fbpx
Wikipedia

Lyme disease

Lyme disease, also known as Lyme borreliosis, is a vector-borne disease caused by Borrelia bacteria, which are spread by ticks in the genus Ixodes.[3][8][9] The most common sign of infection is an expanding red rash, known as erythema migrans (EM), which appears at the site of the tick bite about a week afterwards.[1] The rash is typically neither itchy nor painful.[1] Approximately 70–80% of infected people develop a rash.[1] Early diagnosis can be difficult.[10] Other early symptoms may include fever, headaches and tiredness.[1] If untreated, symptoms may include loss of the ability to move one or both sides of the face, joint pains, severe headaches with neck stiffness or heart palpitations.[1] Months to years later, repeated episodes of joint pain and swelling may occur.[1] Occasionally, shooting pains or tingling in the arms and legs may develop.[1] Despite appropriate treatment, about 10 to 20% of those affected develop joint pains, memory problems, and tiredness for at least six months.[1][11]

Lyme disease
Other namesLyme borreliosis
An adult deer tick (most cases of Lyme are caused by nymphal rather than adult ticks)
SpecialtyInfectious disease
SymptomsExpanding area of redness at the site of a tick bite, fever, headache, tiredness[1]
ComplicationsFacial nerve paralysis, arthritis, meningitis,[1] Heart rhythm irregularities[2]
Usual onsetA week after a bite[1]
CausesBorrelia spread by ticks[3]
Diagnostic methodBased on symptoms, tick exposure, blood tests[4]
PreventionPrevention of tick bites (clothing the limbs, DEET), doxycycline[3]
MedicationDoxycycline, amoxicillin, ceftriaxone, cefuroxime[3]
Frequency~476k/year in U.S. (a likely overestimate), 200k/year in Europe[5][6][7]

Lyme disease is transmitted to humans by the bites of infected ticks of the genus Ixodes.[12] In the United States, ticks of concern are usually of the Ixodes scapularis type. "In most cases, a tick must be attached for 36 to 48 hours or more before the Lyme disease bacterium can be transmitted. If you remove a tick quickly (within 24 hours), you can greatly reduce your chances of getting Lyme disease."[13][14] In Europe, Ixodes ricinus ticks may spread the bacteria more quickly.[14][15] In North America, the bacterial species Borrelia burgdorferi and B. mayonii cause Lyme disease.[3][16] In Europe and Asia, Borrelia afzelii, Borrelia garinii, B. spielmanii and four other species also cause the disease.[3] The disease does not appear to be transmissible between people, by other animals nor through food.[13] Diagnosis is based on a combination of symptoms, history of tick exposure and possibly testing for specific antibodies in the blood.[4][17] Blood tests are often falsely negative in the early stages of the disease.[3] Testing of individual ticks is not typically useful.[18]

Prevention includes efforts to prevent tick bites by wearing clothing to cover the arms and legs and using DEET or picaridin-based insect repellents.[3][8] Using pesticides to reduce tick numbers may also be effective.[3] Ticks can be removed using tweezers.[19] If the removed tick is full of blood a single dose of doxycycline may be used to prevent the development of infection but is not generally recommended since the development of infection is rare.[3] If an infection develops, a number of antibiotics are effective, including doxycycline, amoxicillin and cefuroxime.[3] Standard treatment usually lasts for two or three weeks.[3] Some people develop a fever and muscle and joint pains from treatment, which may last for one or two days.[3] In those who develop persistent symptoms, long-term antibiotic therapy has not been found to be useful.[3][20]

Lyme disease is the most common disease spread by ticks in the Northern Hemisphere.[21][7] Infections are most common in the spring and early summer.[3] Lyme disease was diagnosed as a separate condition for the first time in 1975 in Lyme, Connecticut. It was originally mistaken for juvenile rheumatoid arthritis.[22] The bacterium involved was first described in 1981 by Willy Burgdorfer.[23] Chronic symptoms following treatment are known as "post-treatment Lyme disease syndrome" (PTLDS).[20] PTLDS is different from chronic Lyme disease, a term no longer supported by scientists and used in different ways by different groups.[20][24] Some healthcare providers claim that PTLDS is caused by persistent infection, but this is not believed to be true because no evidence of persistent infection can be found after standard treatment.[25]

As of 2023 clinical trials of proposed human vaccines for Lyme disease were being carried out, but no vaccine was available. A vaccine, LYMERix, was produced, but discontinued in 2002 due to insufficient demand.[26] There are several vaccines for the prevention of Lyme disease in dogs.

Signs and symptoms edit

 
An expanding rash is an initial sign of about 80% of Lyme infections. The rash may look like a 'bull's eye', as pictured, in about 80% of cases in Europe and 20% of cases in the US.[27][28][29][30]

Lyme disease can affect several body systems and produce a broad range of symptoms. Not everyone with Lyme disease has all of the symptoms and many of the symptoms are not specific to Lyme disease, but can occur with other diseases, as well.[31]

The incubation period from infection to the onset of symptoms is usually one to two weeks, but can be much shorter (days) or much longer (months to years).[32] Lyme symptoms most often occur from May to September in the Northern Hemisphere because the nymphal stage of the tick is responsible for most cases.[32] Asymptomatic infection exists, but occurs in less than 7% of infected individuals in the United States.[33] Asymptomatic infection may be much more common among those infected in Europe.[34]

Early localized infection edit

Early, localized infection can occur when the infection has not yet spread throughout the body. Only the site where the infection has first come into contact with the skin is affected. The initial sign of about 80% of Lyme infections is an erythema migrans (EM) rash at the site of a tick bite, often near skin folds such as the armpit, groin, or back of the knee, on the trunk, under clothing straps, or in children's hair, ears, or neck.[27][3] Most people who get infected do not remember seeing a tick or a bite. The rash appears typically one or two weeks (range 3–32 days) after the bite and expands 2–3 cm per day up to a diameter of 5–70 cm (median 16 cm).[27][3][28] The rash is usually circular or oval, red or bluish, and may have an elevated or darker center.[3][29][30] In about 79% of cases in Europe but only 19% of cases in endemic areas of the U.S., the rash gradually clears from the center toward the edges, possibly forming a "bull's eye" pattern.[28][29][30] The rash may feel warm, but usually is not itchy, is rarely tender or painful, and takes up to four weeks to resolve if untreated.[3]

The EM rash is often accompanied by symptoms of a viral-like illness, including fatigue, headache, body aches, fever, and chills, but usually not nausea or upper-respiratory problems. These symptoms may also appear without a rash or linger after the rash has disappeared. Lyme can progress to later stages without these symptoms or a rash.[3]

People with high fever for more than two days or whose other symptoms of viral-like illness do not improve despite antibiotic treatment for Lyme disease, or who have abnormally low levels of white or red cells or platelets in the blood, should be investigated for possible coinfection with other tick-borne diseases such as ehrlichiosis and babesiosis.[35]

Early disseminated infection edit

Within days to weeks after the onset of local infection, the Borrelia bacteria may spread through the lymphatic system or bloodstream. In 10–20% of untreated cases, EM rashes develop at sites across the body that bear no relation to the original tick bite.[27] Transient muscle pains and joint pains are also common.[27]

In about 10–15% of untreated people, Lyme causes neurological problems known as neuroborreliosis.[36] Early neuroborreliosis typically appears 4–6 weeks (range 1–12 weeks) after the tick bite and involves some combination of lymphocytic meningitis, cranial neuritis, radiculopathy, and/or mononeuritis multiplex.[35][37] Lymphocytic meningitis causes characteristic changes in the cerebrospinal fluid (CSF) and may be accompanied for several weeks by variable headache and, less commonly, usually mild meningitis signs such as inability to flex the neck fully and intolerance to bright lights but typically no or only very low fever.[38] After several months neuroborreliosis can also present otolaryngological symptoms. Up to 76.5% of them present as tinnitus, the most common symptom. Vertigo and dizziness (53.7%) and hearing loss (16.7%) were the next most common symptoms.[39] In children partial loss of vision may also occur.[35] Cranial neuritis is an inflammation of cranial nerves. When due to Lyme, it most typically causes facial palsy, impairing blinking, smiling, and chewing on one or both sides of the face. It may also cause intermittent double vision.[35][38] Lyme radiculopathy is an inflammation of spinal nerve roots that often causes pain and less often weakness, numbness, or altered sensation in the areas of the body served by nerves connected to the affected roots, e.g. limb(s) or part(s) of trunk. The pain is often described as unlike any other previously felt, excruciating, migrating, worse at night, rarely symmetrical, and often accompanied by extreme sleep disturbance.[37][40] Mononeuritis multiplex is an inflammation causing similar symptoms in one or more unrelated peripheral nerves.[36][35] Rarely, early neuroborreliosis may involve inflammation of the brain or spinal cord, with symptoms such as confusion, abnormal gait, ocular movements, or speech, impaired movement, impaired motor planning, or shaking.[35][37]

In North America, facial palsy is the typical early neuroborreliosis presentation, occurring in 5–10% of untreated people, in about 75% of cases accompanied by lymphocytic meningitis.[35][41] Lyme radiculopathy is reported half as frequently, but many cases may be unrecognized.[42] In European adults, the most common presentation is a combination of lymphocytic meningitis and radiculopathy known as Bannwarth syndrome, accompanied in 36-89% of cases by facial palsy.[37][40] In this syndrome, radicular pain tends to start in the same body region as the initial erythema migrans rash, if there was one, and precedes possible facial palsy and other impaired movement.[40] In extreme cases, permanent impairment of motor or sensory function of the lower limbs may occur.[34] In European children, the most common manifestations are facial palsy (in 55%), other cranial neuritis, and lymphocytic meningitis (in 27%).[37]

In about 4–10% of untreated cases in the U.S. and 0.3–4% of untreated cases in Europe, typically between June and December, about one month (range 4 days – 7 months) after the tick bite, the infection may cause heart complications known as Lyme carditis.[43][44] Symptoms may include heart palpitations (in 69% of people), dizziness, fainting, shortness of breath, and chest pain.[43] Other symptoms of Lyme disease may also be present, such as EM rash, joint aches, facial palsy, headaches, or radicular pain.[43] In some people, however, carditis may be the first manifestation of Lyme disease.[43] Lyme carditis in 19–87% of people adversely impacts the heart's electrical conduction system, causing atrioventricular block that often manifests as heart rhythms that alternate within minutes between abnormally slow and abnormally fast.[43][44] In 10–15% of people, Lyme causes myocardial complications such as cardiomegaly, left ventricular dysfunction, or congestive heart failure.[43]

Another skin condition, found in Europe but not in North America, is borrelial lymphocytoma, a purplish lump that develops on the ear lobe, nipple, or scrotum.[45]

Late disseminated infection edit

 
Lyme arthritis caused this 3-year-old girl's knee to become swollen. Though painless, it did make her limp.[46]

Lyme arthritis occurs in up to 60% of untreated people, typically starting about six months after infection.[27] It usually affects only one or a few joints, often a knee or possibly the hip, other large joints, or the temporomandibular joint.[35][47] Usually, large joint effusion and swelling occur, but only mild or moderate pain.[35] Without treatment, swelling and pain typically resolve over time, but periodically return.[35] Baker's cysts may form and rupture.

In early US studies of Lyme disease, a rare peripheral neuropathy was described that included numbness, tingling, or burning starting at the feet or hands and over time possibly moving up the limbs. In a later analysis that discovered poor documentation of this manifestation, experts wondered if it exists at all in the US or is merely very rare.[35][48]

A neurologic syndrome called Lyme encephalopathy is associated with subtle memory and cognitive difficulties, insomnia, a general sense of feeling unwell, and changes in personality.[49] Lyme encephalopathy is controversial in the US and has not been reported in Europe.[7] Problems such as depression and fibromyalgia are as common in people with Lyme disease as in the general population.[50][51] There is no compelling evidence that Lyme disease causes psychiatric disorders, behavioral disorders (e.g. ADHD), or developmental disorders (e.g. autism).[52]

Acrodermatitis chronica atrophicans is a chronic skin disorder observed primarily in Europe among the elderly.[45] It begins as a reddish-blue patch of discolored skin, often on the backs of the hands or feet. The lesion slowly atrophies over several weeks or months, with the skin becoming first thin and wrinkled and then, if untreated, completely dry and hairless.[53] It is also associated with peripheral neuropathy.[48]

Cause edit

 
Borrelia bacteria, the causative agents of Lyme disease, magnified

Lyme disease is caused by spirochetes, spiral bacteria from the genus Borrelia. Spirochetes are surrounded by peptidoglycan and flagella, along with an outer membrane similar to Gram-negative bacteria. Because of their double-membrane envelope, Borrelia bacteria are often mistakenly described as Gram negative despite the considerable differences in their envelope components from Gram-negative bacteria.[54] The Lyme-related Borrelia species are collectively known as Borrelia burgdorferi sensu lato, and show a great deal of genetic diversity.[55]

B. burgdorferi sensu lato is a species complex made up of 20 accepted and three proposed genospecies. Eight species are known to cause Lyme disease: B. mayonii (found in North America), B. burgdorferi sensu stricto (found in North America and Europe), B. afzelii, B. garinii, B. spielmanii, and B. lusitaniae (all found in Eurasia).[56][57][16][9] Some studies have also proposed that B. valaisiana may sometimes infect humans, but this species does not seem to be an important cause of disease.[58][59]

Tick life cycle edit

 
Deer tick life cycle
 
Ixodes scapularis, the primary vector of Lyme disease in eastern North America

Three stages occur in the life cycle of a tick - larva, nymph, and adult. During the nymph stage, ticks most frequently transmit Lyme disease and are usually most active in late spring and early summer in regions where the climate is mild. During the adult stage, Lyme disease transmission is less common because adult ticks are less likely to bite humans and tend to be larger in size, so can be easily seen and removed.[60]

Transmission edit

Lyme disease is classified as a zoonosis, as it is transmitted to humans from a natural reservoir among small mammals and birds by ticks that feed on both sets of hosts.[61] Hard-bodied ticks of the genus Ixodes are the vectors of Lyme disease (also the vector for Babesia).[62] Most infections are caused by ticks in the nymphal stage, because they are very small, thus may feed for long periods of time undetected.[61] Nymphal ticks are generally the size of a poppy seed and sometimes with a dark head and a translucent body.[63] Or, the nymphal ticks can be darker.[64] The younger larval ticks are very rarely infected.[65] Although deer are the preferred hosts of adult deer ticks, and tick populations are much lower in the absence of deer, ticks generally do not acquire Borrelia from deer, instead they obtain them from infected small mammals such as the white-footed mouse, and occasionally birds.[66] Areas where Lyme is common are expanding.[67]

Within the tick midgut, the Borrelia's outer surface protein A (OspA) binds to the tick receptor for OspA, known as TROSPA. When the tick feeds, the Borrelia downregulates OspA and upregulates OspC, another surface protein. After the bacteria migrate from the midgut to the salivary glands, OspC binds to Salp15, a tick salivary protein that appears to have immunosuppressive effects that enhance infection.[68] Successful infection of the mammalian host depends on bacterial expression of OspC.[69]

Tick bites often go unnoticed because of the small size of the tick in its nymphal stage, as well as tick secretions that prevent the host from feeling any itch or pain from the bite. However, transmission is quite rare, with only about 1.2 to 1.4 percent of recognized tick bites resulting in Lyme disease.[70]

 
Tick Ixodes ricinus, developmental stages

In Europe, the main vector is Ixodes ricinus, which is also called the sheep tick or castor bean tick.[71] In China, Ixodes persulcatus (the taiga tick) is probably the most important vector.[72] In North America, the black-legged tick or deer tick (Ixodes scapularis) is the main vector on the East Coast.[65]

The lone star tick (Amblyomma americanum), which is found throughout the Southeastern United States as far west as Texas, is unlikely to transmit the Lyme disease spirochetes,[73] though it may be implicated in a related syndrome called southern tick-associated rash illness, which resembles a mild form of Lyme disease.[74]

On the West Coast of the United States, the main vector is the western black-legged tick (Ixodes pacificus).[75] The tendency of this tick species to feed predominantly on host species such as the Western Fence Lizard that are resistant to Borrelia infection appears to diminish transmission of Lyme disease in the West.[76][77]

Transmission can occur across the placenta during pregnancy and as with a number of other spirochetal diseases, adverse pregnancy outcomes are possible with untreated infection; prompt treatment with antibiotics reduces or eliminates this risk.[78][79][80][81][82]

There is no scientific evidence to support Lyme disease transmission via blood transfusion, sexual contact, or breast milk.[83]

Tick-borne co-infections edit

Ticks that transmit B. burgdorferi to humans can also carry and transmit several other microbes, such as Babesia microti and Anaplasma phagocytophilum, which cause the diseases babesiosis and human granulocytic anaplasmosis (HGA), respectively.[84] Among people with early Lyme disease, depending on their location, 2–12% will also have HGA and 2–10% will have babesiosis.[52] Ticks in certain regions also transmit viruses that cause tick-borne encephalitis and Powassan virus disease.[85][52] Co-infections of Lyme disease may not require additional treatment, since they may resolve on their own or—as in the case of HGA—can be treated with the doxycycline prescribed for Lyme.[35] Persistent fever or compatible anomalous laboratory findings may be indicative of a co-infection.[52]

Pathophysiology edit

B. burgdorferi can spread throughout the body during the course of the disease, and has been found in the skin, heart, joints, peripheral nervous system, and central nervous system.[69][86] B. Burgdorferi does not produce toxins.[87] Therefore, many of the signs and symptoms of Lyme disease are a consequence of the immune response to spirochete in those tissues.[88]

B. burgdorferi is injected into the skin by the bite of an infected Ixodes tick. Tick saliva, which accompanies the spirochete into the skin during the feeding process, contains substances that disrupt the immune response at the site of the bite.[89] This provides a protective environment where the spirochete can establish infection. The spirochetes multiply and migrate outward within the dermis. The host inflammatory response to the bacteria in the skin causes the characteristic circular EM lesion.[69] Neutrophils, however, which are necessary to eliminate the spirochetes from the skin, fail to appear in necessary numbers in the developing EM lesion because tick saliva inhibits neutrophil function. This allows the bacteria to survive and eventually spread throughout the body.[90]

Days to weeks following the tick bite, the spirochetes spread via the bloodstream to joints, heart, nervous system, and distant skin sites, where their presence gives rise to the variety of symptoms of the disseminated disease. The spread of B. burgdorferi is aided by the attachment of the host protease plasmin to the surface of the spirochete.[91]

If untreated, the bacteria may persist in the body for months or even years, despite the production of B. burgdorferi antibodies by the immune system.[92] The spirochetes may avoid the immune response by decreasing expression of surface proteins that are targeted by antibodies, antigenic variation of the VlsE surface protein, inactivating key immune components such as complement, and hiding in the extracellular matrix, which may interfere with the function of immune factors.[93][94]

In the brain, B. burgdorferi may induce astrocytes to undergo astrogliosis (proliferation followed by apoptosis), which may contribute to neurodysfunction.[95] The spirochetes may also induce host cells to secrete quinolinic acid, which stimulates the NMDA receptor on nerve cells, which may account for the fatigue and malaise observed with Lyme encephalopathy.[96] In addition, diffuse white matter pathology during Lyme encephalopathy may disrupt gray matter connections, and could account for deficits in attention, memory, visuospatial ability, complex cognition, and emotional status. White matter disease may have a greater potential for recovery than gray matter disease, perhaps because neuronal loss is less common. Resolution of MRI white matter hyperintensities after antibiotic treatment has been observed.[97]

Tryptophan, a precursor to serotonin, appears to be reduced within the central nervous system in a number of infectious diseases that affect the brain, including Lyme.[98] Researchers are investigating if this neurohormone secretion is the cause of neuropsychiatric disorders developing in some people with borreliosis.[99]

Immunological studies edit

Exposure to the Borrelia bacterium during Lyme disease possibly causes a long-lived and damaging inflammatory response,[100] a form of pathogen-induced autoimmune disease.[101] The production of this reaction might be due to a form of molecular mimicry, where Borrelia avoids being killed by the immune system by resembling normal parts of the body's tissues.[102][103]

Chronic symptoms from an autoimmune reaction could explain why some symptoms persist even after the spirochetes have been eliminated from the body. This hypothesis may explain why chronic arthritis persists after antibiotic therapy, similar to rheumatic fever, but its wider application is controversial.[104][105]

Diagnosis edit

Lyme disease is diagnosed based on symptoms, objective physical findings (such as erythema migrans (EM) rash, facial palsy, or arthritis), history of possible exposure to infected ticks, and possibly laboratory tests.[3][27] People with symptoms of early Lyme disease should have a total body skin examination for EM rashes and asked whether EM-type rashes had manifested within the last 1–2 months.[35] Presence of an EM rash and recent tick exposure (i.e., being outdoors in a likely tick habitat where Lyme is common, within 30 days of the appearance of the rash) are sufficient for Lyme diagnosis; no laboratory confirmation is needed or recommended.[3][27][106][107] Most people who get infected do not remember a tick or a bite, and the EM rash need not look like a bull's eye (most EM rashes in the U.S. do not) or be accompanied by any other symptoms.[3][108] In the U.S., Lyme is most common in the New England and Mid-Atlantic states and parts of Wisconsin and Minnesota, but it is expanding into other areas.[67] Several bordering areas of Canada also have high Lyme risk.[109]

In the absence of an EM rash or history of tick exposure, Lyme diagnosis depends on laboratory confirmation.[62][110] The bacteria that cause Lyme disease are difficult to observe directly in body tissues and also difficult and too time-consuming to grow in the laboratory.[3][62] The most widely used tests look instead for presence of antibodies against those bacteria in the blood.[111] A positive antibody test result does not by itself prove active infection but can confirm an infection that is suspected because of symptoms, objective findings, and history of tick exposure in a person.[62] Because as many as 5–20% of the normal population have antibodies against Lyme, people without history and symptoms suggestive of Lyme disease should not be tested for Lyme antibodies: a positive result would likely be false, possibly causing unnecessary treatment.[35][37]

In some cases, when history, signs, and symptoms are strongly suggestive of early disseminated Lyme disease, empiric treatment may be started and reevaluated as laboratory test results become available.[41][112]

Laboratory testing edit

Tests for antibodies in the blood by ELISA and Western blot is the most widely used method for Lyme diagnosis. A two-tiered protocol is recommended by the Centers for Disease Control and Prevention (CDC): the sensitive ELISA test is performed first, and if it is positive or equivocal, then the more specific Western blot is run.[113] The immune system takes some time to produce antibodies in quantity. After Lyme infection onset, antibodies of types IgM and IgG usually can first be detected respectively at 2–4 weeks and 4–6 weeks, and peak at 6–8 weeks.[114] When an EM rash first appears, detectable antibodies may not be present. Therefore, it is recommended that testing not be performed and diagnosis be based on the presence of the EM rash.[35] Up to 30 days after suspected Lyme infection onset, infection can be confirmed by detection of IgM or IgG antibodies; after that, it is recommended that only IgG antibodies be considered.[114] A positive IgM and negative IgG test result after the first month of infection is generally indicative of a false-positive result.[115] The number of IgM antibodies usually collapses 4–6 months after infection, while IgG antibodies can remain detectable for years.[114]

Other tests may be used in neuroborreliosis cases. In Europe, neuroborreliosis is usually caused by Borrelia garinii and almost always involves lymphocytic pleocytosis, i.e. the densities of lymphocytes (infection-fighting cells) and protein in the cerebrospinal fluid (CSF) typically rise to characteristically abnormal levels, while glucose level remains normal.[38][35][40] Additionally, the immune system produces antibodies against Lyme inside the intrathecal space, which contains the CSF.[35][40] Demonstration by lumbar puncture and CSF analysis of pleocytosis and intrathecal antibody production are required for definite diagnosis of neuroborreliosis in Europe (except in cases of peripheral neuropathy associated with acrodermatitis chronica atrophicans, which usually is caused by Borrelia afzelii and confirmed by blood antibody tests).[37] In North America, neuroborreliosis is caused by Borrelia burgdorferi and may not be accompanied by the same CSF signs; they confirm a diagnosis of central nervous system (CNS) neuroborreliosis if positive, but do not exclude it if negative.[116] American guidelines consider CSF analysis optional when symptoms appear to be confined to the peripheral nervous system (PNS), e.g. facial palsy without overt meningitis symptoms.[35][117] Unlike blood and intrathecal antibody tests, CSF pleocytosis tests revert to normal after infection ends and therefore can be used as objective markers of treatment success and inform decisions on whether to retreat.[40] In infection involving the PNS, electromyography and nerve conduction studies can be used to monitor objectively the response to treatment.[38]

In Lyme carditis, electrocardiograms are used to evidence heart conduction abnormalities, while echocardiography may show myocardial dysfunction.[43] Biopsy and confirmation of Borrelia cells in myocardial tissue may be used in specific cases but are usually not done because of risk of the procedure.[43]

Polymerase chain reaction (PCR) tests for Lyme disease have also been developed to detect the genetic material (DNA) of the Lyme disease spirochete. Culture or PCR are the current means for detecting the presence of the organism, as serologic studies only test for antibodies of Borrelia. PCR has the advantage of being much faster than culture. However, PCR tests are susceptible to false positive results, e.g. by detection of debris of dead Borrelia cells or specimen contamination.[118][37] Even when properly performed, PCR often shows false-negative results because few Borrelia cells can be found in blood and cerebrospinal fluid (CSF) during infection.[119][37] Hence, PCR tests are recommended only in special cases, e.g. diagnosis of Lyme arthritis, because it is a highly sensitive way of detecting ospA DNA in synovial fluid.[120] Although sensitivity of PCR in CSF is low, its use may be considered when intrathecal antibody production test results are suspected of being falsely negative, e.g. in very early (< 6 weeks) neuroborreliosis or in immunosuppressed people.[37]

Several other forms of laboratory testing for Lyme disease are available, some of which have not been adequately validated. OspA antigens, shed by live Borrelia bacteria into urine, are a promising technique being studied.[121] The use of nanotrap particles for their detection is being looked at and the OspA has been linked to active symptoms of Lyme.[122][123] High titers of either immunoglobulin G (IgG) or immunoglobulin M (IgM) antibodies to Borrelia antigens indicate disease, but lower titers can be misleading, because the IgM antibodies may remain after the initial infection, and IgG antibodies may remain for years.[124]

The CDC does not recommend urine antigen tests, PCR tests on urine, immunofluorescent staining for cell-wall-deficient forms of B. burgdorferi, and lymphocyte transformation tests.[119]

Imaging edit

Neuroimaging is controversial in whether it provides specific patterns unique to neuroborreliosis, but may aid in differential diagnosis and in understanding the pathophysiology of the disease.[125] Though controversial, some evidence shows certain neuroimaging tests can provide data that are helpful in the diagnosis of a person. Magnetic resonance imaging (MRI) and single-photon emission computed tomography (SPECT) are two of the tests that can identify abnormalities in the brain of a person affected with this disease. Neuroimaging findings in an MRI include lesions in the periventricular white matter, as well as enlarged ventricles and cortical atrophy. The findings are considered somewhat unexceptional because the lesions have been found to be reversible following antibiotic treatment. Images produced using SPECT show numerous areas where an insufficient amount of blood is being delivered to the cortex and subcortical white matter. However, SPECT images are known to be nonspecific because they show a heterogeneous pattern in the imaging. The abnormalities seen in the SPECT images are very similar to those seen in people with cerebral vasculitis and Creutzfeldt–Jakob disease, which makes them questionable.[126]

Differential diagnosis edit

Community clinics have been reported to misdiagnose 23–28% of Erythema migrans (EM) rashes and 83% of other objective manifestations of early Lyme disease.[110] EM rashes are often misdiagnosed as spider bites, cellulitis, or shingles.[110] Many misdiagnoses are credited to the widespread misconception that EM rashes should look like a bull's eye.[3] Actually, the key distinguishing features of the EM rash are the speed and extent to which it expands, respectively up to 2–3 cm/day and a diameter of at least 5 cm, and in 50% of cases more than 16 cm. The rash expands away from its center, which may or may not look different or be separated by ring-like clearing from the rest of the rash.[27][28] Compared to EM rashes, spider bites are more common in the limbs, tend to be more painful and itchy or become swollen, and some may cause necrosis (sinking dark blue patch of dead skin).[27][3] Cellulitis most commonly develops around a wound or ulcer, is rarely circular, and is more likely to become swollen and tender.[27][3] EM rashes often appear at sites that are unusual for cellulitis, such as the armpit, groin, abdomen, or back of knee.[27] Like Lyme, shingles often begins with headache, fever, and fatigue, which are followed by pain or numbness. However, unlike Lyme, in shingles these symptoms are usually followed by appearance of rashes composed of multiple small blisters along with a nerve's dermatome, and shingles can also be confirmed by quick laboratory tests.[127]

Facial palsy caused by Lyme disease (LDFP) is often misdiagnosed as Bell's palsy.[41] Although Bell's palsy is the most common type of one-sided facial palsy (about 70% of cases), LDFP can account for about 25% of cases of facial palsy in areas where Lyme disease is common.[41] Compared to LDFP, Bell's palsy much less frequently affects both sides of the face.[41] Even though LDFP and Bell's palsy have similar symptoms and evolve similarly if untreated, corticosteroid treatment is beneficial for Bell's Palsy, while being detrimental for LDFP.[41] Recent history of exposure to a likely tick habitat during warmer months, EM rash, viral-like symptoms such as headache and fever, and/or palsy in both sides of the face should be evaluated for the likelihood of LDFP; if it is more than minimal, empiric therapy with antibiotics should be initiated, without corticosteroids, and reevaluated upon completion of laboratory tests for Lyme disease.[41]

Unlike viral meningitis, Lyme lymphocytic meningitis tends to not cause fever, last longer, and recur.[38][35] Lymphocytic meningitis is also characterized by possibly co-occurring with EM rash, facial palsy, or partial vision obstruction and having much lower percentage of polymorphonuclear leukocytes in CSF.[35]

Lyme radiculopathy affecting the limbs is often misdiagnosed as a radiculopathy caused by nerve root compression, such as sciatica.[110][128] Although most cases of radiculopathy are compressive and resolve with conservative treatment (e.g., rest) within 4–6 weeks, guidelines for managing radiculopathy recommend first evaluating risks of other possible causes that, although less frequent, require immediate diagnosis and treatment, including infections such as Lyme and shingles.[129] A history of outdoor activities in likely tick habitats in the last 3 months possibly followed by a rash or viral-like symptoms, and current headache, other symptoms of lymphocytic meningitis, or facial palsy would lead to suspicion of Lyme disease and recommendation of serological and lumbar puncture tests for confirmation.[129]

Lyme radiculopathy affecting the trunk can be misdiagnosed as myriad other conditions, such as diverticulitis and acute coronary syndrome.[42][110] Diagnosis of late-stage Lyme disease is often complicated by a multifaceted appearance and nonspecific symptoms, prompting one reviewer to call Lyme the new "great imitator".[130] As all people with later-stage infection will have a positive antibody test, simple blood tests can exclude Lyme disease as a possible cause of a person's symptoms.[131]

Prevention edit

Tick bites may be prevented by avoiding or reducing time in likely tick habitats and taking precautions while in and when getting out of one.[132][8]

Most Lyme human infections are caused by Ixodes nymph bites between April and September.[27][132] Ticks prefer moist, shaded locations in woodlands, shrubs, tall grasses and leaf litter or wood piles.[27][133] Tick densities tend to be highest in woodlands, followed by unmaintained edges between woods and lawns (about half as high), ornamental plants and perennial groundcover (about a quarter), and lawns (about 30 times less).[134] Ixodes larvae and nymphs tend to be abundant also where mice nest, such as stone walls and wood logs.[134] Ixodes larvae and nymphs typically wait for potential hosts ("quest") on leaves or grasses close to the ground with forelegs outstretched; when a host brushes against its limbs, the tick rapidly clings and climbs on the host looking for a skin location to bite.[135] In Northeastern United States, 69% of tick bites are estimated to happen in residences, 11% in schools or camps, 9% in parks or recreational areas, 4% at work, 3% while hunting, and 4% in other areas.[134] Activities associated with tick bites around residences include yard work, brush clearing, gardening, playing in the yard, and letting into the house dogs or cats that roam outside in woody or grassy areas.[134][132] In parks, tick bites often happen while hiking or camping.[134] Walking on a mowed lawn or center of a trail without touching adjacent vegetation is less risky than crawling or sitting on a log or stone wall.[134][136] Pets should not be allowed to roam freely in likely tick habitats.[133]

As a precaution, CDC recommends soaking or spraying clothes, shoes, and camping gear such as tents, backpacks and sleeping bags with 0.5% permethrin solution and hanging them to dry before use.[132][137] Permethrin is odorless and safe for humans but highly toxic to ticks.[138] After crawling on permethrin-treated fabric for as few as 10–20 seconds, tick nymphs become irritated and fall off or die.[138][139] Permethrin-treated closed-toed shoes and socks reduce by 74 times the number of bites from nymphs that make first contact with a shoe of a person also wearing treated shorts (because nymphs usually quest near the ground, this is a typical contact scenario).[138] Better protection can be achieved by tucking permethrin-treated trousers (pants) into treated socks and a treated long-sleeve shirt into the trousers so as to minimize gaps through which a tick might reach the wearer's skin.[136] Light-colored clothing may make it easier to see ticks and remove them before they bite.[136] Military and outdoor workers' uniforms treated with permethrin have been found to reduce the number of bite cases by 80–95%.[139] Permethrin protection lasts several weeks of wear and washings in customer-treated items and up to 70 washings for factory-treated items.[137] Permethrin should not be used on human skin, underwear or cats.[137][140]

The EPA recommends several tick repellents for use on exposed skin, including DEET, picaridin, IR3535 (a derivative of amino acid beta-alanine), oil of lemon eucalyptus (OLE, a natural compound) and OLE's active ingredient para-menthane-diol (PMD).[132][141][142] Unlike permethrin, repellents repel but do not kill ticks, protect for only several hours after application, and may be washed off by sweat or water.[137] The most popular repellent is DEET in the U.S. and picaridin in Europe.[142] Unlike DEET, picaridin is odorless and is less likely to irritate the skin or harm fabric or plastics.[142] Repellents with higher concentration may last longer but are not more effective; against ticks, 20% picaridin may work for 8 hours vs. 55–98.11% DEET for 5–6 hours or 30–40% OLE for 6 hours.[137][141] Repellents should not be used under clothes, on eyes, mouth, wounds or cuts, or on babies younger than 2 months (3 years for OLE or PMD).[137][132] If sunscreen is used, repellent should be applied on top of it.[137] Repellents should not be sprayed directly on a face, but should instead be sprayed on a hand and then rubbed on the face.[137]

After coming indoors, clothes, gear and pets should be checked for ticks.[132] Clothes can be put into a hot dryer for 10 minutes to kill ticks (just washing or warm dryer are not enough).[132] Showering as soon as possible, looking for ticks over the entire body, and removing them reduce risk of infection.[132] Unfed tick nymphs are the size of a poppy seed, but a day or two after biting and attaching themselves to a person, they look like a small blood blister.[143] The following areas should be checked especially carefully: armpits, between legs, back of knee, bellybutton, trunk, and in children ears, neck and hair.[132]

Tick removal edit

 
Removal of a tick using tweezers

Attached ticks should be removed promptly. Risk of infection increases with time of attachment, but in North America risk of Lyme disease is small if the tick is removed within 36 hours.[144] CDC recommends inserting a fine-tipped tweezer between the skin and the tick, grasping very firmly, and pulling the closed tweezer straight away from the skin without twisting, jerking, squeezing or crushing the tick.[145] After tick removal, any tick parts remaining in the skin should be removed with a clean tweezer, if possible.[145] The wound and hands should then be cleaned with alcohol or soap and water.[145] The tick may be disposed by placing it in a container with alcohol, sealed bag, tape or flushed down the toilet.[145] The bitten person should write down where and when the bite happened so that this can be informed to a doctor if the person gets a rash or flu-like symptoms in the following several weeks.[145] CDC recommends not using fingers, nail polish, petroleum jelly or heat on the tick to try to remove it.[145]

In Australia, where the Australian paralysis tick is prevalent, the Australasian Society of Clinical Immunology and Allergy recommends not using tweezers to remove ticks, because if the person is allergic, anaphylaxis could result.[146] Instead, a product should be sprayed on the tick to cause it to freeze and then drop off.[146] Another method consists in using about 20 cm of dental floss or fishing line for slowly tying an overhand knot between the skin and the tick and then pulling it away from the skin.[147][148]

Preventive antibiotics edit

The risk of infectious transmission increases with the duration of tick attachment.[27] It requires between 36 and 48 hours of attachment for the bacteria that causes Lyme to travel from within the tick into its saliva.[27] If a deer tick that is sufficiently likely to be carrying Borrelia is found attached to a person and removed, and if the tick has been attached for 36 hours or is engorged, a single dose of doxycycline administered within the 72 hours after removal may reduce the risk of Lyme disease. It is not generally recommended for all people bitten, as development of infection is rare: about 50 bitten people would have to be treated this way to prevent one case of erythema migrans (i.e. the typical rash found in about 70–80% of people infected).[3][27]

Garden landscaping edit

Several landscaping practices may reduce the risk of tick bites in residential yards.[143][149] These include keeping lawns mowed, removing leaf litter and weeds, and avoiding the use of ground cover.[143] A 3-ft-wide rock or woodchip barrier is recommended to separate lawns from wood piles, woodlands, stone walls, and shrubs.[149] Without vegetation on the barrier, ticks will tend not to cross it; acaricides may also be sprayed on it to kill ticks.[149] A sun-exposed tick-safe zone at least 9 ft from the barrier should concentrate human activity on the yard, including any patios, playgrounds and gardening.[149] Materials such as wood decking, concrete, bricks, gravel or woodchips used on the ground under patios and playgrounds would discourage ticks there.[143] An 8-ft-high fence may be added to keep deer away from the tick-safe zone.[149][143]

Occupational exposure edit

Outdoor workers are at risk of Lyme disease if they work at sites with infected ticks. This includes construction, landscaping, forestry, brush clearing, land surveying, farming, railroad work, oil field work, utility line work, park or wildlife management.[150][151] U.S. workers in the northeastern and north-central states are at highest risk of exposure to infected ticks. Ticks may also transmit other tick-borne diseases to workers in these and other regions of the country. Worksites with woods, bushes, high grass or leaf litter are likely to have more ticks. Outdoor workers should be most careful to protect themselves in the late spring and summer when young ticks are most active.[152]

Host animals edit

Ticks can feed upon the blood of a wide array of possible host species, including lizards, birds, mice, cats, dogs, deer, cattle and humans. The extent to which a tick can feed, reproduce, and spread will depend on the type and availability of its hosts. Whether it will spread disease is also affected by its available hosts. Some species, such as lizards, are referred to as "dilution hosts" because they don't tend to support Lyme disease pathogens and so decrease the likelihood that the disease will be passed on by ticks feeding on them. White-tailed deer are both a food source and a "reproductive host", where ticks tend to mate. The white-footed mouse is a reservoir host in which the pathogen for Lyme disease can survive. Availability of hosts can have significant impacts on the transmission of Lyme disease. A greater diversity of hosts, or of those that don't support the pathogen, tends to decrease the likelihood that the disease will be transmitted.[153]

In the United States, one approach to reducing the incidence of Lyme and other deer tick-borne diseases has been to greatly reduce the deer population on which the adult ticks depend for feeding and reproduction. Lyme disease cases fell following deer eradication on an island, Monhegan, Maine,[154] and following deer control in Mumford Cove, Connecticut.[155] Advocates have suggested reducing the deer population to levels of 8 to 10 deer per square mile, compared to levels of 60 or more deer per square mile in the areas of the country with the highest Lyme disease rates.[156]

Others have noted that while deer are reproductive hosts, they are not Borrelia burgdorferi reservoirs. Researchers have suggested that smaller, less obviously visible Lyme reservoirs, like white-footed mice and Eastern chipmunks, may more strongly impact Lyme disease occurrence. Ecosystem studies in New York state suggest that white-footed mice thrive when forests are broken into smaller isolated chunks of woodland with fewer rodent predators. With more rodents harboring the disease, the odds increase that a tick will feed on a disease-harboring rodent and that someone will pick up a disease-carrying tick in their garden or walking in the woods. Data indicates that the smaller the wooded area, the more ticks it will contain and the likely they are to carry Lyme disease, supporting the idea that deforestation and habitat fragmentation affect ticks, hosts and disease transmission.[153]

Tick-borne diseases are estimated to affect ~80 % of cattle worldwide.[157] They also affect cats, dogs, and other pets. Routine veterinary control of ticks of domestic animals through the use of acaricides has been suggested as a way to reduce exposure of humans to ticks. However, chemical control with acaricides is now criticized on a number of grounds. Ticks appear to develop resistance to acaricides; acaricides are costly; and there are concerns over their toxicity and the potential for chemical residues to affect food and the environment.[158]

In Europe, known reservoirs of Borrelia burgdorferi were 9 small mammals, 7 medium-sized mammals and 16 species of birds (including passerines, sea-birds and pheasants).[159] These animals seem to transmit spirochetes to ticks and thus participate in the natural circulation of B. burgdorferi in Europe. The house mouse is also suspected as well as other species of small rodents, particularly in Eastern Europe and Russia.[159] "The reservoir species that contain the most pathogens are the European roe deer Capreolus capreolus;[160] "it does not appear to serve as a major reservoir of B. burgdorferi" thought Jaenson & al. (1992)[161] (incompetent host for B. burgdorferi and TBE virus) but it is important for feeding the ticks,[162] as red deer and wild boars (Sus scrofa),[160] in which one Rickettsia and three Borrelia species were identified",[160] with high risks of coinfection in roe deer.[163] Nevertheless, in the 2000s, in roe deer in Europe "two species of Rickettsia and two species of Borrelia were identified".[160]

Vaccination edit

As of 2023 no human vaccines for Lyme disease were available.[26] The only human vaccine to advance to market was LYMErix, which was available from 1998, but discontinued in 2002.[164] The vaccine candidate VLA15 was scheduled to start a phase 3 trial in the third quarter of 2022, with other research ongoing. Multiple vaccines are available for the prevention of Lyme disease in dogs.

LYMErix edit

The vaccine LYMErix was available from 1998 to 2002. The recombinant vaccine against Lyme disease, based on the outer surface protein A (OspA) of B. burgdorferi with aluminum hydroxide as adjuvant, was developed by SmithKline Beecham. In clinical trials involving more than 10,000 people, the vaccine was found to confer protective immunity to Lyme disease in 76% of adults after three doses with only mild or moderate and transient adverse effects.[165][166] On 21 December 1998, the Food and Drug Administration (FDA) approved LYMErix on the basis of these trials for persons of ages 15 through 70.[166][164]

Following approval of the vaccine, its entry into clinical practice was slow for a variety of reasons, including its cost, which was often not reimbursed by insurance companies.[167] Subsequently, hundreds of vaccine recipients reported they had developed autoimmune and other side effects. Supported by some advocacy groups, a number of class-action lawsuits were filed against GlaxoSmithKline, alleging the vaccine had caused these health problems. These claims were investigated by the FDA and the Centers for Disease Control, which found no connection between the vaccine and the autoimmune complaints.[168]

Despite the lack of evidence that the complaints were caused by the vaccine, sales plummeted and LYMErix was withdrawn from the U.S. market by GlaxoSmithKline in February 2002,[169] in the setting of negative media coverage and fears of vaccine side effects.[168][170] The fate of LYMErix was described in the medical literature as a "cautionary tale";[170] an editorial in Nature cited the withdrawal of LYMErix as an instance in which "unfounded public fears place pressures on vaccine developers that go beyond reasonable safety considerations."[171] The original developer of the OspA vaccine at the Max Planck Institute told Nature: "This just shows how irrational the world can be ... There was no scientific justification for the first OspA vaccine LYMErix being pulled."[168][172]

VLA15 edit

The hexavalent (OspA) protein subunit-based vaccine candidate VLA15 was developed by Valneva. It was granted fast track designation by the U.S. Food and Drug Administration in July 2017.[173][174] In April 2020 Pfizer paid $130 million for the rights to the vaccine, and the companies are developing it together, performing multiple phase 2 trials.[175]

A phase 3 trial of VLA15 was scheduled for late 2022, recruiting volunteers at test sites located across the northeastern United States and in Europe.[176][177] Participants were scheduled to receive an initial three-dose series of vaccines over the course of five to nine months, followed by a booster dose after twelve months, with both the initial series and the booster dose scheduled to be complete before the year's peak Lyme disease season.[177]

Other research edit

An mRNA vaccine designed to cause a strong fast immune response to tick saliva allowed the immune system to detect and remove the ticks from test animals before they were able to transmit the infectious bacteria.[178] The vaccine contains mRNAs for the body to build 19 proteins in tick saliva which, by enabling quick development of erythema (itchy redness) at the bite site, protects guinea pigs against Lyme disease. It also protected the test animals if the tick is not removed if only one tick, but not three, remain attached.[179][180]

Canine vaccines edit

Canine vaccines have been formulated and approved for the prevention of Lyme disease in dogs. Currently, three Lyme disease vaccines are available. LymeVax, formulated by Fort Dodge Laboratories, contains intact dead spirochetes which expose the host to the organism. Galaxy Lyme, Intervet-Schering-Plough's vaccine, targets proteins OspC and OspA. The OspC antibodies kill any of the bacteria that have not been killed by the OspA antibodies. Canine Recombinant Lyme, formulated by Merial, generates antibodies against the OspA protein so a tick feeding on a vaccinated dog draws in blood full of anti-OspA antibodies, which kill the spirochetes in the tick's gut before they are transmitted to the dog.[181]

Treatment edit

Antibiotics are the primary treatment.[3][27] The specific approach to their use is dependent on the individual affected and the stage of the disease.[27] For most people with early localized infection, oral administration of doxycycline is widely recommended as the first choice, as it is effective against not only Borrelia bacteria but also a variety of other illnesses carried by ticks.[27] People taking doxycycline should avoid sun exposure because of higher risk of sunburns.[35] Doxycycline is contraindicated in children younger than eight years of age and women who are pregnant or breastfeeding;[27] alternatives to doxycycline are amoxicillin, cefuroxime axetil, and azithromycin.[27] Azithromycin is recommended only in case of intolerance to the other antibiotics.[35] The standard treatment for cellulitis, cephalexin, is not useful for Lyme disease.[35] When it is unclear if a rash is caused by Lyme or cellulitis, the IDSA recommends treatment with cefuroxime or amoxicillin/clavulanic acid, as these are effective against both infections.[35] Individuals with early disseminated or late Lyme infection may have symptomatic cardiac disease, Lyme arthritis, or neurologic symptoms like facial palsy, radiculopathy, meningitis, or peripheral neuropathy.[27] Intravenous administration of ceftriaxone is recommended as the first choice in these cases;[27] cefotaxime and doxycycline are available as alternatives.[27]

Treatment regimens for Lyme disease range from 14 days in early localized disease, to 14–21 days in early disseminated disease to 14–28 days in late disseminated disease.[182] Neurologic complications of Lyme disease may be treated with doxycycline as it can be taken by mouth and has a lower cost, although in North America evidence of efficacy is only indirect.[117] In case of failure, guidelines recommend retreatment with injectable ceftriaxone.[117] Several months after treatment for Lyme arthritis, if joint swelling persists or returns, a second round of antibiotics may be considered; intravenous antibiotics are preferred for retreatment in case of poor response to oral antibiotics.[27][35] Outside of that, a prolonged antibiotic regimen lasting more than 28 days is not recommended as no evidence shows it to be effective.[27][183] IgM and IgG antibody levels may be elevated for years even after successful treatment with antibiotics.[27] As antibody levels are not indicative of treatment success, testing for them is not recommended.[27]

Facial palsy may resolve without treatment; however, antibiotic treatment is recommended to stop other Lyme complications.[35] Corticosteroids are not recommended when facial palsy is caused by Lyme disease.[41] In those with facial palsy, frequent use of artificial tears while awake is recommended, along with ointment and a patch or taping the eye closed when sleeping.[41][184]

About a third of people with Lyme carditis need a temporary pacemaker until their heart conduction abnormality resolves, and 21% need to be hospitalized.[43] Lyme carditis should not be treated with corticosteroids.[43]

People with Lyme arthritis should limit their level of physical activity to avoid damaging affected joints, and in case of limping should use crutches.[185] Pain associated with Lyme disease may be treated with nonsteroidal anti-inflammatory drugs (NSAIDs).[35] Corticosteroid joint injections are not recommended for Lyme arthritis that is being treated with antibiotics.[35][185] People with Lyme arthritis treated with intravenous antibiotics or two months of oral antibiotics who continue to have joint swelling two months after treatment and have negative PCR test for Borrelia DNA in the synovial fluid are said to have post-antibiotic Lyme arthritis; this is more common after infection by certain Borrelia strains in people with certain genetic and immunologic characteristics.[35][52][185] Post-antibiotic Lyme arthritis may be symptomatically treated with NSAIDs, disease-modifying antirheumatic drugs (DMARDs), arthroscopic synovectomy, or physical therapy.[52][185]

People receiving treatment should be advised that reinfection is possible and how to prevent it.[112]

Prognosis edit

Lyme disease's typical first sign, the erythema migrans (EM) rash, resolves within several weeks even without treatment.[3] However, in untreated people, the infection often disseminates to the nervous system, heart, or joints, possibly causing permanent damage to body tissues.[35]

People who receive recommended antibiotic treatment within several days of appearance of an initial EM rash have the best prospects.[110] Recovery may not be total or immediate. The percentage of people achieving full recovery in the United States increases from about 64–71% at end of treatment for EM rash to about 84–90% after 30 months; higher percentages are reported in Europe.[186][187] Treatment failure, i.e. persistence of original or appearance of new signs of the disease, occurs only in a few people.[186] Remaining people are considered cured but continue to experience subjective symptoms, e.g. joint or muscle pains or fatigue.[188] These symptoms usually are mild and nondisabling.[188]

People treated only after nervous system manifestations of the disease may end up with objective neurological deficits, in addition to subjective symptoms.[35] In Europe, an average of 32–33 months after initial Lyme symptoms in people treated mostly with doxycycline 200 mg for 14–21 days, the percentage of people with lingering symptoms was much higher among those diagnosed with neuroborreliosis (50%) than among those with only an EM rash (16%).[189] In another European study, 5 years after treatment for neuroborreliosis, lingering symptoms were less common among children (15%) than adults (30%), and in the latter was less common among those treated within 30 days of the first symptom (16%) than among those treated later (39%); among those with lingering symptoms, 54% had daily activities restricted and 19% were on sick leave or incapacitated.[190]

Some data suggest that about 90% of Lyme facial palsies treated with antibiotics recover fully a median of 24 days after appearing and most of the rest recover with only mild abnormality.[191][192] However, in Europe 41% of people treated for facial palsy had other lingering symptoms at followup up to 6 months later, including 28% with numbness or altered sensation and 14% with fatigue or concentration problems.[192] Palsies in both sides of the face are associated with worse and longer time to recovery.[191][192] Historical data suggests that untreated people with facial palsies recover at nearly the same rate, but 88% subsequently have Lyme arthritis.[191][193] Other research shows that synkinesis (involuntary movement of a facial muscle when another one is voluntarily moved) can become evident only 6–12 months after facial palsy appears to be resolved, as damaged nerves regrow and sometimes connect to incorrect muscles.[194] Synkinesis is associated with corticosteroid use.[194] In longer-term follow-up, 16–23% of Lyme facial palsies do not fully recover.[194]

In Europe, about a quarter of people with Bannwarth syndrome (Lyme radiculopathy and lymphocytic meningitis) treated with intravenous ceftriaxone for 14 days an average of 30 days after first symptoms had to be retreated 3–6 months later because of unsatisfactory clinical response or continued objective markers of infection in cerebrospinal fluid; after 12 months, 64% recovered fully, 31% had nondisabling mild or infrequent symptoms that did not require regular use of analgesics, and 5% had symptoms that were disabling or required substantial use of analgesics.[40] The most common lingering nondisabling symptoms were headache, fatigue, altered sensation, joint pains, memory disturbances, malaise, radicular pain, sleep disturbances, muscle pains, and concentration disturbances. Lingering disabling symptoms included facial palsy and other impaired movement.[40]

Recovery from late neuroborreliosis tends to take longer and be less complete than from early neuroborreliosis, probably because of irreversible neurologic damage.[35]

About half the people with Lyme carditis progress to complete heart block, but it usually resolves in a week.[43] Other Lyme heart conduction abnormalities resolve typically within 6 weeks.[43] About 94% of people have full recovery, but 5% need a permanent pacemaker and 1% end up with persistent heart block (the actual percentage may be higher because of unrecognized cases).[43] Lyme myocardial complications usually are mild and self-limiting.[43] However, in some cases Lyme carditis can be fatal.[43]

Recommended antibiotic treatments are effective in about 90% of Lyme arthritis cases, although it can take several months for inflammation to resolve and a second round of antibiotics is often necessary.[35] Antibiotic-refractory Lyme arthritis also eventually resolves, typically within 9–14 months (range 4 months – 4 years); DMARDs or synovectomy can accelerate recovery.[185]

Reinfection is not uncommon. In a U.S. study, 6–11% of people treated for an EM rash had another EM rash within 30 months.[186] The second rash typically is due to infection by a different Borrelia strain.[195]

Post-treatment Lyme disease syndrome edit

Chronic symptoms like pain, fatigue, or cognitive impairment are experienced by 10–20% of people who contract Lyme disease, even after completing treatment.[196][197] The cause is unknown. One hypothesis is autoimmunity triggered by the infection, which may persist long-term after the triggering infection is resolved. Auto–immune responses are known to occur following other infections, including Campylobacter (Guillain-Barré syndrome), Chlamydia (reactive arthritis), and strep throat (rheumatic heart disease). Another is that a persistent, difficult to detect infection causes PTLDS. A third proposal is that symptoms are unrelated to a previous case of Lyme Disease, but have other causes.[197]

There is no proven treatment for PTLDS. While short-term antibiotics are effective in early Lyme disease, prolonged antibiotics are not. They have been shown ineffective in placebo-controlled trials and carry the risk of serious, sometimes deadly complications. Generally, treatment is symptomatic and is similar to the management of fibromyalgia or ME/CFS. PTLDS usually gets better over time, but recovery may take many months.[197]

Epidemiology edit

 
Countries with reported Lyme disease cases

Lyme disease occurs regularly in Northern Hemisphere temperate regions.[198] An estimated 476,000 people a year are diagnosed and treated for the disease in the United States. This number is likely an overestimate due to overdiagnosis and overtreatment.[5][6] Over 200,000 people a year are diagnosed and treated in Europe.[7][3][199] There is a suggestion that tick populations and Lyme disease occurrence are increasing and spreading into new areas, due in part to the warming temperatures of climate change. However, tick-borne disease systems are complex, and determining whether changes are due to climate change or other drivers can be difficult.[153][200]

Africa edit

In northern Africa, B. burgdorferi sensu lato has been identified in Morocco, Algeria, Egypt and Tunisia.[201][202][203]

Lyme disease in sub-Saharan Africa is presently unknown, but evidence indicates it may occur in humans in this region. The abundance of hosts and tick vectors would favor the establishment of Lyme infection in Africa.[204] In East Africa, two cases of Lyme disease have been reported in Kenya.[205] According The Federation of Infectious Diseases Societies of Southern Africa, Lyme disease is not known to be endemic in either South Africa or Mozambique.[206]

Asia edit

B. burgdorferi sensu lato-infested ticks are being found more frequently in Japan, as well as in northwest China, Nepal, Thailand and far eastern Russia.[207][208] Borrelia has also been isolated in Mongolia.[209]

Australia edit

Lyme disease is not considered endemic to Australia.[210] While there have been reports of people acquiring Lyme disease in Australia, and even evidence of closely related Borrelia species in ticks,[211] the evidence linking these cases to local transmission is limited. Ongoing research on resolving potential Borrelia species to Debilitating Symptom Complexes Attributed to Ticks (DSCATT) in Australia are ongoing.[212]

Europe edit

In Europe, Lyme disease is caused by infection with one or more pathogenic European genospecies of the spirochaete B. burgdorferi sensu lato, mainly transmitted by the tick Ixodes ricinus.[213] Cases of B. burgdorferi sensu lato-infected ticks are found predominantly in central Europe, particularly in Slovenia and Austria, but have been isolated in almost every country on the continent.[214] Number of cases in southern Europe, such as Italy and Portugal, is much lower.[215] Diagnosed cases in some Western countries, such as Iceland, are rising.[216] Lyme disease is rare in Iceland. On average around 6 to 7 cases are diagnosed every year, primarily localised infections presenting as erythema migrans. None of the cases had a definitive Icelandic origin and the yearly number of cases has not been increasing.[217]

United Kingdom edit

In the United Kingdom the number of laboratory-confirmed cases of Lyme disease has been rising steadily since voluntary reporting was introduced in 1986[218] when 68 cases were recorded in the UK and Ireland combined.[219] In the UK there were 23 confirmed cases in 1988 and 19 in 1990,[220] but 973 in 2009[218] and 953 in 2010.[221] Provisional figures for the first 3 quarters of 2011 show a 26% increase on the same period in 2010.[222]

It is thought, however, that the actual number of cases is significantly higher than suggested by the above figures, with the UK's Health Protection Agency estimating that there are between 2,000 and 3,000 cases per year,[221] (with an average of around 15% of the infections acquired overseas[218]), while Dr Darrel Ho-Yen, Director of the Scottish Toxoplasma Reference Laboratory and National Lyme Disease Testing Service, believes that the number of confirmed cases should be multiplied by 10 "to take account of wrongly diagnosed cases, tests giving false results, sufferers who weren't tested, people who are infected but not showing symptoms, failures to notify and infected individuals who don't consult a doctor."[223][224]

Despite Lyme disease (Borrelia burgdorferi infection) being a notifiable disease in Scotland[225] since January 1990[226] which should therefore be reported on the basis of clinical suspicion, it is believed that many GPs are unaware of the requirement.[227] Mandatory reporting, limited to laboratory test results only, was introduced throughout the UK in October 2010, under the Health Protection (Notification) Regulations 2010.[218]

Although there is a greater number of cases of Lyme disease in the New Forest, Salisbury Plain, Exmoor, the South Downs, parts of Wiltshire and Berkshire, Thetford Forest[228] and the West coast and islands of Scotland,[229] infected ticks are widespread, and can even be found in the parks of London.[220][230] A 1989 report found that 25% of forestry workers in the New Forest were seropositive, as were between 2% and 4–5% of the general local population of the area.[231][232]

Tests on pet dogs carried out throughout the country in 2009 indicated that around 2.5% of ticks in the UK may be infected, considerably higher than previously thought.[233][234] It is speculated that global warming may lead to an increase in tick activity in the future, as well as an increase in the amount of time that people spend in public parks, thus increasing the risk of infection.[235] However no published research has proven this to be so.

North America edit

Many studies in North America have examined ecological and environmental correlates of the number of people affected by Lyme disease. A 2005 study using climate suitability modelling of I. scapularis projected that climate change would cause an overall 213% increase in suitable vector habitat by 2080, with northward expansions in Canada, increased suitability in the central U.S., and decreased suitable habitat and vector retraction in the southern U.S.[236] A 2008 review of published studies concluded that the presence of forests or forested areas was the only variable that consistently elevated the risk of Lyme disease whereas other environmental variables showed little or no concordance between studies.[237] The authors argued that the factors influencing tick density and human risk between sites are still poorly understood, and that future studies should be conducted over longer time periods, become more standardized across regions, and incorporate existing knowledge of regional Lyme disease ecology.[237]

Canada edit

The range of ticks able to carry Lyme disease has expanded from a limited area of Ontario to include areas of southern Quebec, Manitoba, northern Ontario, southern New Brunswick, southwest Nova Scotia and limited parts of Saskatchewan and Alberta, as well as British Columbia. Cases have been reported as far east as the island of Newfoundland.[109][238][239][240] A model-based prediction by Leighton et al. (2012) suggests that the range of the I. scapularis tick will expand into Canada by 46 km/year over the next decade, with warming climatic temperatures as the main driver of increased speed of spread.[241]

Mexico edit

A 2007 study suggests Borrelia burgdorferi infections are endemic to Mexico, from four cases reported between 1999 and 2000.[242]

United States edit

 
CDC map showing the risk of Lyme disease in the United States, particularly its concentration in the Northeast Megalopolis and western Wisconsin

Lyme disease is the most common tick-borne disease in North America and Europe, and one of the fastest-growing infectious diseases in the United States. Of cases reported to the United States CDC, the ratio of Lyme disease infection is 7.9 cases for every 100,000 persons. In the ten states where Lyme disease is most common, the average was 31.6 cases for every 100,000 persons for the year 2005.[243][244][245]

Although Lyme disease has been reported in all states due to travel-associated infections, about 99% of all reported cases are confined to just five geographic areas (New England, Mid-Atlantic, East-North Central, South Atlantic, and West North-Central).[83][246][247] CDC implemented national surveillance of Lyme disease cases in 1991. Since then, reporting criteria has been modified multiple times.[248] The 2022 surveillance case definition classifies cases as confirmed, probable, and suspect.[249]

CDC emphasizes that, while surveillance data has limitations, it is useful due to "uniformity, simplicity, and timeliness." While cases are under-reported in high-incidence areas, over-reporting is likely in low-incidence areas. Additionally, surveillance cases are reported by county of residence and not where an infection was necessarily contracted.[250][83]

The number of reported cases of the disease has been increasing, as are endemic regions in North America. For example, B. burgdorferi sensu lato was previously thought to be hindered in its ability to be maintained in an enzootic cycle in California, because it was assumed the large lizard population would dilute the number of people affected by B. burgdorferi in local tick populations; this has since been brought into question, as some evidence has suggested lizards can become infected.[251]

Except for one study in Europe,[252] much of the data implicating lizards is based on DNA detection of the spirochete and has not demonstrated that lizards are able to infect ticks feeding upon them.[251][253][254][255] As some experiments suggest lizards are refractory to infection with Borrelia, it appears likely their involvement in the enzootic cycle is more complex and species-specific.[77]

While B. burgdorferi is most associated with ticks hosted by white-tailed deer and white-footed mice, Borrelia afzelii is most frequently detected in rodent-feeding vector ticks, and Borrelia garinii and Borrelia valaisiana appear to be associated with birds. Both rodents and birds are competent reservoir hosts for B. burgdorferi sensu stricto. The resistance of a genospecies of Lyme disease spirochetes to the bacteriolytic activities of the alternative complement pathway of various host species may determine its reservoir host association.[252]

Several similar but apparently distinct conditions may exist, caused by various species or subspecies of Borrelia in North America. A regionally restricted condition that may be related to Borrelia infection is southern tick-associated rash illness (STARI), also known as Masters disease. Amblyomma americanum, known commonly as the lone-star tick, is recognized as the primary vector for STARI. In some parts of the geographical distribution of STARI, Lyme disease is quite rare (e.g., Arkansas), so people in these regions experiencing Lyme-like symptoms—especially if they follow a bite from a lone-star tick—should consider STARI as a possibility. It is generally a milder condition than Lyme and typically responds well to antibiotic treatment.[256]

In recent years there have been 5 to 10 cases a year of a disease similar to Lyme occurring in Montana. It occurs primarily in pockets along the Yellowstone River in central Montana. People have developed a red bull's-eye rash around a tick bite followed by weeks of fatigue and a fever.[247]

Lyme disease effects are comparable among males and females. A wide range of age groups is affected, though the number of cases is highest among 10- to 19-year-olds. For unknown reasons, Lyme disease is seven times more common among Asians.[257]

South America edit

In Brazil, a Lyme-like disease known as Baggio–Yoshinari syndrome was identified, caused by microorganisms that do not belong to the B. burgdorferi sensu lato complex and transmitted by ticks of the Amblyomma and Rhipicephalus genera.[258] The first reported case of BYS in Brazil was made in 1992 in Cotia, São Paulo.[259]

Etymology edit

Lyme disease was diagnosed as a separate condition for the first time in 1975 in Lyme, Connecticut.[260]

History edit

The evolutionary history of Borrelia burgdorferi genetics has been the subject of recent studies. One study has found that prior to the reforestation that accompanied post-colonial farm abandonment in New England and the wholesale migration into the mid-west that occurred during the early 19th century, Lyme disease was present for thousands of years in America and had spread along with its tick hosts from the Northeast to the Midwest.[261]

John Josselyn, who visited New England in 1638 and again from 1663 to 1670, wrote "there be infinite numbers of ticks hanging upon the bushes in summertime that will cleave to man's garments and creep into his breeches, eating themselves in a short time into the very flesh of a man. I have seen the stockings of those that have gone through the woods covered with them."[262]

This is also confirmed by the writings of Peter Kalm, a Swedish botanist who was sent to America by Linnaeus, and who found the forests of New York "abound" with ticks when he visited in 1749. When Kalm's journey was retraced 100 years later, the forests were gone and the Lyme bacterium had probably become isolated to a few pockets along the northeast coast, Wisconsin, and Minnesota.[263]

Perhaps the first detailed description of what is now known as Lyme disease appeared in the writings of John Walker after a visit to the island of Jura (Deer Island) off the west coast of Scotland in 1764.[264] He gives a good description both of the symptoms of Lyme disease (with "exquisite pain [in] the interior parts of the limbs") and of the tick vector itself, which he describes as a "worm" with a body which is "of a reddish color and of a compressed shape with a row of feet on each side" that "penetrates the skin". Many people from this area of Great Britain emigrated to North America between 1717 and the end of the 18th century.[citation needed]

The examination of preserved museum specimens has found Borrelia DNA in an infected Ixodes ricinus tick from Germany that dates back to 1884, and from an infected mouse from Cape Cod that died in 1894.[263] The 2010 autopsy of Ötzi the Iceman, a 5,300-year-old mummy, revealed the presence of the DNA sequence of Borrelia burgdorferi making him the earliest known human with Lyme disease.[265]

The early European studies of what is now known as Lyme disease described its skin manifestations. The first study dates to 1883 in Breslau, Germany (now Wrocław, Poland), where physician Alfred Buchwald described a man who for 16 years had had a degenerative skin disorder now known as acrodermatitis chronica atrophicans.[266]

At a 1909 research conference, Swedish dermatologist Arvid Afzelius presented a study about an expanding, ring-like lesion he had observed in an older woman following the bite of a sheep tick. He named the lesion erythema migrans.[266] The skin condition now known as borrelial lymphocytoma was first described in 1911.[267]

The modern history of medical understanding of the disease, including its cause, diagnosis, and treatment, has been difficult.[268]

Neurological problems following tick bites were recognized starting in the 1920s. French physicians Garin and Bujadoux described a farmer with a painful sensory radiculitis accompanied by mild meningitis following a tick bite. A large, ring-shaped rash was also noted, although the doctors did not relate it to the meningoradiculitis. In 1930, the Swedish dermatologist Sven Hellerström was the first to propose EM and neurological symptoms following a tick bite were related.[269] In the 1940s, German neurologist Alfred Bannwarth described several cases of chronic lymphocytic meningitis and polyradiculoneuritis, some of which were accompanied by erythematous skin lesions.

Carl Lennhoff, who worked at the Karolinska Institute in Sweden, believed many skin conditions were caused by spirochetes. In 1948, he used a special stain to microscopically observe what he believed were spirochetes in various types of skin lesions, including EM.[270] Although his conclusions were later shown to be erroneous, interest in the study of spirochetes was sparked. In 1949, Nils Thyresson, who also worked at the Karolinska Institute, was the first to treat ACA with penicillin.[271] In the 1950s, the relationship among tick bite, lymphocytoma, EM and Bannwarth's syndrome was recognized throughout Europe leading to the widespread use of penicillin for treatment in Europe.[272][273]

In 1970, a dermatologist in Wisconsin named Rudolph Scrimenti recognized an EM lesion in a person after recalling a paper by Hellerström that had been reprinted in an American science journal in 1950. This was the first documented case of EM in the United States. Based on the European literature, he treated the person with penicillin.[274]

The full syndrome now known as Lyme disease was not recognized until a cluster of cases originally thought to be juvenile rheumatoid arthritis was identified in three towns in southeastern Connecticut in 1975, including the towns Lyme and Old Lyme, which gave the disease its popular name.[275] This was investigated by physicians David Snydman and Allen Steere of the Epidemic Intelligence Service, and by others from Yale University, including Stephen Malawista, who is credited as a co-discover of the disease.[276] The recognition that the people in the United States had EM led to the recognition that "Lyme arthritis" was one manifestation of the same tick-borne condition known in Europe.[277]

Before 1976, the elements of B. burgdorferi sensu lato infection were called or known as tick-borne meningopolyneuritis, Garin-Bujadoux syndrome, Bannwarth syndrome, Afzelius's disease,[278] Montauk Knee or sheep tick fever. Since 1976 the disease is most often referred to as Lyme disease,[279][280] Lyme borreliosis or simply borreliosis.[281][282]

In 1980, Steere, et al., began to test antibiotic regimens in adults with Lyme disease.[283] In the same year, New York State Health Dept. epidemiologist Jorge Benach provided Willy Burgdorfer, a researcher at the Rocky Mountain Biological Laboratory, with collections of I. dammini [scapularis] from Shelter Island, New York, a known Lyme-endemic area as part of an ongoing investigation of Rocky Mountain spotted fever. In examining the ticks for rickettsiae, Burgdorfer noticed "poorly stained, rather long, irregularly coiled spirochetes." Further examination revealed spirochetes in 60% of the ticks. Burgdorfer credited his familiarity with the European literature for his realization that the spirochetes might be the "long-sought cause of ECM and Lyme disease." Benach supplied him with more ticks from Shelter Island and sera from people diagnosed with Lyme disease. University of Texas Health Science Center researcher Alan Barbour "offered his expertise to culture and immunochemically characterize the organism." Burgdorfer subsequently confirmed his discovery by isolating, from people with Lyme disease, spirochetes identical to those found in ticks.[284] In June 1982, he published his findings in Science, and the spirochete was named Borrelia burgdorferi in his honor.[285]

After the identification of B. burgdorferi as the causative agent of Lyme disease, antibiotics were selected for testing, guided by in vitro antibiotic sensitivities, including tetracycline antibiotics, amoxicillin, cefuroxime axetil, intravenous and intramuscular penicillin and intravenous ceftriaxone.[286][287] The mechanism of tick transmission was also the subject of much discussion. B. burgdorferi spirochetes were identified in tick saliva in 1987, confirming the hypothesis that transmission occurred via tick salivary glands.[288]

Society, culture, & controversy edit

Landscape changes & urbanization

Urbanization and other anthropogenic factors can be implicated in the spread of Lyme disease to humans. In many areas, expansion of suburban neighborhoods has led to gradual deforestation of surrounding wooded areas and increased border contact between humans and tick-dense areas. Human expansion has also resulted in a reduction of predators that hunt deer as well as mice, chipmunks and other small rodents—the primary reservoirs for Lyme disease. As a consequence of increased human contact with host and vector, the likelihood of transmission of the disease has greatly increased.[289][290] Researchers are investigating possible links between global warming and the spread of vector-borne diseases, including Lyme disease.[291]

The dilution effect

Given these habitat-host dynamics, some researchers have begun to postulate whether the dilution effect could mitigate the spread of Lyme disease.[289] The dilution effect is a hypothesis that predicts that an increase in host biodiversity will result in a decrease in the number of vectors infected with B. burgdorferi.[289] Scientific research has shown that nymphal infection prevalence (NIP) decreases as the number of host species increases, supporting the dilution effect.[289] That said, these findings should not be misinterpreted to suggest that there is a direct relationship between decreased NIP and decreased epidemiological risk, as this has yet to be proven.[289] Additionally, it is important to note that, thus far, the dilution effect is only supported in the Northeastern United States, and has been disproved in other parts of the world that also experience high Lyme disease incidence rates [292]

Chronic Lyme disease

The term "chronic Lyme disease" is controversial and not recognized in the medical literature,[293] and most medical authorities advise against long-term antibiotic treatment for Lyme disease.[35][117][294] Studies have shown that most people diagnosed with "chronic Lyme disease" either have no objective evidence of previous or current infection with B. burgdorferi or are people who should be classified as having post-treatment Lyme disease syndrome (PTLDS), which is defined as continuing or relapsing non-specific symptoms (such as fatigue, musculoskeletal pain, and cognitive complaints) in a person previously treated for Lyme disease.[295]

The 2008 documentary Under Our Skin is known for promoting controversial and unrecognized theories about "chronic Lyme disease".[296]

Other animals edit

Dogs edit

Prevention of Lyme disease is an important step in keeping dogs safe in endemic areas. Prevention education and a number of preventive measures are available. First, for dog owners who live near or who often frequent tick-infested areas, routine vaccinations of their dogs is an important step.[297]

Another crucial preventive measure is the use of persistent acaricides, such as topical repellents or pesticides that contain triazapentadienes (Amitraz), phenylpyrazoles (Fipronil), or permethrin (pyrethroids).[298] These acaricides target primarily the adult stages of Lyme-carrying ticks and reduce the number of reproductively active ticks in the environment.[297] Formulations of these ingredients are available in a variety of topical forms, including spot-ons, sprays, powders, impregnated collars, solutions, and shampoos.[298]

Examination of a dog for ticks after being in a tick-infested area is an important precautionary measure to take in the prevention of Lyme disease. Key spots to examine include the head, neck, and ears.[299]

In dogs, a serious long-term prognosis may result in glomerular disease,[300] which is a category of kidney damage that may cause chronic kidney disease.[181] Dogs may also experience chronic joint disease if the disease is left untreated. However, the majority of cases of Lyme disease in dogs result in complete recovery with, and sometimes without, treatment with antibiotics.[301][verification needed] In rare cases, Lyme disease can be fatal to both humans and dogs.[302]

Cats edit

Unlike dogs, it is very rare for a cat to be infected with Lyme disease. However, cats are nevertheless capable of being infected with B. burgdorferi , following a bite from an infected tick. Cats who are infected with Lyme Disease may show symptoms including but not limited to lameness, fatigue, or loss of appetite.[303] In two cases, the infected cats experienced cardiac irregularities similar to symptoms of Lyme in both dogs and humans.[304] However, cats who are infected with Lyme disease are likely to be asymptomatic, and show no noticeable signs of the disease.[304][303] Cats with Lyme are often treated with antibiotics, much like other animals. In some cases, additional treatment or therapy may be required.[303]

Horses edit

While Lyme disease can occur in horses, not every infection with B. burgdorferi is associated with symptoms.[305] Especially, detection of specific antibodies against B. burgdorferi alone is not sufficient for a diagnosis of equine Lyme disease and unspecific testing for antibodies is not recommended.[306]

References edit

  1. ^ a b c d e f g h i j k . cdc.gov. 11 January 2013. Archived from the original on 16 January 2013. Retrieved 2 March 2015.
  2. ^ "Lyme disease - Symptoms and causes". Mayo Clinic. Retrieved 7 June 2022.
  3. ^ a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae Shapiro ED (May 2014). (PDF). The New England Journal of Medicine. 370 (18): 1724–1731. doi:10.1056/NEJMcp1314325. PMC 4487875. PMID 24785207. Archived from the original (PDF) on 21 August 2016. Retrieved 5 July 2016.
  4. ^ a b "Lyme Disease Diagnosis and Testing". cdc.gov. 10 January 2013. from the original on 2 March 2015. Retrieved 2 March 2015.
  5. ^ a b "How many people get Lyme disease?". Centers for Disease Control and Prevention. 13 January 2021. Retrieved 11 January 2023.
  6. ^ a b Kugeler KJ, Schwartz AM, Delorey MJ, Mead PS, Hinckley AF (February 2021). "Estimating the Frequency of Lyme Disease Diagnoses, United States, 2010-2018". Emerging Infectious Diseases. 27 (2): 616–619. doi:10.3201/eid2702.202731. PMC 7853543. PMID 33496229.
  7. ^ a b c d Marques AR, Strle F, Wormser GP (August 2021). "Comparison of Lyme Disease in the United States and Europe". Emerging Infectious Diseases. 27 (8): 2017–2024. doi:10.3201/eid2708.204763. PMC 8314816. PMID 34286689.
  8. ^ a b c Wenner M (11 June 2021). "Let's Do a Tick Check - These pervasive bloodsuckers can give you more than just Lyme disease. Here's how to protect yourself. (Interactive)". The New York Times. Retrieved 19 June 2021.
  9. ^ a b Wolcott KA, Margos G, Fingerle V, Becker NS (September 2021). "Host association of Borrelia burgdorferi sensu lato: A review". Ticks and Tick-Borne Diseases. 12 (5): 101766. doi:10.1016/j.ttbdis.2021.101766. PMID 34161868.
  10. ^ Aucott J, Morrison C, Munoz B, Rowe PC, Schwarzwalder A, West SK (June 2009). "Diagnostic challenges of early Lyme disease: lessons from a community case series". BMC Infectious Diseases. 9: 79. doi:10.1186/1471-2334-9-79. PMC 2698836. PMID 19486523.
  11. ^ Aucott JN (June 2015). "Posttreatment Lyme disease syndrome". Infectious Disease Clinics of North America. 29 (2): 309–323. doi:10.1016/j.idc.2015.02.012. PMID 25999226.
  12. ^ Johnson RC (1996). "Borrelia". In Baron S, et al. (eds.). Baron's Medical Microbiology (4th ed.). Univ of Texas Medical Branch. ISBN 978-0-9631172-1-2. PMID 21413339. from the original on 7 February 2009.
  13. ^ a b "Lyme disease transmission". cdc.gov. 11 January 2013. from the original on 3 March 2015. Retrieved 2 March 2015.
  14. ^ a b Steere AC, Strle F, Wormser GP, Hu LT, Branda JA, Hovius JW, et al. (December 2016). "Lyme borreliosis". Nature Reviews. Disease Primers. 2: 16090. doi:10.1038/nrdp.2016.90. PMC 5539539. PMID 27976670.
  15. ^ "Lyme borreliosis" (PDF). ECDC. (PDF) from the original on 29 September 2018. Retrieved 29 September 2018.
  16. ^ a b Pritt BS, Mead PS, Johnson DK, Neitzel DF, Respicio-Kingry LB, Davis JP, et al. (May 2016). "Identification of a novel pathogenic Borrelia species causing Lyme borreliosis with unusually high spirochaetaemia: a descriptive study". The Lancet. Infectious Diseases. 16 (5): 556–564. doi:10.1016/S1473-3099(15)00464-8. PMC 4975683. PMID 26856777.
  17. ^ "Two-step Laboratory Testing Process". cdc.gov. 15 November 2011. from the original on 12 March 2015. Retrieved 2 March 2015.
  18. ^ "Testing of Ticks". cdc.gov. 4 June 2013. from the original on 19 February 2015. Retrieved 2 March 2015. Although some commercial groups offer testing, in general it is not recommended
  19. ^ "Tick Removal". cdc.gov. 23 June 2014. from the original on 10 March 2015. Retrieved 2 March 2015.
  20. ^ a b c "Post-Treatment Lyme Disease Syndrome". cdc.gov. 1 December 2017. from the original on 27 February 2015. Retrieved 20 June 2018.
  21. ^ Regional Disease Vector Ecology Profile: Central Europe. DIANE Publishing. April 2001. p. 136. ISBN 978-1-4289-1143-7. from the original on 8 September 2017.
  22. ^ Williams C (2007). Infectious disease epidemiology : theory and practice (2nd ed.). Sudbury, Mass.: Jones and Bartlett Publishers. p. 447. ISBN 978-0-7637-2879-3. from the original on 8 September 2017.
  23. ^ "Willy Burgdorfer – obituary". Daily Telegraph. 1 December 2014. from the original on 1 December 2014. Retrieved 1 December 2014.
  24. ^ Baker PJ (November 2010). "Chronic Lyme disease: in defense of the scientific enterprise". FASEB Journal. 24 (11): 4175–4177. doi:10.1096/fj.10-167247. PMID 20631327. S2CID 36141950.
  25. ^ Lantos PM (June 2015). "Chronic Lyme disease". Infectious Disease Clinics of North America. 29 (2): 325–340. doi:10.1016/j.idc.2015.02.006. PMC 4477530. PMID 25999227.
  26. ^ a b "Lyme disease vaccine". Centers for Disease Control and Prevention. 11 August 2022. Retrieved 7 June 2023.
  27. ^ a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac Wright WF, Riedel DJ, Talwani R, Gilliam BL (June 2012). "Diagnosis and management of Lyme disease". American Family Physician. 85 (11): 1086–1093. PMID 22962880. from the original on 27 September 2013.
  28. ^ a b c d Tibbles CD, Edlow JA (June 2007). "Does this patient have erythema migrans?". JAMA. 297 (23): 2617–2627. doi:10.1001/jama.297.23.2617. PMID 17579230.
  29. ^ a b c "Lyme disease rashes and look-alikes". Lyme Disease. Centers for Disease Control and Prevention. 21 December 2018. from the original on 2 April 2019. Retrieved 18 April 2019.
  30. ^ a b c "Lyme disease: erythema migrans". Lyme disease NICE guideline [NG95]. National Institute for Health and Care Excellence. from the original on 9 May 2019. Retrieved 8 May 2019.
  31. ^ "Lyme Disease". The Lecturio Medical Concept Library. Retrieved 9 July 2021.
  32. ^ a b Lyme disease at eMedicine
  33. ^ Steere AC, Sikand VK, Schoen RT, Nowakowski J (August 2003). "Asymptomatic infection with Borrelia burgdorferi". Clinical Infectious Diseases. 37 (4): 528–532. doi:10.1086/376914. PMID 12905137. (primary source)
  34. ^ a b Biesiada G, Czepiel J, Leśniak MR, Garlicki A, Mach T (December 2012). "Lyme disease: review". Archives of Medical Science. 8 (6): 978–982. doi:10.5114/aoms.2012.30948. PMC 3542482. PMID 23319969.
  35. ^ a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae af ag ah Wormser GP, Dattwyler RJ, Shapiro ED, Halperin JJ, Steere AC, Klempner MS, et al. (November 2006). "The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: clinical practice guidelines by the Infectious Diseases Society of America". Clinical Infectious Diseases. 43 (9): 1089–1134. doi:10.1086/508667. PMID 17029130.
  36. ^ a b Halperin JJ (June 2008). "Nervous system Lyme disease". Infectious Disease Clinics of North America. 22 (2): 261–74, vi. doi:10.1016/j.idc.2007.12.009. PMID 18452800. S2CID 10590435.
  37. ^ a b c d e f g h i j Mygland A, Ljøstad U, Fingerle V, Rupprecht T, Schmutzhard E, Steiner I (January 2010). "EFNS guidelines on the diagnosis and management of European Lyme neuroborreliosis" (PDF). European Journal of Neurology. 17 (1): 8–16. doi:10.1111/j.1468-1331.2009.02862.x. PMID 19930447. S2CID 14166137. (PDF) from the original on 8 August 2017. Retrieved 30 April 2019.
  38. ^ a b c d e Pachner AR, Steere AC (July 1984). "Neurological findings of Lyme disease". The Yale Journal of Biology and Medicine. 57 (4): 481–483. PMC 2590042. PMID 6516450.
  39. ^ Moscatello AL, Worden DL, Nadelman RB, Wormser G, Lucente F (June 1991). "Otolaryngologic aspects of Lyme disease". The Laryngoscope. 101 (6 Pt 1): 592–595. doi:10.1288/00005537-199106000-00004. PMID 2041438. S2CID 35345898.
  40. ^ a b c d e f g h Ogrinc K, Lusa L, Lotrič-Furlan S, Bogovič P, Stupica D, Cerar T, et al. (August 2016). "Course and Outcome of Early European Lyme Neuroborreliosis (Bannwarth Syndrome): Clinical and Laboratory Findings". Clinical Infectious Diseases. 63 (3): 346–353. doi:10.1093/cid/ciw299. PMID 27161773.
  41. ^ a b c d e f g h i Garro A, Nigrovic LE (May 2018). "Managing Peripheral Facial Palsy". Annals of Emergency Medicine. 71 (5): 618–624. doi:10.1016/j.annemergmed.2017.08.039. PMID 29110887.
  42. ^ a b Halperin JJ (September 2010). "Nervous system Lyme disease". The Journal of the Royal College of Physicians of Edinburgh. 40 (3): 248–255. doi:10.4997/JRCPE.2010.314. PMID 21127770.
  43. ^ a b c d e f g h i j k l m n o Fish AE, Pride YB, Pinto DS (June 2008). "Lyme carditis" (PDF). Infectious Disease Clinics of North America. 22 (2): 275–88, vi. doi:10.1016/j.idc.2007.12.008. PMID 18452801. (PDF) from the original on 29 August 2017. Retrieved 9 May 2019.
  44. ^ a b Stanek G, Wormser GP, Gray J, Strle F (February 2012). "Lyme borreliosis". Lancet. 379 (9814): 461–473. doi:10.1016/S0140-6736(11)60103-7. PMID 21903253. S2CID 31461047.
  45. ^ a b Stanek G, Strle F (June 2008). "Lyme disease: European perspective". Infectious Disease Clinics of North America. 22 (2): 327–39, vii. doi:10.1016/j.idc.2008.01.001. PMID 18452805.
  46. ^ Guardado KE, Sergent S (January 2022). "Pediatric unilateral knee swelling: a case report of a complicated differential diagnosis and often overlooked cause". Journal of Osteopathic Medicine. 122 (2): 105–109. doi:10.1515/jom-2020-0332. PMID 34989214. S2CID 245705028.
  47. ^ Puius YA, Kalish RA (June 2008). "Lyme arthritis: pathogenesis, clinical presentation, and management". Infectious Disease Clinics of North America. 22 (2): 289–300, vi–vii. doi:10.1016/j.idc.2007.12.014. PMID 18452802.
  48. ^ a b Wormser GP, Strle F, Shapiro ED, Dattwyler RJ, Auwaerter PG (February 2017). "A critical appraisal of the mild axonal peripheral neuropathy of late neurologic Lyme disease". Diagnostic Microbiology and Infectious Disease. 87 (2): 163–167. doi:10.1016/j.diagmicrobio.2016.11.003. PMC 5924701. PMID 27914746.
  49. ^ Bratton RL, Whiteside JW, Hovan MJ, Engle RL, Edwards FD (May 2008). "Diagnosis and treatment of Lyme disease". Mayo Clinic Proceedings. 83 (5): 566–571. doi:10.4065/83.5.566. PMID 18452688.
  50. ^ Shadick NA, Phillips CB, Sangha O, Logigian EL, Kaplan RF, Wright EA, et al. (December 1999). "Musculoskeletal and neurologic outcomes in patients with previously treated Lyme disease". Annals of Internal Medicine. 131 (12): 919–926. doi:10.7326/0003-4819-131-12-199912210-00003. PMID 10610642. S2CID 20746489.
  51. ^ Seltzer EG, Gerber MA, Cartter ML, Freudigman K, Shapiro ED (February 2000). "Long-term outcomes of persons with Lyme disease". JAMA. 283 (5): 609–616. doi:10.1001/jama.283.5.609. PMID 10665700.
  52. ^ a b c d e f Lantos PM, Rumbaugh J, Bockenstedt LK, Falck-Ytter YT, Aguero-Rosenfeld ME, Auwaerter PG, et al. (January 2021). "Clinical Practice Guidelines by the Infectious Diseases Society of America (IDSA), American Academy of Neurology (AAN), and American College of Rheumatology (ACR): 2020 Guidelines for the Prevention, Diagnosis and Treatment of Lyme Disease". Clinical Infectious Diseases. 72 (1): e1–e48. doi:10.1093/cid/ciaa1215. PMID 33417672.
  53. ^ Mullegger RR (2004). "Dermatological manifestations of Lyme borreliosis". European Journal of Dermatology. 14 (5): 296–309. PMID 15358567.
  54. ^ Samuels DS, Radolf JD, eds. (2010). "Chapter 6, Structure, Function and Biogenesis of the Borrelia Cell Envelope". Borrelia: Molecular Biology, Host Interaction and Pathogenesis. Caister Academic Press. ISBN 978-1-904455-58-5.
  55. ^ Radolf JD, Samuels DS, eds. (2021). Lyme Disease and Relapsing Fever Spirochetes: Genomics, Molecular Biology, Host Interactions, and Disease Pathogenesis. Caister Academic Press. ISBN 978-1-913652-61-6.
  56. ^ Cutler SJ, Ruzic-Sabljic E, Potkonjak A (February 2017). "Emerging borreliae - Expanding beyond Lyme borreliosis" (PDF). Molecular and Cellular Probes. 31: 22–27. doi:10.1016/j.mcp.2016.08.003. PMID 27523487.
  57. ^ Stanek G, Reiter M (April 2011). "The expanding Lyme Borrelia complex--clinical significance of genomic species?". Clinical Microbiology and Infection. 17 (4): 487–493. doi:10.1111/j.1469-0691.2011.03492.x. PMID 21414082.
  58. ^ Schneider BS, Schriefer ME, Dietrich G, Dolan MC, Morshed MG, Zeidner NS (October 2008). "Borrelia bissettii isolates induce pathology in a murine model of disease". Vector Borne and Zoonotic Diseases. 8 (5): 623–633. doi:10.1089/vbz.2007.0251. PMID 18454594. from the original on 29 January 2020. Retrieved 29 January 2020.
  59. ^ Rudenko N, Golovchenko M, Mokrácek A, Piskunová N, Ruzek D, Mallatová N, Grubhoffer L (October 2008). "Detection of Borrelia bissettii in cardiac valve tissue of a patient with endocarditis and aortic valve stenosis in the Czech Republic". Journal of Clinical Microbiology. 46 (10): 3540–3543. doi:10.1128/JCM.01032-08. PMC 2566110. PMID 18650352.
  60. ^ "UpToDate". www.uptodate.com. Retrieved 10 July 2021.
  61. ^ a b Tilly K, Rosa PA, Stewart PE (June 2008). "Biology of infection with Borrelia burgdorferi". Infectious Disease Clinics of North America. 22 (2): 217–34, v. doi:10.1016/j.idc.2007.12.013. PMC 2440571. PMID 18452798.
  62. ^ a b c d Ryan KJ, Ray CG, eds. (2004). Sherris Medical Microbiology (4th ed.). McGraw Hill. pp. 434–37. ISBN 978-0-8385-8529-0.
  63. ^ Smith-Fiola D, Hamilton GC (March 2005). (PDF). Rutgers University, Rutgers Cooperative Research & Extension, New Jersey Agricultural Experiment Station. Archived from the original (PDF) on 26 June 2013.
  64. ^ Scribner H (31 May 2019). . Deseret News. Utah. Archived from the original on 25 January 2020. The U.S. CDC published a picture of a poppy seed muffin in which some of the "poppy seeds" were actually ticks
  65. ^ a b Lo Re V, Occi JL, MacGregor RR (April 2004). "Identifying the vector of Lyme disease". American Family Physician. 69 (8): 1935–1937. PMID 15117014.
  66. ^ . 2004. Archived from the original on 29 September 2013. Retrieved 26 September 2013.
  67. ^ a b "Lyme Disease Data and surveillance". Lyme Disease. Centers for Disease Control and Prevention. 5 February 2019. from the original on 13 April 2019. Retrieved 12 April 2019.
  68. ^ Hovius JW, van Dam AP, Fikrig E (September 2007). "Tick-host-pathogen interactions in Lyme borreliosis" (PDF). Trends in Parasitology. 23 (9): 434–438. doi:10.1016/j.pt.2007.07.001. PMID 17656156. (PDF) from the original on 16 February 2019. Retrieved 9 September 2019.
  69. ^ a b c Steere AC, Coburn J, Glickstein L (April 2004). "The emergence of Lyme disease". The Journal of Clinical Investigation. 113 (8): 1093–1101. doi:10.1172/JCI21681. PMC 385417. PMID 15085185.
  70. ^ Hu L (13 May 2019). "Patient education: What to do after a tick bite to prevent Lyme disease (Beyond the Basics)". www.uptodate.com. UpToDate. from the original on 1 January 2020. Retrieved 30 January 2020.
  71. ^ de Mik EL, van Pelt W, Docters-van Leeuwen BD, van der Veen A, Schellekens JF, Borgdorff MW (April 1997). "The geographical distribution of tick bites and erythema migrans in general practice in The Netherlands". International Journal of Epidemiology. 26 (2): 451–457. doi:10.1093/ije/26.2.451. PMID 9169184.
  72. ^ Sun Y, Xu R (2003). "Ability of Ixodes persulcatus, Haemaphysalis concinna and Dermacentor silvarum ticks to acquire and transstadially transmit Borrelia garinii". Experimental & Applied Acarology. 31 (1–2): 151–160. doi:10.1023/B:APPA.0000005119.30172.43. PMID 14756409. S2CID 19214181.
  73. ^ Ledin KE, Zeidner NS, Ribeiro JM, Biggerstaff BJ, Dolan MC, Dietrich G, et al. (March 2005). "Borreliacidal activity of saliva of the tick Amblyomma americanum". Medical and Veterinary Entomology. 19 (1): 90–95. doi:10.1111/j.0269-283X.2005.00546.x. PMID 15752182. S2CID 270178.
  74. ^ Masters EJ, Grigery CN, Masters RW (June 2008). "STARI, or Masters disease: Lone Star tick-vectored Lyme-like illness". Infectious Disease Clinics of North America. 22 (2): 361–76, viii. doi:10.1016/j.idc.2007.12.010. PMID 18452807.
  75. ^ Clark K (November 2004). "Borrelia species in host-seeking ticks and small mammals in northern Florida". Journal of Clinical Microbiology. 42 (11): 5076–5086. doi:10.1128/JCM.42.11.5076-5086.2004. PMC 525154. PMID 15528699.
  76. ^ Eisen L, Eisen RJ, Lane RS (December 2004). "The roles of birds, lizards, and rodents as hosts for the western black-legged tick Ixodes pacificus". Journal of Vector Ecology. 29 (2): 295–308. PMID 15709249.
  77. ^ a b Lane RS, Mun J, Eisen L, Eisen RJ (August 2006). "Refractoriness of the western fence lizard (Sceloporus occidentalis) to the Lyme disease group spirochete Borrelia bissettii". The Journal of Parasitology. 92 (4): 691–696. doi:10.1645/GE-738R1.1. PMID 16995383. S2CID 24200639.
  78. ^ Walsh CA, Mayer EW, Baxi LV (January 2007). "Lyme disease in pregnancy: case report and review of the literature". Obstetrical & Gynecological Survey. 62 (1): 41–50. doi:10.1097/01.ogx.0000251024.43400.9a. PMID 17176487. S2CID 31929160.
  79. ^ Dotters-Katz SK, Kuller J, Heine RP (September 2013). "Arthropod-borne bacterial diseases in pregnancy". Obstetrical & Gynecological Survey. 68 (9): 635–649. doi:10.1097/OGX.0b013e3182a5ed46. PMID 25102120. S2CID 26801402.
  80. ^ Lakos A, Solymosi N (June 2010). "Maternal Lyme borreliosis and pregnancy outcome". International Journal of Infectious Diseases. 14 (6): e494–e498. doi:10.1016/j.ijid.2009.07.019. PMID 19926325.
  81. ^ Steere AC (August 1989). "Lyme disease". The New England Journal of Medicine. 321 (9): 586–596. doi:10.1056/NEJM198908313210906. PMID 2668764.
  82. ^ "Tick Attachment and Tickborne Diseases". www.maine.gov. from the original on 1 October 2018. Retrieved 1 October 2018.
  83. ^ a b c "CDC Lyme disease FAQ". Centers for Disease Control and Prevention. 8 January 2021. Retrieved 11 January 2023.
  84. ^ Swanson SJ, Neitzel D, Reed KD, Belongia EA (October 2006). "Coinfections acquired from ixodes ticks". Clinical Microbiology Reviews. 19 (4): 708–727. doi:10.1128/CMR.00011-06. PMC 1592693. PMID 17041141.
  85. ^ Lindgren E, Gustafson R (July 2001). "Tick-borne encephalitis in Sweden and climate change". Lancet. 358 (9275): 16–18. doi:10.1016/S0140-6736(00)05250-8. PMID 11454371. S2CID 336323.
  86. ^ Pachner AR, Steiner I (June 2007). "Lyme neuroborreliosis: infection, immunity, and inflammation". The Lancet. Neurology. 6 (6): 544–552. doi:10.1016/S1474-4422(07)70128-X. PMID 17509489. S2CID 11600373.
  87. ^ "Does Borrelia burgdorferi produce a neurotoxin ?". American Lyme Disease Foundation. Retrieved 30 July 2022.
  88. ^ Auwaerter PG, Aucott J, Dumler JS (January 2004). "Lyme borreliosis (Lyme disease): molecular and cellular pathobiology and prospects for prevention, diagnosis and treatment". Expert Reviews in Molecular Medicine. 6 (2): 1–22. doi:10.1017/S1462399404007276. PMID 14987414. S2CID 24311718.
  89. ^ Fikrig E, Narasimhan S (April 2006). "Borrelia burgdorferi--traveling incognito?". Microbes and Infection. 8 (5): 1390–1399. doi:10.1016/j.micinf.2005.12.022. PMID 16698304.
  90. ^ Xu Q, Seemanapalli SV, Reif KE, Brown CR, Liang FT (April 2007). "Increasing the recruitment of neutrophils to the site of infection dramatically attenuates Borrelia burgdorferi infectivity". Journal of Immunology. 178 (8): 5109–5115. doi:10.4049/jimmunol.178.8.5109. PMID 17404293.
  91. ^ Coleman JL, Gebbia JA, Piesman J, Degen JL, Bugge TH, Benach JL (June 1997). "Plasminogen is required for efficient dissemination of B. burgdorferi in ticks and for enhancement of spirochetemia in mice". Cell. 89 (7): 1111–1119. doi:10.1016/S0092-8674(00)80298-6. PMID 9215633. S2CID 18750257.
  92. ^ Steere AC (July 2001). "Lyme disease". The New England Journal of Medicine. 345 (2): 115–125. doi:10.1056/NEJM200107123450207. PMID 11450660.
  93. ^ Rupprecht TA, Koedel U, Fingerle V, Pfister HW (2008). "The pathogenesis of lyme neuroborreliosis: from infection to inflammation". Molecular Medicine. 14 (3–4): 205–212. doi:10.2119/2007-00091.Rupprecht. PMC 2148032. PMID 18097481. from the original on 3 April 2015.
  94. ^ Cabello FC, Godfrey HP, Newman SA (August 2007). "Hidden in plain sight: Borrelia burgdorferi and the extracellular matrix". Trends in Microbiology. 15 (8): 350–354. doi:10.1016/j.tim.2007.06.003. PMID 17600717. from the original on 7 July 2019. Retrieved 5 July 2019.
  95. ^ Ramesh G, Alvarez AL, Roberts ED, Dennis VA, Lasater BL, Alvarez X, Philipp MT (September 2003). "Pathogenesis of Lyme neuroborreliosis: Borrelia burgdorferi lipoproteins induce both proliferation and apoptosis in rhesus monkey astrocytes". European Journal of Immunology. 33 (9): 2539–2550. doi:10.1002/eji.200323872. PMID 12938230. S2CID 25448372.
  96. ^ Halperin JJ, Heyes MP (January 1992). "Neuroactive kynurenines in Lyme borreliosis". Neurology. 42 (1): 43–50. doi:10.1212/WNL.42.1.43. PMID 1531156. S2CID 37495839.
  97. ^ Fallon BA, Keilp J, Prohovnik I, Heertum RV, Mann JJ (2003). "Regional cerebral blood flow and cognitive deficits in chronic lyme disease". The Journal of Neuropsychiatry and Clinical Neurosciences. 15 (3): 326–332. doi:10.1176/appi.neuropsych.15.3.326. PMID 12928508.
  98. ^ Gasse T, Murr C, Meyersbach P, Schmutzhard E, Wachter H, Fuchs D (September 1994). "Neopterin production and tryptophan degradation in acute Lyme neuroborreliosis versus late Lyme encephalopathy". European Journal of Clinical Chemistry and Clinical Biochemistry (Submitted manuscript). 32 (9): 685–689. doi:10.1515/cclm.1994.32.9.685. PMID 7865624. S2CID 22214184.
  99. ^ Zajkowska J, Grygorczuk S, Kondrusik M, Pancewicz S, Hermanowska-Szpakowicz T (2006). "[New aspects of pathogenesis of Lyme borreliosis]". Przeglad Epidemiologiczny (in Polish). 60 (Suppl 1): 167–170. PMID 16909797.
  100. ^ Ercolini AM, Miller SD (January 2009). "The role of infections in autoimmune disease". Clinical and Experimental Immunology. 155 (1): 1–15. doi:10.1111/j.1365-2249.2008.03834.x. PMC 2665673. PMID 19076824.
  101. ^ Singh SK, Girschick HJ (July 2004). "Lyme borreliosis: from infection to autoimmunity". Clinical Microbiology and Infection. 10 (7): 598–614. doi:10.1111/j.1469-0691.2004.00895.x. PMID 15214872.
  102. ^ Oldstone MB (October 1998). "Molecular mimicry and immune-mediated diseases". FASEB Journal. 12 (13): 1255–1265. doi:10.1096/fasebj.12.13.1255. PMC 7164021. PMID 9761770. S2CID 16089636.
  103. ^ Raveche ES, Schutzer SE, Fernandes H, Bateman H, McCarthy BA, Nickell SP, Cunningham MW (February 2005). "Evidence of Borrelia autoimmunity-induced component of Lyme carditis and arthritis". Journal of Clinical Microbiology. 43 (2): 850–856. doi:10.1128/JCM.43.2.850-856.2005. PMC 548028. PMID 15695691.
  104. ^ Weinstein A, Britchkov M (July 2002). "Lyme arthritis and post-Lyme disease syndrome". Current Opinion in Rheumatology. 14 (4): 383–387. doi:10.1097/00002281-200207000-00008. PMID 12118171.
  105. ^ Bolz DD, Weis JJ (August 2004). "Molecular mimicry to Borrelia burgdorferi: pathway to autoimmunity?". Autoimmunity. 37 (5): 387–392. doi:10.1080/08916930410001713098. PMID 15621562. S2CID 43045224.
  106. ^ Brown SL, Hansen SL, Langone JJ (July 1999). "Role of serology in the diagnosis of Lyme disease". JAMA. 282 (1): 62–66. doi:10.1001/jama.282.1.62. PMID 10404913.
  107. ^ Hofmann H (1996). "Lyme borreliosis--problems of serological diagnosis". Infection. 24 (6): 470–472. doi:10.1007/BF01713052. PMID 9007597. S2CID 19701481.
  108. ^ Wormser GP, Masters E, Nowakowski J, McKenna D, Holmgren D, Ma K, et al. (October 2005). "Prospective clinical evaluation of patients from Missouri and New York with erythema migrans-like skin lesions". Clinical Infectious Diseases. 41 (7): 958–965. doi:10.1086/432935. PMID 16142659.
  109. ^ a b "Lyme Disease risk areas map". Risk of Lyme disease to Canadians. Government of Canada. 27 January 2015. from the original on 10 May 2019. Retrieved 8 May 2019.
  110. ^ a b c d e f Aucott J, Morrison C, Munoz B, Rowe PC, Schwarzwalder A, West SK (June 2009). "Diagnostic challenges of early Lyme disease: lessons from a community case series". BMC Infectious Diseases. 9 (79): 79. doi:10.1186/1471-2334-9-79. PMC 2698836. PMID 19486523.
  111. ^ "Lyme disease diagnosis". Centers for Disease Control and Prevention (CDC). 7 October 2008. from the original on 28 August 2009. Retrieved 6 July 2009.
  112. ^ a b "Lyme disease". NICE guideline [NG95]. National Institute for Health and Care Excellence. 11 April 2018. from the original on 11 May 2019. Retrieved 24 May 2019.
  113. ^ Wilske B (2005). "Epidemiology and diagnosis of Lyme borreliosis". Annals of Medicine. 37 (8): 568–579. doi:10.1080/07853890500431934. PMID 16338759. S2CID 30818459.
  114. ^ a b c DePietropaolo DL, Powers JH, Gill JM, Foy AJ (July 2005). "Diagnosis of lyme disease". American Family Physician. 72 (2): 297–304. doi:10.1093/cid/cir464. PMID 16050454.
  115. ^ "Are serological tests of any value in the diagnosis of Lyme disease?". American Lyme Disease Foundation. from the original on 19 December 2019. Retrieved 2 December 2019.
  116. ^ Coyle PK, Schutzer SE, Deng Z, Krupp LB, Belman AL, Benach JL, Luft BJ (November 1995). "Detection of Borrelia burgdorferi-specific antigen in antibody-negative cerebrospinal fluid in neurologic Lyme disease". Neurology. 45 (11): 2010–2015. doi:10.1212/WNL.45.11.2010. PMID 7501150. S2CID 23797801.
  117. ^ a b c d Halperin JJ, Shapiro ED, Logigian E, Belman AL, Dotevall L, Wormser GP, et al. (July 2007). "Practice parameter: treatment of nervous system Lyme disease (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology". Neurology. 69 (1): 91–102. doi:10.1212/01.wnl.0000265517.66976.28. PMID 17522387.
  118. ^ Molloy PJ, Persing DH, Berardi VP (August 2001). "False-positive results of PCR testing for Lyme disease". Clinical Infectious Diseases. 33 (3): 412–413. doi:10.1086/321911. PMID 11438915.
  119. ^ a b Aguero-Rosenfeld ME, Wang G, Schwartz I, Wormser GP (July 2005). "Diagnosis of lyme borreliosis". Clinical Microbiology Reviews. 18 (3): 484–509. doi:10.1128/CMR.18.3.484-509.2005. PMC 1195970. PMID 16020686.
  120. ^ Nocton JJ, Dressler F, Rutledge BJ, Rys PN, Persing DH, Steere AC (January 1994). "Detection of Borrelia burgdorferi DNA by polymerase chain reaction in synovial fluid from patients with Lyme arthritis". The New England Journal of Medicine. 330 (4): 229–234. doi:10.1056/NEJM199401273300401. PMID 8272083.
  121. ^ Hyde FW, Johnson RC, White TJ, Shelburne CE (January 1989). "Detection of antigens in urine of mice and humans infected with Borrelia burgdorferi, etiologic agent of Lyme disease". Journal of Clinical Microbiology. 27 (1): 58–61. doi:10.1128/JCM.27.1.58-61.1989. PMC 267232. PMID 2913036.
  122. ^ Shafagati N, Patanarut A, Luchini A, Lundberg L, Bailey C, Petricoin E, et al. (July 2014). "The use of Nanotrap particles for biodefense and emerging infectious disease diagnostics". Pathogens and Disease. 71 (2): 164–176. doi:10.1111/2049-632X.12136. PMC 7108521. PMID 24449537.
  123. ^ Shafagati N, Patanarut A, Luchini A, Lundberg L, Bailey C, Petricoin E, et al. (July 2014). "The use of Nanotrap particles for biodefense and emerging infectious disease diagnostics". Pathogens and Disease. 71 (2): 164–176. doi:10.1111/2049-632x.12136. PMC 7108521. PMID 24449537.
  124. ^ Burdash N, Fernandes J (June 1991). "Lyme borreliosis: detecting the great imitator". The Journal of the American Osteopathic Association. 91 (6): 573–4, 577–8. doi:10.1515/jom-1991-910610. PMID 1874654. S2CID 245118295.
  125. ^ Hildenbrand P, Craven DE, Jones R, Nemeskal P (June 2009). "Lyme neuroborreliosis: manifestations of a rapidly emerging zoonosis". AJNR. American Journal of Neuroradiology. 30 (6): 1079–1087. doi:10.3174/ajnr.A1579. PMC 7051319. PMID 19346313.
  126. ^ Westervelt HJ, McCaffrey RJ (September 2002). "Neuropsychological functioning in chronic Lyme disease". Neuropsychology Review. 12 (3): 153–177. doi:10.1023/A:1020381913563. PMID 12428915. S2CID 2807397.
  127. ^ Dworkin RH, Johnson RW, Breuer J, Gnann JW, Levin MJ, Backonja M, et al. (January 2007). "Recommendations for the management of herpes zoster". Clinical Infectious Diseases. 44 (Suppl 1): S1-26. doi:10.1086/510206. PMID 17143845. S2CID 10894629.
  128. ^ Halperin JJ (May 2015). "Chronic Lyme disease: misconceptions and challenges for patient management". Infection and Drug Resistance. 8: 119–128. doi:10.2147/IDR.S66739. PMC 4440423. PMID 26028977.
  129. ^ a b Tarulli AW, Raynor EM (May 2007). (PDF). Neurologic Clinics. 25 (2): 387–405. doi:10.1016/j.ncl.2007.01.008. PMID 17445735. S2CID 15518713. Archived from the original (PDF) on 20 February 2019.
  130. ^ Pachner AR (1989). "Neurologic manifestations of Lyme disease, the new "great imitator"". Reviews of Infectious Diseases. 11 (Suppl 6): S1482–S1486. doi:10.1093/clinids/11.Supplement_6.S1482. PMID 2682960. S2CID 3862308.
  131. ^ Branda JA, Linskey K, Kim YA, Steere AC, Ferraro MJ (September 2011). "Two-tiered antibody testing for Lyme disease with use of 2 enzyme immunoassays, a whole-cell sonicate enzyme immunoassay followed by a VlsE C6 peptide enzyme immunoassay". Clinical Infectious Diseases. 53 (6): 541–547. doi:10.1093/cid/cir464. PMID 21865190.
  132. ^ a b c d e f g h i j "Preventing tick bites on people". Lyme Disease. Centers for Disease Control and Prevention. 8 March 2019. from the original on 15 June 2019. Retrieved 21 May 2019.
  133. ^ a b "About ticks – preventative measures". Tick-borne diseases. Companion Vector-Borne Diseases. from the original on 7 June 2019. Retrieved 21 May 2019.
  134. ^ a b c d e f Eisen L, Eisen RJ (September 2016). "Critical Evaluation of the Linkage Between Tick-Based Risk Measures and the Occurrence of Lyme Disease Cases". Journal of Medical Entomology. 53 (5): 1050–1062. doi:10.1093/jme/tjw092. PMC 5777907. PMID 27330093.
  135. ^ "Host seeking". CVBD: Companion Vector-Borne Diseases. from the original on 16 October 2016. Retrieved 8 December 2016.
  136. ^ a b c "Tips to prevent tick bytes". Insect repellents. Environmental Protection Agency. 15 July 2013. from the original on 15 April 2019. Retrieved 21 May 2019.
  137. ^ a b c d e f g h "Protection against Mosquitoes, Ticks, & Other Arthropods". Travelers' Health. Centers for Disease Control and Prevention. from the original on 9 May 2019. Retrieved 21 May 2019.
  138. ^ a b c Miller NJ, Rainone EE, Dyer MC, González ML, Mather TN (March 2011). "Tick bite protection with permethrin-treated summer-weight clothing". Journal of Medical Entomology. 48 (2): 327–333. doi:10.1603/me10158. PMID 21485369.
  139. ^ a b Eisen L, Rose D, Prose R, Breuner NE, Dolan MC, Thompson K, Connally N (October 2017). "Bioassays to evaluate non-contact spatial repellency, contact irritancy, and acute toxicity of permethrin-treated clothing against nymphal Ixodes scapularis ticks". Ticks and Tick-Borne Diseases. 8 (6): 837–849. doi:10.1016/j.ttbdis.2017.06.010. PMC 5665650. PMID 28754599.
  140. ^ Dymond NL, Swift IM (June 2008). "Permethrin toxicity in cats: a retrospective study of 20 cases". Australian Veterinary Journal. 86 (6): 219–223. doi:10.1111/j.1751-0813.2008.00298.x. PMID 18498556.
  141. ^ a b "Repellents: protection against mosquitoes, ticks and other arthropods". Insect repellents. Environmental Protection Agency. 9 July 2013. from the original on 20 May 2019. Retrieved 21 May 2019.
  142. ^ a b c Nguyen QD, Vu MN, Hebert AA (January 2023). "Insect repellents: An updated review for the clinician". Journal of the American Academy of Dermatology. 88 (1): 123–130. doi:10.1016/j.jaad.2018.10.053. PMID 30395919. S2CID 53246686.
  143. ^ a b c d e Stafford KC (2007). "Tick management handbook (rev. ed.)" (PDF). Bulletins. Connecticut Agricultural Experiment Station. (PDF) from the original on 3 April 2019. Retrieved 21 May 2019.
  144. ^ Piesman J, Dolan MC (May 2002). "Protection against lyme disease spirochete transmission provided by prompt removal of nymphal Ixodes scapularis (Acari: Ixodidae)". Journal of Medical Entomology. 39 (3): 509–512. doi:10.1603/0022-2585-39.3.509. PMID 12061448. S2CID 39805040.
  145. ^ a b c d e f "Tick removal and testing". Lyme Disease. Centers for Disease Control and Prevention. 22 April 2019. from the original on 23 November 2017. Retrieved 21 May 2019.
  146. ^ a b "Tick Allergy". 2014. from the original on 14 May 2015. Retrieved 30 April 2015.
  147. ^ Röggla G (2002). "Death of unknown origin in alpine medicine". Wilderness & Environmental Medicine. 13 (2): 181. doi:10.1580/1080-6032(2002)013[0181:ltte]2.0.co;2. PMID 12092975.
  148. ^ Ghirga G, Ghirga P (September 2010). "Effective tick removal with a fishing line knot". Wilderness & Environmental Medicine. 21 (3): 270–271. doi:10.1016/j.wem.2010.04.005. PMID 20832708.
  149. ^ a b c d e "Preventing tick bites in the yard". Lyme Disease. Centers for Disease Control and Prevention. 8 March 2019. from the original on 15 June 2019. Retrieved 21 May 2019.
  150. ^ "CDC – Lyme Disease – NIOSH Workplace Safety and Health Topic". www.cdc.gov. 3 August 2017. from the original on 13 November 2015. Retrieved 3 November 2015.
  151. ^ "Risks: Lyme Disease | NIOSH | CDC". www.cdc.gov. 14 November 2018. from the original on 26 April 2019. Retrieved 26 April 2019.
  152. ^ "Lyme disease". The Centers for Disease Control and Prevention. 12 October 2016. from the original on 17 June 2017. Retrieved 22 June 2017.  This article incorporates public domain material from websites or documents of the Centers for Disease Control and Prevention.
  153. ^ a b c Chrobak U (3 February 2022). "Lyme and other tick-borne diseases are on the rise. But why?". Knowable Magazine. doi:10.1146/knowable-020222-1. Retrieved 4 March 2022.
  154. ^ Rand PW, Lubelczyk C, Holman MS, Lacombe EH, Smith RP (July 2004). "Abundance of Ixodes scapularis (Acari: Ixodidae) after the complete removal of deer from an isolated offshore island, endemic for Lyme Disease". Journal of Medical Entomology. 41 (4): 779–784. doi:10.1603/0022-2585-41.4.779. PMID 15311475. S2CID 5853201.
  155. ^ "Figure 2: Changes in deer density and cases of Lyme disease in Mumford Cove, Connecticut, 1996–2004 (CT DEP data)" (PDF). Managing Urban Deer in Connecticut (2nd ed.). Connecticut Department of Environmental Protection – Wildlife Division. June 2007. p. 4. (PDF) from the original on 2 June 2013.
  156. ^ Stafford KC (2004). Tick Management Handbook (PDF). Connecticut Agricultural Experiment Station and Connecticut Department of Public Health. p. 46. (PDF) from the original on 25 October 2007. Retrieved 21 August 2007.
  157. ^ Rochlin I, Toledo A (June 2020). "Emerging tick-borne pathogens of public health importance: a mini-review". Journal of Medical Microbiology. 69 (6): 781–791. doi:10.1099/jmm.0.001206. PMC 7451033. PMID 32478654.
  158. ^ Rajput ZI, Hu SH, Chen WJ, Arijo AG, Xiao CW (November 2006). "Importance of ticks and their chemical and immunological control in livestock". Journal of Zhejiang University. Science. B. 7 (11): 912–921. doi:10.1631/jzus.2006.B0912. PMC 1635821. PMID 17048307.
  159. ^ a b Gern L, Estrada-Peña A, Frandsen F, Gray JS, Jaenson TG, Jongejan F, et al. (March 1998). "European reservoir hosts of Borrelia burgdorferi sensu lato". Zentralblatt Fur Bakteriologie. 287 (3): 196–204. doi:10.1016/S0934-8840(98)80121-7. PMID 9580423.
  160. ^ a b c d Wodecka B, Rymaszewska A, Skotarczak B (April 2014). "Host and pathogen DNA identification in blood meals of nymphal Ixodes ricinus ticks from forest parks and rural forests of Poland". Experimental & Applied Acarology. 62 (4): 543–555. doi:10.1007/s10493-013-9763-x. PMC 3933768. PMID 24352572.
  161. ^ Jaenson TG, Tälleklint L (September 1992). "Incompetence of roe deer as reservoirs of the Lyme borreliosis spirochete". Journal of Medical Entomology. 29 (5): 813–817. doi:10.1093/jmedent/29.5.813. PMID 1404260.
  162. ^ Tälleklint L, Jaenson TG (November 1994). "Transmission of Borrelia burgdorferi s.l. from mammal reservoirs to the primary vector of Lyme borreliosis, Ixodes ricinus (Acari: Ixodidae), in Sweden". Journal of Medical Entomology. 31 (6): 880–886. doi:10.1093/jmedent/31.6.880. PMID 7815401.
  163. ^ Overzier E, Pfister K, Herb I, Mahling M, Böck G, Silaghi C (June 2013). "Detection of tick-borne pathogens in roe deer (Capreolus capreolus), in questing ticks (Ixodes ricinus), and in ticks infesting roe deer in southern Germany". Ticks and Tick-Borne Diseases. 4 (4): 320–328. doi:10.1016/j.ttbdis.2013.01.004. PMID 23571115.[permanent dead link]
  164. ^ a b O'Bier NS, Hatke AL, Camire AC, Marconi RT (2021). "Human and Veterinary Vaccines for Lyme Disease". Current Issues in Molecular Biology. 42: 191–222. doi:10.21775/cimb.042.191. PMC 7946718. PMID 33289681.
  165. ^ Poland GA, Jacobson RM (March 2001). "The prevention of Lyme disease with vaccine". Vaccine. 19 (17–19): 2303–2308. doi:10.1016/S0264-410X(00)00520-X. PMID 11257352.
  166. ^ a b Poland GA (February 2011). "Vaccines against Lyme disease: What happened and what lessons can we learn?". Clinical Infectious Diseases. 52 (Suppl 3): s253–s258. doi:10.1093/cid/ciq116. PMID 21217172.
  167. ^ Rowe C (13 June 1999). "Lukewarm Response To New Lyme Vaccine". The New York Times. from the original on 10 February 2012. Retrieved 11 July 2008.
  168. ^ a b c Abbott A (February 2006). "Lyme disease: uphill struggle". Nature. 439 (7076): 524–525. doi:10.1038/439524a. PMID 16452949. S2CID 4315588.
  169. ^ "Sole Lyme Vaccine Is Pulled Off Market". The New York Times. 28 February 2002. from the original on 30 August 2010. Retrieved 11 July 2008.
  170. ^ a b Nigrovic LE, Thompson KM (January 2007). "The Lyme vaccine: a cautionary tale". Epidemiology and Infection. 135 (1): 1–8. doi:10.1017/S0950268806007096. PMC 2870557. PMID 16893489.
  171. ^ "When a vaccine is safe". Nature. 439 (7076): 509. February 2006. Bibcode:2006Natur.439Q.509.. doi:10.1038/439509a. PMID 16452935.
  172. ^ Aronowitz RA (June 2012). "The rise and fall of the lyme disease vaccines: a cautionary tale for risk interventions in American medicine and public health". The Milbank Quarterly. 90 (2): 250–277. doi:10.1111/j.1468-0009.2012.00663.x. PMC 3460208. PMID 22709388.
  173. ^ "Anti-vaxxers stopped the last Lyme disease vaccine. The FDA has just fast-tracked a new one". Newsweek. 25 July 2017. from the original on 25 December 2017. Retrieved 25 December 2017.
  174. ^ Brooks D (6 August 2018). "A Lyme vaccine for humans is getting closer, says French biotech firm". Concord Monitor. Retrieved 20 July 2021.
  175. ^ Taylor NP (4 February 2022). "Pfizer's $130M Lyme disease vaccine advances to phase 3 after 3rd shot shown to increase antibodies". Fierce Biotech. Retrieved 5 June 2022.
  176. ^ Neergaard L, Lum S (8 August 2022). "Major test of first possible Lyme vaccine in 20 years begins". Associated Press. Retrieved 11 August 2022.
  177. ^ a b "An Efficacy, Safety, Tolerability, Immunogenicity, and Lot-Consistency Clinical Trial of a 6-Valent OspA-Based Lyme Disease Vaccine (VLA15) (VALOR)". ClinicalTrials.gov. National Institute of Health. 28 July 2022. Retrieved 11 August 2022.
  178. ^ Klein A (17 November 2021). "An mRNA vaccine that causes a red, itchy skin rash in response to bites by ticks may allow them to be removed before they transmit Lyme disease-causing bacteria". New Scientist.
  179. ^ Hathaway B. "Novel Lyme vaccine shows promise". Yale University. Retrieved 13 December 2021. Compared to non-immunized guinea pigs, vaccinated animals exposed to infected ticks quickly developed redness at the tick bite site. None of the immunized animals developed Lyme disease if ticks were removed when redness developed. In contrast, about half of the control group became infected with B. burgdorferi after tick removal. When a single infected tick was attached to immunized guinea pigs and not removed, none of vaccinated animals were infected compared to 60 percent of control animals. However, protection waned in immunized guinea pigs if three ticks remained attached to the animal. Ticks in immunized animals were unable to feed aggressively and dislodged more quickly than those on guinea pigs in the control group.
  180. ^ Sajid A, Matias J, Arora G, Kurokawa C, DePonte K, Tang X, et al. (November 2021). "mRNA vaccination induces tick resistance and prevents transmission of the Lyme disease agent". Science Translational Medicine. 13 (620): eabj9827. doi:10.1126/scitranslmed.abj9827. PMID 34788080. S2CID 244375227.
  181. ^ a b Brooks WC. "Lyme Disease". Veterinary Information Network. from the original on 4 April 2015. Retrieved 10 February 2012.
  182. ^ Pace EJ, O'Reilly M (May 2020). "Tickborne Diseases: Diagnosis and Management". American Family Physician. 101 (9): 530–540. PMID 32352736.
  183. ^ Berende A, ter Hofstede HJ, Vos FJ, van Middendorp H, Vogelaar ML, Tromp M, et al. (March 2016). "Randomized Trial of Longer-Term Therapy for Symptoms Attributed to Lyme Disease". The New England Journal of Medicine. 374 (13): 1209–1220. doi:10.1056/NEJMoa1505425. PMID 27028911.
  184. ^ Stephenson M (4 October 2012). "OTC Drops: Telling the Tears Apart". Review of Ophtalmology. Jobson Medical Information LLC. from the original on 17 April 2019. Retrieved 16 April 2019.
  185. ^ a b c d e Arvikar SL, Steere AC (June 2015). "Diagnosis and treatment of Lyme arthritis". Infectious Disease Clinics of North America. 29 (2): 269–280. doi:10.1016/j.idc.2015.02.004. PMC 4443866. PMID 25999223.
  186. ^ a b c Wormser GP, Ramanathan R, Nowakowski J, McKenna D, Holmgren D, Visintainer P, et al. (May 2003). "Duration of antibiotic therapy for early Lyme disease. A randomized, double-blind, placebo-controlled trial". Annals of Internal Medicine. 138 (9): 697–704. doi:10.7326/0003-4819-138-9-200305060-00005. PMID 12729423. S2CID 3083800.
  187. ^ Stupica D, Lusa L, Ruzić-Sabljić E, Cerar T, Strle F (August 2012). "Treatment of erythema migrans with doxycycline for 10 days versus 15 days". Clinical Infectious Diseases. 55 (3): 343–350. doi:10.1093/cid/cis402. PMID 22523260.
  188. ^ a b Dattwyler RJ, Luft BJ, Kunkel MJ, Finkel MF, Wormser GP, Rush TJ, et al. (July 1997). "Ceftriaxone compared with doxycycline for the treatment of acute disseminated Lyme disease". The New England Journal of Medicine. 337 (5): 289–294. doi:10.1056/NEJM199707313370501. PMID 9233865.
  189. ^ Vrethem M, Hellblom L, Widlund M, Ahl M, Danielsson O, Ernerudh J, Forsberg P (October 2002). "Chronic symptoms are common in patients with neuroborreliosis -- a questionnaire follow-up study". Acta Neurologica Scandinavica. 106 (4): 205–208. doi:10.1034/j.1600-0404.2002.01358.x. PMID 12225315. S2CID 42290158.
  190. ^ Berglund J, Stjernberg L, Ornstein K, Tykesson-Joelsson K, Walter H (2002). "5-y Follow-up study of patients with neuroborreliosis". Scandinavian Journal of Infectious Diseases. 34 (6): 421–425. doi:10.1080/00365540110080421. PMID 12160168. S2CID 28306612.
  191. ^ a b c Clark JR, Carlson RD, Sasaki CT, Pachner AR, Steere AC (November 1985). "Facial paralysis in Lyme disease". The Laryngoscope. 95 (11): 1341–1345. doi:10.1288/00005537-198511000-00009. PMID 4058212. S2CID 85021.
  192. ^ a b c Dotevall L, Hagberg L (March 1999). "Successful oral doxycycline treatment of Lyme disease-associated facial palsy and meningitis". Clinical Infectious Diseases. 28 (3): 569–574. doi:10.1086/515145. PMID 10194080.
  193. ^ Kalish RA, Kaplan RF, Taylor E, Jones-Woodward L, Workman K, Steere AC (February 2001). "Evaluation of study patients with Lyme disease, 10-20-year follow-up". The Journal of Infectious Diseases. 183 (3): 453–460. doi:10.1086/318082. PMID 11133377.
  194. ^ a b c Jowett N, Gaudin RA, Banks CA, Hadlock TA (June 2017). "Steroid use in Lyme disease-associated facial palsy is associated with worse long-term outcomes". The Laryngoscope. 127 (6): 1451–1458. doi:10.1002/lary.26273. PMID 27598389. S2CID 25596860.
lyme, disease, confused, with, phytophotodermatitis, also, known, lime, disease, also, known, lyme, borreliosis, vector, borne, disease, caused, borrelia, bacteria, which, spread, ticks, genus, ixodes, most, common, sign, infection, expanding, rash, known, ery. Not to be confused with phytophotodermatitis also known as lime disease Lyme disease also known as Lyme borreliosis is a vector borne disease caused by Borrelia bacteria which are spread by ticks in the genus Ixodes 3 8 9 The most common sign of infection is an expanding red rash known as erythema migrans EM which appears at the site of the tick bite about a week afterwards 1 The rash is typically neither itchy nor painful 1 Approximately 70 80 of infected people develop a rash 1 Early diagnosis can be difficult 10 Other early symptoms may include fever headaches and tiredness 1 If untreated symptoms may include loss of the ability to move one or both sides of the face joint pains severe headaches with neck stiffness or heart palpitations 1 Months to years later repeated episodes of joint pain and swelling may occur 1 Occasionally shooting pains or tingling in the arms and legs may develop 1 Despite appropriate treatment about 10 to 20 of those affected develop joint pains memory problems and tiredness for at least six months 1 11 Lyme diseaseOther namesLyme borreliosisAn adult deer tick most cases of Lyme are caused by nymphal rather than adult ticks SpecialtyInfectious diseaseSymptomsExpanding area of redness at the site of a tick bite fever headache tiredness 1 ComplicationsFacial nerve paralysis arthritis meningitis 1 Heart rhythm irregularities 2 Usual onsetA week after a bite 1 CausesBorrelia spread by ticks 3 Diagnostic methodBased on symptoms tick exposure blood tests 4 PreventionPrevention of tick bites clothing the limbs DEET doxycycline 3 MedicationDoxycycline amoxicillin ceftriaxone cefuroxime 3 Frequency 476k year in U S a likely overestimate 200k year in Europe 5 6 7 Lyme disease is transmitted to humans by the bites of infected ticks of the genus Ixodes 12 In the United States ticks of concern are usually of the Ixodes scapularis type In most cases a tick must be attached for 36 to 48 hours or more before the Lyme disease bacterium can be transmitted If you remove a tick quickly within 24 hours you can greatly reduce your chances of getting Lyme disease 13 14 In Europe Ixodes ricinus ticks may spread the bacteria more quickly 14 15 In North America the bacterial species Borrelia burgdorferiand B mayonii cause Lyme disease 3 16 In Europe and Asia Borrelia afzelii Borrelia garinii B spielmanii and four other species also cause the disease 3 The disease does not appear to be transmissible between people by other animals nor through food 13 Diagnosis is based on a combination of symptoms history of tick exposure and possibly testing for specific antibodies in the blood 4 17 Blood tests are often falsely negative in the early stages of the disease 3 Testing of individual ticks is not typically useful 18 Prevention includes efforts to prevent tick bites by wearing clothing to cover the arms and legs and using DEET or picaridin based insect repellents 3 8 Using pesticides to reduce tick numbers may also be effective 3 Ticks can be removed using tweezers 19 If the removed tick is full of blood a single dose of doxycycline may be used to prevent the development of infection but is not generally recommended since the development of infection is rare 3 If an infection develops a number of antibiotics are effective including doxycycline amoxicillin and cefuroxime 3 Standard treatment usually lasts for two or three weeks 3 Some people develop a fever and muscle and joint pains from treatment which may last for one or two days 3 In those who develop persistent symptoms long term antibiotic therapy has not been found to be useful 3 20 Lyme disease is the most common disease spread by ticks in the Northern Hemisphere 21 7 Infections are most common in the spring and early summer 3 Lyme disease was diagnosed as a separate condition for the first time in 1975 in Lyme Connecticut It was originally mistaken for juvenile rheumatoid arthritis 22 The bacterium involved was first described in 1981 by Willy Burgdorfer 23 Chronic symptoms following treatment are known as post treatment Lyme disease syndrome PTLDS 20 PTLDS is different from chronic Lyme disease a term no longer supported by scientists and used in different ways by different groups 20 24 Some healthcare providers claim that PTLDS is caused by persistent infection but this is not believed to be true because no evidence of persistent infection can be found after standard treatment 25 As of 2023 update clinical trials of proposed human vaccines for Lyme disease were being carried out but no vaccine was available A vaccine LYMERix was produced but discontinued in 2002 due to insufficient demand 26 There are several vaccines for the prevention of Lyme disease in dogs Contents 1 Signs and symptoms 1 1 Early localized infection 1 2 Early disseminated infection 1 3 Late disseminated infection 2 Cause 2 1 Tick life cycle 2 2 Transmission 2 3 Tick borne co infections 3 Pathophysiology 3 1 Immunological studies 4 Diagnosis 4 1 Laboratory testing 4 2 Imaging 4 3 Differential diagnosis 5 Prevention 5 1 Tick removal 5 2 Preventive antibiotics 5 3 Garden landscaping 5 4 Occupational exposure 5 5 Host animals 6 Vaccination 6 1 LYMErix 6 2 VLA15 6 3 Other research 6 4 Canine vaccines 7 Treatment 8 Prognosis 8 1 Post treatment Lyme disease syndrome 9 Epidemiology 9 1 Africa 9 2 Asia 9 3 Australia 9 4 Europe 9 4 1 United Kingdom 9 5 North America 9 5 1 Canada 9 5 2 Mexico 9 5 3 United States 9 6 South America 10 Etymology 11 History 12 Society culture amp controversy 13 Other animals 13 1 Dogs 13 2 Cats 13 3 Horses 14 References 15 Further reading 16 External linksSigns and symptoms edit nbsp An expanding rash is an initial sign of about 80 of Lyme infections The rash may look like a bull s eye as pictured in about 80 of cases in Europe and 20 of cases in the US 27 28 29 30 Lyme disease can affect several body systems and produce a broad range of symptoms Not everyone with Lyme disease has all of the symptoms and many of the symptoms are not specific to Lyme disease but can occur with other diseases as well 31 The incubation period from infection to the onset of symptoms is usually one to two weeks but can be much shorter days or much longer months to years 32 Lyme symptoms most often occur from May to September in the Northern Hemisphere because the nymphal stage of the tick is responsible for most cases 32 Asymptomatic infection exists but occurs in less than 7 of infected individuals in the United States 33 Asymptomatic infection may be much more common among those infected in Europe 34 Early localized infection edit Early localized infection can occur when the infection has not yet spread throughout the body Only the site where the infection has first come into contact with the skin is affected The initial sign of about 80 of Lyme infections is an erythema migrans EM rash at the site of a tick bite often near skin folds such as the armpit groin or back of the knee on the trunk under clothing straps or in children s hair ears or neck 27 3 Most people who get infected do not remember seeing a tick or a bite The rash appears typically one or two weeks range 3 32 days after the bite and expands 2 3 cm per day up to a diameter of 5 70 cm median 16 cm 27 3 28 The rash is usually circular or oval red or bluish and may have an elevated or darker center 3 29 30 In about 79 of cases in Europe but only 19 of cases in endemic areas of the U S the rash gradually clears from the center toward the edges possibly forming a bull s eye pattern 28 29 30 The rash may feel warm but usually is not itchy is rarely tender or painful and takes up to four weeks to resolve if untreated 3 The EM rash is often accompanied by symptoms of a viral like illness including fatigue headache body aches fever and chills but usually not nausea or upper respiratory problems These symptoms may also appear without a rash or linger after the rash has disappeared Lyme can progress to later stages without these symptoms or a rash 3 People with high fever for more than two days or whose other symptoms of viral like illness do not improve despite antibiotic treatment for Lyme disease or who have abnormally low levels of white or red cells or platelets in the blood should be investigated for possible coinfection with other tick borne diseases such as ehrlichiosis and babesiosis 35 Early disseminated infection edit Within days to weeks after the onset of local infection the Borrelia bacteria may spread through the lymphatic system or bloodstream In 10 20 of untreated cases EM rashes develop at sites across the body that bear no relation to the original tick bite 27 Transient muscle pains and joint pains are also common 27 In about 10 15 of untreated people Lyme causes neurological problems known as neuroborreliosis 36 Early neuroborreliosis typically appears 4 6 weeks range 1 12 weeks after the tick bite and involves some combination of lymphocytic meningitis cranial neuritis radiculopathy and or mononeuritis multiplex 35 37 Lymphocytic meningitis causes characteristic changes in the cerebrospinal fluid CSF and may be accompanied for several weeks by variable headache and less commonly usually mild meningitis signs such as inability to flex the neck fully and intolerance to bright lights but typically no or only very low fever 38 After several months neuroborreliosis can also present otolaryngological symptoms Up to 76 5 of them present as tinnitus the most common symptom Vertigo and dizziness 53 7 and hearing loss 16 7 were the next most common symptoms 39 In children partial loss of vision may also occur 35 Cranial neuritis is an inflammation of cranial nerves When due to Lyme it most typically causes facial palsy impairing blinking smiling and chewing on one or both sides of the face It may also cause intermittent double vision 35 38 Lyme radiculopathy is an inflammation of spinal nerve roots that often causes pain and less often weakness numbness or altered sensation in the areas of the body served by nerves connected to the affected roots e g limb s or part s of trunk The pain is often described as unlike any other previously felt excruciating migrating worse at night rarely symmetrical and often accompanied by extreme sleep disturbance 37 40 Mononeuritis multiplex is an inflammation causing similar symptoms in one or more unrelated peripheral nerves 36 35 Rarely early neuroborreliosis may involve inflammation of the brain or spinal cord with symptoms such as confusion abnormal gait ocular movements or speech impaired movement impaired motor planning or shaking 35 37 In North America facial palsy is the typical early neuroborreliosis presentation occurring in 5 10 of untreated people in about 75 of cases accompanied by lymphocytic meningitis 35 41 Lyme radiculopathy is reported half as frequently but many cases may be unrecognized 42 In European adults the most common presentation is a combination of lymphocytic meningitis and radiculopathy known as Bannwarth syndrome accompanied in 36 89 of cases by facial palsy 37 40 In this syndrome radicular pain tends to start in the same body region as the initial erythema migrans rash if there was one and precedes possible facial palsy and other impaired movement 40 In extreme cases permanent impairment of motor or sensory function of the lower limbs may occur 34 In European children the most common manifestations are facial palsy in 55 other cranial neuritis and lymphocytic meningitis in 27 37 In about 4 10 of untreated cases in the U S and 0 3 4 of untreated cases in Europe typically between June and December about one month range 4 days 7 months after the tick bite the infection may cause heart complications known as Lyme carditis 43 44 Symptoms may include heart palpitations in 69 of people dizziness fainting shortness of breath and chest pain 43 Other symptoms of Lyme disease may also be present such as EM rash joint aches facial palsy headaches or radicular pain 43 In some people however carditis may be the first manifestation of Lyme disease 43 Lyme carditis in 19 87 of people adversely impacts the heart s electrical conduction system causing atrioventricular block that often manifests as heart rhythms that alternate within minutes between abnormally slow and abnormally fast 43 44 In 10 15 of people Lyme causes myocardial complications such as cardiomegaly left ventricular dysfunction or congestive heart failure 43 Another skin condition found in Europe but not in North America is borrelial lymphocytoma a purplish lump that develops on the ear lobe nipple or scrotum 45 Late disseminated infection edit nbsp Lyme arthritis caused this 3 year old girl s knee to become swollen Though painless it did make her limp 46 Lyme arthritis occurs in up to 60 of untreated people typically starting about six months after infection 27 It usually affects only one or a few joints often a knee or possibly the hip other large joints or the temporomandibular joint 35 47 Usually large joint effusion and swelling occur but only mild or moderate pain 35 Without treatment swelling and pain typically resolve over time but periodically return 35 Baker s cysts may form and rupture In early US studies of Lyme disease a rare peripheral neuropathy was described that included numbness tingling or burning starting at the feet or hands and over time possibly moving up the limbs In a later analysis that discovered poor documentation of this manifestation experts wondered if it exists at all in the US or is merely very rare 35 48 A neurologic syndrome called Lyme encephalopathy is associated with subtle memory and cognitive difficulties insomnia a general sense of feeling unwell and changes in personality 49 Lyme encephalopathy is controversial in the US and has not been reported in Europe 7 Problems such as depression and fibromyalgia are as common in people with Lyme disease as in the general population 50 51 There is no compelling evidence that Lyme disease causes psychiatric disorders behavioral disorders e g ADHD or developmental disorders e g autism 52 Acrodermatitis chronica atrophicans is a chronic skin disorder observed primarily in Europe among the elderly 45 It begins as a reddish blue patch of discolored skin often on the backs of the hands or feet The lesion slowly atrophies over several weeks or months with the skin becoming first thin and wrinkled and then if untreated completely dry and hairless 53 It is also associated with peripheral neuropathy 48 Cause editMain article Lyme disease microbiology nbsp Borrelia bacteria the causative agents of Lyme disease magnifiedLyme disease is caused by spirochetes spiral bacteria from the genus Borrelia Spirochetes are surrounded by peptidoglycan and flagella along with an outer membrane similar to Gram negative bacteria Because of their double membrane envelope Borrelia bacteria are often mistakenly described as Gram negative despite the considerable differences in their envelope components from Gram negative bacteria 54 The Lyme related Borrelia species are collectively known as Borrelia burgdorferi sensu lato and show a great deal of genetic diversity 55 B burgdorferi sensu lato is a species complex made up of 20 accepted and three proposed genospecies Eight species are known to cause Lyme disease B mayonii found in North America B burgdorferi sensu stricto found in North America and Europe B afzelii B garinii B spielmanii and B lusitaniae all found in Eurasia 56 57 16 9 Some studies have also proposed that B valaisiana may sometimes infect humans but this species does not seem to be an important cause of disease 58 59 Further information Weather and climate effects on Lyme disease exposure Tick life cycle edit nbsp Deer tick life cycle nbsp Ixodes scapularis the primary vector of Lyme disease in eastern North AmericaThree stages occur in the life cycle of a tick larva nymph and adult During the nymph stage ticks most frequently transmit Lyme disease and are usually most active in late spring and early summer in regions where the climate is mild During the adult stage Lyme disease transmission is less common because adult ticks are less likely to bite humans and tend to be larger in size so can be easily seen and removed 60 Transmission edit Lyme disease is classified as a zoonosis as it is transmitted to humans from a natural reservoir among small mammals and birds by ticks that feed on both sets of hosts 61 Hard bodied ticks of the genus Ixodes are the vectors of Lyme disease also the vector for Babesia 62 Most infections are caused by ticks in the nymphal stage because they are very small thus may feed for long periods of time undetected 61 Nymphal ticks are generally the size of a poppy seed and sometimes with a dark head and a translucent body 63 Or the nymphal ticks can be darker 64 The younger larval ticks are very rarely infected 65 Although deer are the preferred hosts of adult deer ticks and tick populations are much lower in the absence of deer ticks generally do not acquire Borrelia from deer instead they obtain them from infected small mammals such as the white footed mouse and occasionally birds 66 Areas where Lyme is common are expanding 67 Within the tick midgut the Borrelia s outer surface protein A OspA binds to the tick receptor for OspA known as TROSPA When the tick feeds theBorrelia downregulates OspA and upregulates OspC another surface protein After the bacteria migrate from the midgut to the salivary glands OspC binds to Salp15 a tick salivary protein that appears to have immunosuppressive effects that enhance infection 68 Successful infection of the mammalian host depends on bacterial expression of OspC 69 Tick bites often go unnoticed because of the small size of the tick in its nymphal stage as well as tick secretions that prevent the host from feeling any itch or pain from the bite However transmission is quite rare with only about 1 2 to 1 4 percent of recognized tick bites resulting in Lyme disease 70 nbsp Tick Ixodes ricinus developmental stagesIn Europe the main vector is Ixodes ricinus which is also called the sheep tick or castor bean tick 71 In China Ixodes persulcatus the taiga tick is probably the most important vector 72 In North America the black legged tick or deer tick Ixodes scapularis is the main vector on the East Coast 65 The lone star tick Amblyomma americanum which is found throughout the Southeastern United States as far west as Texas is unlikely to transmit the Lyme disease spirochetes 73 though it may be implicated in a related syndrome called southern tick associated rash illness which resembles a mild form of Lyme disease 74 On the West Coast of the United States the main vector is the western black legged tick Ixodes pacificus 75 The tendency of this tick species to feed predominantly on host species such as the Western Fence Lizard that are resistant to Borrelia infection appears to diminish transmission of Lyme disease in the West 76 77 Transmission can occur across the placenta during pregnancy and as with a number of other spirochetal diseases adverse pregnancy outcomes are possible with untreated infection prompt treatment with antibiotics reduces or eliminates this risk 78 79 80 81 82 There is no scientific evidence to support Lyme disease transmission via blood transfusion sexual contact or breast milk 83 Tick borne co infections edit Main article Tick borne disease Ticks that transmit B burgdorferi to humans can also carry and transmit several other microbes such as Babesia microti and Anaplasma phagocytophilum which cause the diseases babesiosis and human granulocytic anaplasmosis HGA respectively 84 Among people with early Lyme disease depending on their location 2 12 will also have HGA and 2 10 will have babesiosis 52 Ticks in certain regions also transmit viruses that cause tick borne encephalitis and Powassan virus disease 85 52 Co infections of Lyme disease may not require additional treatment since they may resolve on their own or as in the case of HGA can be treated with the doxycycline prescribed for Lyme 35 Persistent fever or compatible anomalous laboratory findings may be indicative of a co infection 52 Pathophysiology editB burgdorferi can spread throughout the body during the course of the disease and has been found in the skin heart joints peripheral nervous system and central nervous system 69 86 B Burgdorferi does not produce toxins 87 Therefore many of the signs and symptoms of Lyme disease are a consequence of the immune response to spirochete in those tissues 88 B burgdorferi is injected into the skin by the bite of an infected Ixodes tick Tick saliva which accompanies the spirochete into the skin during the feeding process contains substances that disrupt the immune response at the site of the bite 89 This provides a protective environment where the spirochete can establish infection The spirochetes multiply and migrate outward within the dermis The host inflammatory response to the bacteria in the skin causes the characteristic circular EM lesion 69 Neutrophils however which are necessary to eliminate the spirochetes from the skin fail to appear in necessary numbers in the developing EM lesion because tick saliva inhibits neutrophil function This allows the bacteria to survive and eventually spread throughout the body 90 Days to weeks following the tick bite the spirochetes spread via the bloodstream to joints heart nervous system and distant skin sites where their presence gives rise to the variety of symptoms of the disseminated disease The spread of B burgdorferi is aided by the attachment of the host protease plasmin to the surface of the spirochete 91 If untreated the bacteria may persist in the body for months or even years despite the production of B burgdorferi antibodies by the immune system 92 The spirochetes may avoid the immune response by decreasing expression of surface proteins that are targeted by antibodies antigenic variation of the VlsE surface protein inactivating key immune components such as complement and hiding in the extracellular matrix which may interfere with the function of immune factors 93 94 In the brain B burgdorferi may induce astrocytes to undergo astrogliosis proliferation followed by apoptosis which may contribute to neurodysfunction 95 The spirochetes may also induce host cells to secrete quinolinic acid which stimulates the NMDA receptor on nerve cells which may account for the fatigue and malaise observed with Lyme encephalopathy 96 In addition diffuse white matter pathology during Lyme encephalopathy may disrupt gray matter connections and could account for deficits in attention memory visuospatial ability complex cognition and emotional status White matter disease may have a greater potential for recovery than gray matter disease perhaps because neuronal loss is less common Resolution of MRI white matter hyperintensities after antibiotic treatment has been observed 97 Tryptophan a precursor to serotonin appears to be reduced within the central nervous system in a number of infectious diseases that affect the brain including Lyme 98 Researchers are investigating if this neurohormone secretion is the cause of neuropsychiatric disorders developing in some people with borreliosis 99 Immunological studies edit Exposure to the Borrelia bacterium during Lyme disease possibly causes a long lived and damaging inflammatory response 100 a form of pathogen induced autoimmune disease 101 The production of this reaction might be due to a form of molecular mimicry where Borrelia avoids being killed by the immune system by resembling normal parts of the body s tissues 102 103 Chronic symptoms from an autoimmune reaction could explain why some symptoms persist even after the spirochetes have been eliminated from the body This hypothesis may explain why chronic arthritis persists after antibiotic therapy similar to rheumatic fever but its wider application is controversial 104 105 Diagnosis editLyme disease is diagnosed based on symptoms objective physical findings such as erythema migrans EM rash facial palsy or arthritis history of possible exposure to infected ticks and possibly laboratory tests 3 27 People with symptoms of early Lyme disease should have a total body skin examination for EM rashes and asked whether EM type rashes had manifested within the last 1 2 months 35 Presence of an EM rash and recent tick exposure i e being outdoors in a likely tick habitat where Lyme is common within 30 days of the appearance of the rash are sufficient for Lyme diagnosis no laboratory confirmation is needed or recommended 3 27 106 107 Most people who get infected do not remember a tick or a bite and the EM rash need not look like a bull s eye most EM rashes in the U S do not or be accompanied by any other symptoms 3 108 In the U S Lyme is most common in the New England and Mid Atlantic states and parts of Wisconsin and Minnesota but it is expanding into other areas 67 Several bordering areas of Canada also have high Lyme risk 109 In the absence of an EM rash or history of tick exposure Lyme diagnosis depends on laboratory confirmation 62 110 The bacteria that cause Lyme disease are difficult to observe directly in body tissues and also difficult and too time consuming to grow in the laboratory 3 62 The most widely used tests look instead for presence of antibodies against those bacteria in the blood 111 A positive antibody test result does not by itself prove active infection but can confirm an infection that is suspected because of symptoms objective findings and history of tick exposure in a person 62 Because as many as 5 20 of the normal population have antibodies against Lyme people without history and symptoms suggestive of Lyme disease should not be tested for Lyme antibodies a positive result would likely be false possibly causing unnecessary treatment 35 37 In some cases when history signs and symptoms are strongly suggestive of early disseminated Lyme disease empiric treatment may be started and reevaluated as laboratory test results become available 41 112 Laboratory testing edit Tests for antibodies in the blood by ELISA and Western blot is the most widely used method for Lyme diagnosis A two tiered protocol is recommended by the Centers for Disease Control and Prevention CDC the sensitive ELISA test is performed first and if it is positive or equivocal then the more specific Western blot is run 113 The immune system takes some time to produce antibodies in quantity After Lyme infection onset antibodies of types IgM and IgG usually can first be detected respectively at 2 4 weeks and 4 6 weeks and peak at 6 8 weeks 114 When an EM rash first appears detectable antibodies may not be present Therefore it is recommended that testing not be performed and diagnosis be based on the presence of the EM rash 35 Up to 30 days after suspected Lyme infection onset infection can be confirmed by detection of IgM or IgG antibodies after that it is recommended that only IgG antibodies be considered 114 A positive IgM and negative IgG test result after the first month of infection is generally indicative of a false positive result 115 The number of IgM antibodies usually collapses 4 6 months after infection while IgG antibodies can remain detectable for years 114 Other tests may be used in neuroborreliosis cases In Europe neuroborreliosis is usually caused by Borrelia garinii and almost always involves lymphocytic pleocytosis i e the densities of lymphocytes infection fighting cells and protein in the cerebrospinal fluid CSF typically rise to characteristically abnormal levels while glucose level remains normal 38 35 40 Additionally the immune system produces antibodies against Lyme inside the intrathecal space which contains the CSF 35 40 Demonstration by lumbar puncture and CSF analysis of pleocytosis and intrathecal antibody production are required for definite diagnosis of neuroborreliosis in Europe except in cases of peripheral neuropathy associated with acrodermatitis chronica atrophicans which usually is caused by Borrelia afzelii and confirmed by blood antibody tests 37 In North America neuroborreliosis is caused by Borrelia burgdorferi and may not be accompanied by the same CSF signs they confirm a diagnosis of central nervous system CNS neuroborreliosis if positive but do not exclude it if negative 116 American guidelines consider CSF analysis optional when symptoms appear to be confined to the peripheral nervous system PNS e g facial palsy without overt meningitis symptoms 35 117 Unlike blood and intrathecal antibody tests CSF pleocytosis tests revert to normal after infection ends and therefore can be used as objective markers of treatment success and inform decisions on whether to retreat 40 In infection involving the PNS electromyography and nerve conduction studies can be used to monitor objectively the response to treatment 38 In Lyme carditis electrocardiograms are used to evidence heart conduction abnormalities while echocardiography may show myocardial dysfunction 43 Biopsy and confirmation of Borrelia cells in myocardial tissue may be used in specific cases but are usually not done because of risk of the procedure 43 Polymerase chain reaction PCR tests for Lyme disease have also been developed to detect the genetic material DNA of the Lyme disease spirochete Culture or PCR are the current means for detecting the presence of the organism as serologic studies only test for antibodies of Borrelia PCR has the advantage of being much faster than culture However PCR tests are susceptible to false positive results e g by detection of debris of dead Borrelia cells or specimen contamination 118 37 Even when properly performed PCR often shows false negative results because few Borrelia cells can be found in blood and cerebrospinal fluid CSF during infection 119 37 Hence PCR tests are recommended only in special cases e g diagnosis of Lyme arthritis because it is a highly sensitive way of detecting ospA DNA in synovial fluid 120 Although sensitivity of PCR in CSF is low its use may be considered when intrathecal antibody production test results are suspected of being falsely negative e g in very early lt 6 weeks neuroborreliosis or in immunosuppressed people 37 Several other forms of laboratory testing for Lyme disease are available some of which have not been adequately validated OspA antigens shed by live Borrelia bacteria into urine are a promising technique being studied 121 The use of nanotrap particles for their detection is being looked at and the OspA has been linked to active symptoms of Lyme 122 123 High titers of either immunoglobulin G IgG or immunoglobulin M IgM antibodies to Borrelia antigens indicate disease but lower titers can be misleading because the IgM antibodies may remain after the initial infection and IgG antibodies may remain for years 124 The CDC does not recommend urine antigen tests PCR tests on urine immunofluorescent staining for cell wall deficient forms of B burgdorferi and lymphocyte transformation tests 119 Imaging edit Neuroimaging is controversial in whether it provides specific patterns unique to neuroborreliosis but may aid in differential diagnosis and in understanding the pathophysiology of the disease 125 Though controversial some evidence shows certain neuroimaging tests can provide data that are helpful in the diagnosis of a person Magnetic resonance imaging MRI and single photon emission computed tomography SPECT are two of the tests that can identify abnormalities in the brain of a person affected with this disease Neuroimaging findings in an MRI include lesions in the periventricular white matter as well as enlarged ventricles and cortical atrophy The findings are considered somewhat unexceptional because the lesions have been found to be reversible following antibiotic treatment Images produced using SPECT show numerous areas where an insufficient amount of blood is being delivered to the cortex and subcortical white matter However SPECT images are known to be nonspecific because they show a heterogeneous pattern in the imaging The abnormalities seen in the SPECT images are very similar to those seen in people with cerebral vasculitis and Creutzfeldt Jakob disease which makes them questionable 126 Differential diagnosis edit Community clinics have been reported to misdiagnose 23 28 of Erythema migrans EM rashes and 83 of other objective manifestations of early Lyme disease 110 EM rashes are often misdiagnosed as spider bites cellulitis or shingles 110 Many misdiagnoses are credited to the widespread misconception that EM rashes should look like a bull s eye 3 Actually the key distinguishing features of the EM rash are the speed and extent to which it expands respectively up to 2 3 cm day and a diameter of at least 5 cm and in 50 of cases more than 16 cm The rash expands away from its center which may or may not look different or be separated by ring like clearing from the rest of the rash 27 28 Compared to EM rashes spider bites are more common in the limbs tend to be more painful and itchy or become swollen and some may cause necrosis sinking dark blue patch of dead skin 27 3 Cellulitis most commonly develops around a wound or ulcer is rarely circular and is more likely to become swollen and tender 27 3 EM rashes often appear at sites that are unusual for cellulitis such as the armpit groin abdomen or back of knee 27 Like Lyme shingles often begins with headache fever and fatigue which are followed by pain or numbness However unlike Lyme in shingles these symptoms are usually followed by appearance of rashes composed of multiple small blisters along with a nerve s dermatome and shingles can also be confirmed by quick laboratory tests 127 Facial palsy caused by Lyme disease LDFP is often misdiagnosed as Bell s palsy 41 Although Bell s palsy is the most common type of one sided facial palsy about 70 of cases LDFP can account for about 25 of cases of facial palsy in areas where Lyme disease is common 41 Compared to LDFP Bell s palsy much less frequently affects both sides of the face 41 Even though LDFP and Bell s palsy have similar symptoms and evolve similarly if untreated corticosteroid treatment is beneficial for Bell s Palsy while being detrimental for LDFP 41 Recent history of exposure to a likely tick habitat during warmer months EM rash viral like symptoms such as headache and fever and or palsy in both sides of the face should be evaluated for the likelihood of LDFP if it is more than minimal empiric therapy with antibiotics should be initiated without corticosteroids and reevaluated upon completion of laboratory tests for Lyme disease 41 Unlike viral meningitis Lyme lymphocytic meningitis tends to not cause fever last longer and recur 38 35 Lymphocytic meningitis is also characterized by possibly co occurring with EM rash facial palsy or partial vision obstruction and having much lower percentage of polymorphonuclear leukocytes in CSF 35 Lyme radiculopathy affecting the limbs is often misdiagnosed as a radiculopathy caused by nerve root compression such as sciatica 110 128 Although most cases of radiculopathy are compressive and resolve with conservative treatment e g rest within 4 6 weeks guidelines for managing radiculopathy recommend first evaluating risks of other possible causes that although less frequent require immediate diagnosis and treatment including infections such as Lyme and shingles 129 A history of outdoor activities in likely tick habitats in the last 3 months possibly followed by a rash or viral like symptoms and current headache other symptoms of lymphocytic meningitis or facial palsy would lead to suspicion of Lyme disease and recommendation of serological and lumbar puncture tests for confirmation 129 Lyme radiculopathy affecting the trunk can be misdiagnosed as myriad other conditions such as diverticulitis and acute coronary syndrome 42 110 Diagnosis of late stage Lyme disease is often complicated by a multifaceted appearance and nonspecific symptoms prompting one reviewer to call Lyme the new great imitator 130 As all people with later stage infection will have a positive antibody test simple blood tests can exclude Lyme disease as a possible cause of a person s symptoms 131 Prevention editTick bites may be prevented by avoiding or reducing time in likely tick habitats and taking precautions while in and when getting out of one 132 8 Most Lyme human infections are caused by Ixodes nymph bites between April and September 27 132 Ticks prefer moist shaded locations in woodlands shrubs tall grasses and leaf litter or wood piles 27 133 Tick densities tend to be highest in woodlands followed by unmaintained edges between woods and lawns about half as high ornamental plants and perennial groundcover about a quarter and lawns about 30 times less 134 Ixodes larvae and nymphs tend to be abundant also where mice nest such as stone walls and wood logs 134 Ixodes larvae and nymphs typically wait for potential hosts quest on leaves or grasses close to the ground with forelegs outstretched when a host brushes against its limbs the tick rapidly clings and climbs on the host looking for a skin location to bite 135 In Northeastern United States 69 of tick bites are estimated to happen in residences 11 in schools or camps 9 in parks or recreational areas 4 at work 3 while hunting and 4 in other areas 134 Activities associated with tick bites around residences include yard work brush clearing gardening playing in the yard and letting into the house dogs or cats that roam outside in woody or grassy areas 134 132 In parks tick bites often happen while hiking or camping 134 Walking on a mowed lawn or center of a trail without touching adjacent vegetation is less risky than crawling or sitting on a log or stone wall 134 136 Pets should not be allowed to roam freely in likely tick habitats 133 As a precaution CDC recommends soaking or spraying clothes shoes and camping gear such as tents backpacks and sleeping bags with 0 5 permethrin solution and hanging them to dry before use 132 137 Permethrin is odorless and safe for humans but highly toxic to ticks 138 After crawling on permethrin treated fabric for as few as 10 20 seconds tick nymphs become irritated and fall off or die 138 139 Permethrin treated closed toed shoes and socks reduce by 74 times the number of bites from nymphs that make first contact with a shoe of a person also wearing treated shorts because nymphs usually quest near the ground this is a typical contact scenario 138 Better protection can be achieved by tucking permethrin treated trousers pants into treated socks and a treated long sleeve shirt into the trousers so as to minimize gaps through which a tick might reach the wearer s skin 136 Light colored clothing may make it easier to see ticks and remove them before they bite 136 Military and outdoor workers uniforms treated with permethrin have been found to reduce the number of bite cases by 80 95 139 Permethrin protection lasts several weeks of wear and washings in customer treated items and up to 70 washings for factory treated items 137 Permethrin should not be used on human skin underwear or cats 137 140 The EPA recommends several tick repellents for use on exposed skin including DEET picaridin IR3535 a derivative of amino acid beta alanine oil of lemon eucalyptus OLE a natural compound and OLE s active ingredient para menthane diol PMD 132 141 142 Unlike permethrin repellents repel but do not kill ticks protect for only several hours after application and may be washed off by sweat or water 137 The most popular repellent is DEET in the U S and picaridin in Europe 142 Unlike DEET picaridin is odorless and is less likely to irritate the skin or harm fabric or plastics 142 Repellents with higher concentration may last longer but are not more effective against ticks 20 picaridin may work for 8 hours vs 55 98 11 DEET for 5 6 hours or 30 40 OLE for 6 hours 137 141 Repellents should not be used under clothes on eyes mouth wounds or cuts or on babies younger than 2 months 3 years for OLE or PMD 137 132 If sunscreen is used repellent should be applied on top of it 137 Repellents should not be sprayed directly on a face but should instead be sprayed on a hand and then rubbed on the face 137 After coming indoors clothes gear and pets should be checked for ticks 132 Clothes can be put into a hot dryer for 10 minutes to kill ticks just washing or warm dryer are not enough 132 Showering as soon as possible looking for ticks over the entire body and removing them reduce risk of infection 132 Unfed tick nymphs are the size of a poppy seed but a day or two after biting and attaching themselves to a person they look like a small blood blister 143 The following areas should be checked especially carefully armpits between legs back of knee bellybutton trunk and in children ears neck and hair 132 Tick removal edit nbsp Removal of a tick using tweezersAttached ticks should be removed promptly Risk of infection increases with time of attachment but in North America risk of Lyme disease is small if the tick is removed within 36 hours 144 CDC recommends inserting a fine tipped tweezer between the skin and the tick grasping very firmly and pulling the closed tweezer straight away from the skin without twisting jerking squeezing or crushing the tick 145 After tick removal any tick parts remaining in the skin should be removed with a clean tweezer if possible 145 The wound and hands should then be cleaned with alcohol or soap and water 145 The tick may be disposed by placing it in a container with alcohol sealed bag tape or flushed down the toilet 145 The bitten person should write down where and when the bite happened so that this can be informed to a doctor if the person gets a rash or flu like symptoms in the following several weeks 145 CDC recommends not using fingers nail polish petroleum jelly or heat on the tick to try to remove it 145 In Australia where the Australian paralysis tick is prevalent the Australasian Society of Clinical Immunology and Allergy recommends not using tweezers to remove ticks because if the person is allergic anaphylaxis could result 146 Instead a product should be sprayed on the tick to cause it to freeze and then drop off 146 Another method consists in using about 20 cm of dental floss or fishing line for slowly tying an overhand knot between the skin and the tick and then pulling it away from the skin 147 148 Preventive antibiotics edit The risk of infectious transmission increases with the duration of tick attachment 27 It requires between 36 and 48 hours of attachment for the bacteria that causes Lyme to travel from within the tick into its saliva 27 If a deer tick that is sufficiently likely to be carrying Borrelia is found attached to a person and removed and if the tick has been attached for 36 hours or is engorged a single dose of doxycycline administered within the 72 hours after removal may reduce the risk of Lyme disease It is not generally recommended for all people bitten as development of infection is rare about 50 bitten people would have to be treated this way to prevent one case of erythema migrans i e the typical rash found in about 70 80 of people infected 3 27 Garden landscaping edit Several landscaping practices may reduce the risk of tick bites in residential yards 143 149 These include keeping lawns mowed removing leaf litter and weeds and avoiding the use of ground cover 143 A 3 ft wide rock or woodchip barrier is recommended to separate lawns from wood piles woodlands stone walls and shrubs 149 Without vegetation on the barrier ticks will tend not to cross it acaricides may also be sprayed on it to kill ticks 149 A sun exposed tick safe zone at least 9 ft from the barrier should concentrate human activity on the yard including any patios playgrounds and gardening 149 Materials such as wood decking concrete bricks gravel or woodchips used on the ground under patios and playgrounds would discourage ticks there 143 An 8 ft high fence may be added to keep deer away from the tick safe zone 149 143 Occupational exposure edit Outdoor workers are at risk of Lyme disease if they work at sites with infected ticks This includes construction landscaping forestry brush clearing land surveying farming railroad work oil field work utility line work park or wildlife management 150 151 U S workers in the northeastern and north central states are at highest risk of exposure to infected ticks Ticks may also transmit other tick borne diseases to workers in these and other regions of the country Worksites with woods bushes high grass or leaf litter are likely to have more ticks Outdoor workers should be most careful to protect themselves in the late spring and summer when young ticks are most active 152 Host animals edit Ticks can feed upon the blood of a wide array of possible host species including lizards birds mice cats dogs deer cattle and humans The extent to which a tick can feed reproduce and spread will depend on the type and availability of its hosts Whether it will spread disease is also affected by its available hosts Some species such as lizards are referred to as dilution hosts because they don t tend to support Lyme disease pathogens and so decrease the likelihood that the disease will be passed on by ticks feeding on them White tailed deer are both a food source and a reproductive host where ticks tend to mate The white footed mouse is a reservoir host in which the pathogen for Lyme disease can survive Availability of hosts can have significant impacts on the transmission of Lyme disease A greater diversity of hosts or of those that don t support the pathogen tends to decrease the likelihood that the disease will be transmitted 153 In the United States one approach to reducing the incidence of Lyme and other deer tick borne diseases has been to greatly reduce the deer population on which the adult ticks depend for feeding and reproduction Lyme disease cases fell following deer eradication on an island Monhegan Maine 154 and following deer control in Mumford Cove Connecticut 155 Advocates have suggested reducing the deer population to levels of 8 to 10 deer per square mile compared to levels of 60 or more deer per square mile in the areas of the country with the highest Lyme disease rates 156 Others have noted that while deer are reproductive hosts they are not Borrelia burgdorferi reservoirs Researchers have suggested that smaller less obviously visible Lyme reservoirs like white footed mice and Eastern chipmunks may more strongly impact Lyme disease occurrence Ecosystem studies in New York state suggest that white footed mice thrive when forests are broken into smaller isolated chunks of woodland with fewer rodent predators With more rodents harboring the disease the odds increase that a tick will feed on a disease harboring rodent and that someone will pick up a disease carrying tick in their garden or walking in the woods Data indicates that the smaller the wooded area the more ticks it will contain and the likely they are to carry Lyme disease supporting the idea that deforestation and habitat fragmentation affect ticks hosts and disease transmission 153 Tick borne diseases are estimated to affect 80 of cattle worldwide 157 They also affect cats dogs and other pets Routine veterinary control of ticks of domestic animals through the use of acaricides has been suggested as a way to reduce exposure of humans to ticks However chemical control with acaricides is now criticized on a number of grounds Ticks appear to develop resistance to acaricides acaricides are costly and there are concerns over their toxicity and the potential for chemical residues to affect food and the environment 158 In Europe known reservoirs of Borrelia burgdorferi were 9 small mammals 7 medium sized mammals and 16 species of birds including passerines sea birds and pheasants 159 These animals seem to transmit spirochetes to ticks and thus participate in the natural circulation of B burgdorferi in Europe The house mouse is also suspected as well as other species of small rodents particularly in Eastern Europe and Russia 159 The reservoir species that contain the most pathogens are the European roe deer Capreolus capreolus 160 it does not appear to serve as a major reservoir of B burgdorferi thought Jaenson amp al 1992 161 incompetent host for B burgdorferi and TBE virus but it is important for feeding the ticks 162 as red deer and wild boars Sus scrofa 160 in which one Rickettsia and three Borrelia species were identified 160 with high risks of coinfection in roe deer 163 Nevertheless in the 2000s in roe deer in Europe two species of Rickettsia and two species of Borrelia were identified 160 Vaccination editAs of 2023 update no human vaccines for Lyme disease were available 26 The only human vaccine to advance to market was LYMErix which was available from 1998 but discontinued in 2002 164 The vaccine candidate VLA15 was scheduled to start a phase 3 trial in the third quarter of 2022 with other research ongoing Multiple vaccines are available for the prevention of Lyme disease in dogs LYMErix edit The vaccine LYMErix was available from 1998 to 2002 The recombinant vaccine against Lyme disease based on the outer surface protein A OspA of B burgdorferi with aluminum hydroxide as adjuvant was developed by SmithKline Beecham In clinical trials involving more than 10 000 people the vaccine was found to confer protective immunity to Lyme disease in 76 of adults after three doses with only mild or moderate and transient adverse effects 165 166 On 21 December 1998 the Food and Drug Administration FDA approved LYMErix on the basis of these trials for persons of ages 15 through 70 166 164 Following approval of the vaccine its entry into clinical practice was slow for a variety of reasons including its cost which was often not reimbursed by insurance companies 167 Subsequently hundreds of vaccine recipients reported they had developed autoimmune and other side effects Supported by some advocacy groups a number of class action lawsuits were filed against GlaxoSmithKline alleging the vaccine had caused these health problems These claims were investigated by the FDA and the Centers for Disease Control which found no connection between the vaccine and the autoimmune complaints 168 Despite the lack of evidence that the complaints were caused by the vaccine sales plummeted and LYMErix was withdrawn from the U S market by GlaxoSmithKline in February 2002 169 in the setting of negative media coverage and fears of vaccine side effects 168 170 The fate of LYMErix was described in the medical literature as a cautionary tale 170 an editorial in Nature cited the withdrawal of LYMErix as an instance in which unfounded public fears place pressures on vaccine developers that go beyond reasonable safety considerations 171 The original developer of the OspA vaccine at the Max Planck Institute told Nature This just shows how irrational the world can be There was no scientific justification for the first OspA vaccine LYMErix being pulled 168 172 VLA15 edit The hexavalent OspA protein subunit based vaccine candidate VLA15 was developed by Valneva It was granted fast track designation by the U S Food and Drug Administration in July 2017 173 174 In April 2020 Pfizer paid 130 million for the rights to the vaccine and the companies are developing it together performing multiple phase 2 trials 175 A phase 3 trial of VLA15 was scheduled for late 2022 recruiting volunteers at test sites located across the northeastern United States and in Europe 176 177 Participants were scheduled to receive an initial three dose series of vaccines over the course of five to nine months followed by a booster dose after twelve months with both the initial series and the booster dose scheduled to be complete before the year s peak Lyme disease season 177 Other research edit An mRNA vaccine designed to cause a strong fast immune response to tick saliva allowed the immune system to detect and remove the ticks from test animals before they were able to transmit the infectious bacteria 178 The vaccine contains mRNAs for the body to build 19 proteins in tick saliva which by enabling quick development of erythema itchy redness at the bite site protects guinea pigs against Lyme disease It also protected the test animals if the tick is not removed if only one tick but not three remain attached 179 180 Canine vaccines edit Canine vaccines have been formulated and approved for the prevention of Lyme disease in dogs Currently three Lyme disease vaccines are available LymeVax formulated by Fort Dodge Laboratories contains intact dead spirochetes which expose the host to the organism Galaxy Lyme Intervet Schering Plough s vaccine targets proteins OspC and OspA The OspC antibodies kill any of the bacteria that have not been killed by the OspA antibodies Canine Recombinant Lyme formulated by Merial generates antibodies against the OspA protein so a tick feeding on a vaccinated dog draws in blood full of anti OspA antibodies which kill the spirochetes in the tick s gut before they are transmitted to the dog 181 Treatment editAntibiotics are the primary treatment 3 27 The specific approach to their use is dependent on the individual affected and the stage of the disease 27 For most people with early localized infection oral administration of doxycycline is widely recommended as the first choice as it is effective against not only Borrelia bacteria but also a variety of other illnesses carried by ticks 27 People taking doxycycline should avoid sun exposure because of higher risk of sunburns 35 Doxycycline is contraindicated in children younger than eight years of age and women who are pregnant or breastfeeding 27 alternatives to doxycycline are amoxicillin cefuroxime axetil and azithromycin 27 Azithromycin is recommended only in case of intolerance to the other antibiotics 35 The standard treatment for cellulitis cephalexin is not useful for Lyme disease 35 When it is unclear if a rash is caused by Lyme or cellulitis the IDSA recommends treatment with cefuroxime or amoxicillin clavulanic acid as these are effective against both infections 35 Individuals with early disseminated or late Lyme infection may have symptomatic cardiac disease Lyme arthritis or neurologic symptoms like facial palsy radiculopathy meningitis or peripheral neuropathy 27 Intravenous administration of ceftriaxone is recommended as the first choice in these cases 27 cefotaxime and doxycycline are available as alternatives 27 Treatment regimens for Lyme disease range from 14 days in early localized disease to 14 21 days in early disseminated disease to 14 28 days in late disseminated disease 182 Neurologic complications of Lyme disease may be treated with doxycycline as it can be taken by mouth and has a lower cost although in North America evidence of efficacy is only indirect 117 In case of failure guidelines recommend retreatment with injectable ceftriaxone 117 Several months after treatment for Lyme arthritis if joint swelling persists or returns a second round of antibiotics may be considered intravenous antibiotics are preferred for retreatment in case of poor response to oral antibiotics 27 35 Outside of that a prolonged antibiotic regimen lasting more than 28 days is not recommended as no evidence shows it to be effective 27 183 IgM and IgG antibody levels may be elevated for years even after successful treatment with antibiotics 27 As antibody levels are not indicative of treatment success testing for them is not recommended 27 Facial palsy may resolve without treatment however antibiotic treatment is recommended to stop other Lyme complications 35 Corticosteroids are not recommended when facial palsy is caused by Lyme disease 41 In those with facial palsy frequent use of artificial tears while awake is recommended along with ointment and a patch or taping the eye closed when sleeping 41 184 About a third of people with Lyme carditis need a temporary pacemaker until their heart conduction abnormality resolves and 21 need to be hospitalized 43 Lyme carditis should not be treated with corticosteroids 43 People with Lyme arthritis should limit their level of physical activity to avoid damaging affected joints and in case of limping should use crutches 185 Pain associated with Lyme disease may be treated with nonsteroidal anti inflammatory drugs NSAIDs 35 Corticosteroid joint injections are not recommended for Lyme arthritis that is being treated with antibiotics 35 185 People with Lyme arthritis treated with intravenous antibiotics or two months of oral antibiotics who continue to have joint swelling two months after treatment and have negative PCR test for Borrelia DNA in the synovial fluid are said to have post antibiotic Lyme arthritis this is more common after infection by certain Borrelia strains in people with certain genetic and immunologic characteristics 35 52 185 Post antibiotic Lyme arthritis may be symptomatically treated with NSAIDs disease modifying antirheumatic drugs DMARDs arthroscopic synovectomy or physical therapy 52 185 People receiving treatment should be advised that reinfection is possible and how to prevent it 112 Prognosis editLyme disease s typical first sign the erythema migrans EM rash resolves within several weeks even without treatment 3 However in untreated people the infection often disseminates to the nervous system heart or joints possibly causing permanent damage to body tissues 35 People who receive recommended antibiotic treatment within several days of appearance of an initial EM rash have the best prospects 110 Recovery may not be total or immediate The percentage of people achieving full recovery in the United States increases from about 64 71 at end of treatment for EM rash to about 84 90 after 30 months higher percentages are reported in Europe 186 187 Treatment failure i e persistence of original or appearance of new signs of the disease occurs only in a few people 186 Remaining people are considered cured but continue to experience subjective symptoms e g joint or muscle pains or fatigue 188 These symptoms usually are mild and nondisabling 188 People treated only after nervous system manifestations of the disease may end up with objective neurological deficits in addition to subjective symptoms 35 In Europe an average of 32 33 months after initial Lyme symptoms in people treated mostly with doxycycline 200 mg for 14 21 days the percentage of people with lingering symptoms was much higher among those diagnosed with neuroborreliosis 50 than among those with only an EM rash 16 189 In another European study 5 years after treatment for neuroborreliosis lingering symptoms were less common among children 15 than adults 30 and in the latter was less common among those treated within 30 days of the first symptom 16 than among those treated later 39 among those with lingering symptoms 54 had daily activities restricted and 19 were on sick leave or incapacitated 190 Some data suggest that about 90 of Lyme facial palsies treated with antibiotics recover fully a median of 24 days after appearing and most of the rest recover with only mild abnormality 191 192 However in Europe 41 of people treated for facial palsy had other lingering symptoms at followup up to 6 months later including 28 with numbness or altered sensation and 14 with fatigue or concentration problems 192 Palsies in both sides of the face are associated with worse and longer time to recovery 191 192 Historical data suggests that untreated people with facial palsies recover at nearly the same rate but 88 subsequently have Lyme arthritis 191 193 Other research shows that synkinesis involuntary movement of a facial muscle when another one is voluntarily moved can become evident only 6 12 months after facial palsy appears to be resolved as damaged nerves regrow and sometimes connect to incorrect muscles 194 Synkinesis is associated with corticosteroid use 194 In longer term follow up 16 23 of Lyme facial palsies do not fully recover 194 In Europe about a quarter of people with Bannwarth syndrome Lyme radiculopathy and lymphocytic meningitis treated with intravenous ceftriaxone for 14 days an average of 30 days after first symptoms had to be retreated 3 6 months later because of unsatisfactory clinical response or continued objective markers of infection in cerebrospinal fluid after 12 months 64 recovered fully 31 had nondisabling mild or infrequent symptoms that did not require regular use of analgesics and 5 had symptoms that were disabling or required substantial use of analgesics 40 The most common lingering nondisabling symptoms were headache fatigue altered sensation joint pains memory disturbances malaise radicular pain sleep disturbances muscle pains and concentration disturbances Lingering disabling symptoms included facial palsy and other impaired movement 40 Recovery from late neuroborreliosis tends to take longer and be less complete than from early neuroborreliosis probably because of irreversible neurologic damage 35 About half the people with Lyme carditis progress to complete heart block but it usually resolves in a week 43 Other Lyme heart conduction abnormalities resolve typically within 6 weeks 43 About 94 of people have full recovery but 5 need a permanent pacemaker and 1 end up with persistent heart block the actual percentage may be higher because of unrecognized cases 43 Lyme myocardial complications usually are mild and self limiting 43 However in some cases Lyme carditis can be fatal 43 Recommended antibiotic treatments are effective in about 90 of Lyme arthritis cases although it can take several months for inflammation to resolve and a second round of antibiotics is often necessary 35 Antibiotic refractory Lyme arthritis also eventually resolves typically within 9 14 months range 4 months 4 years DMARDs or synovectomy can accelerate recovery 185 Reinfection is not uncommon In a U S study 6 11 of people treated for an EM rash had another EM rash within 30 months 186 The second rash typically is due to infection by a different Borrelia strain 195 Post treatment Lyme disease syndrome edit Chronic symptoms like pain fatigue or cognitive impairment are experienced by 10 20 of people who contract Lyme disease even after completing treatment 196 197 The cause is unknown One hypothesis is autoimmunity triggered by the infection which may persist long term after the triggering infection is resolved Auto immune responses are known to occur following other infections including Campylobacter Guillain Barre syndrome Chlamydia reactive arthritis and strep throat rheumatic heart disease Another is that a persistent difficult to detect infection causes PTLDS A third proposal is that symptoms are unrelated to a previous case of Lyme Disease but have other causes 197 There is no proven treatment for PTLDS While short term antibiotics are effective in early Lyme disease prolonged antibiotics are not They have been shown ineffective in placebo controlled trials and carry the risk of serious sometimes deadly complications Generally treatment is symptomatic and is similar to the management of fibromyalgia or ME CFS PTLDS usually gets better over time but recovery may take many months 197 Epidemiology edit nbsp Countries with reported Lyme disease casesLyme disease occurs regularly in Northern Hemisphere temperate regions 198 An estimated 476 000 people a year are diagnosed and treated for the disease in the United States This number is likely an overestimate due to overdiagnosis and overtreatment 5 6 Over 200 000 people a year are diagnosed and treated in Europe 7 3 199 There is a suggestion that tick populations and Lyme disease occurrence are increasing and spreading into new areas due in part to the warming temperatures of climate change However tick borne disease systems are complex and determining whether changes are due to climate change or other drivers can be difficult 153 200 Africa edit In northern Africa B burgdorferi sensu lato has been identified in Morocco Algeria Egypt and Tunisia 201 202 203 Lyme disease in sub Saharan Africa is presently unknown but evidence indicates it may occur in humans in this region The abundance of hosts and tick vectors would favor the establishment of Lyme infection in Africa 204 In East Africa two cases of Lyme disease have been reported in Kenya 205 According The Federation of Infectious Diseases Societies of Southern Africa Lyme disease is not known to be endemic in either South Africa or Mozambique 206 Asia edit B burgdorferi sensu lato infested ticks are being found more frequently in Japan as well as in northwest China Nepal Thailand and far eastern Russia 207 208 Borrelia has also been isolated in Mongolia 209 Australia edit Lyme disease is not considered endemic to Australia 210 While there have been reports of people acquiring Lyme disease in Australia and even evidence of closely related Borrelia species in ticks 211 the evidence linking these cases to local transmission is limited Ongoing research on resolving potential Borrelia species to Debilitating Symptom Complexes Attributed to Ticks DSCATT in Australia are ongoing 212 Europe edit In Europe Lyme disease is caused by infection with one or more pathogenic European genospecies of the spirochaete B burgdorferi sensu lato mainly transmitted by the tick Ixodes ricinus 213 Cases of B burgdorferi sensu lato infected ticks are found predominantly in central Europe particularly in Slovenia and Austria but have been isolated in almost every country on the continent 214 Number of cases in southern Europe such as Italy and Portugal is much lower 215 Diagnosed cases in some Western countries such as Iceland are rising 216 Lyme disease is rare in Iceland On average around 6 to 7 cases are diagnosed every year primarily localised infections presenting as erythema migrans None of the cases had a definitive Icelandic origin and the yearly number of cases has not been increasing 217 United Kingdom edit In the United Kingdom the number of laboratory confirmed cases of Lyme disease has been rising steadily since voluntary reporting was introduced in 1986 218 when 68 cases were recorded in the UK and Ireland combined 219 In the UK there were 23 confirmed cases in 1988 and 19 in 1990 220 but 973 in 2009 218 and 953 in 2010 221 Provisional figures for the first 3 quarters of 2011 show a 26 increase on the same period in 2010 222 It is thought however that the actual number of cases is significantly higher than suggested by the above figures with the UK s Health Protection Agency estimating that there are between 2 000 and 3 000 cases per year 221 with an average of around 15 of the infections acquired overseas 218 while Dr Darrel Ho Yen Director of the Scottish Toxoplasma Reference Laboratory and National Lyme Disease Testing Service believes that the number of confirmed cases should be multiplied by 10 to take account of wrongly diagnosed cases tests giving false results sufferers who weren t tested people who are infected but not showing symptoms failures to notify and infected individuals who don t consult a doctor 223 224 Despite Lyme disease Borrelia burgdorferi infection being a notifiable disease in Scotland 225 since January 1990 226 which should therefore be reported on the basis of clinical suspicion it is believed that many GPs are unaware of the requirement 227 Mandatory reporting limited to laboratory test results only was introduced throughout the UK in October 2010 under the Health Protection Notification Regulations 2010 218 Although there is a greater number of cases of Lyme disease in the New Forest Salisbury Plain Exmoor the South Downs parts of Wiltshire and Berkshire Thetford Forest 228 and the West coast and islands of Scotland 229 infected ticks are widespread and can even be found in the parks of London 220 230 A 1989 report found that 25 of forestry workers in the New Forest were seropositive as were between 2 and 4 5 of the general local population of the area 231 232 Tests on pet dogs carried out throughout the country in 2009 indicated that around 2 5 of ticks in the UK may be infected considerably higher than previously thought 233 234 It is speculated that global warming may lead to an increase in tick activity in the future as well as an increase in the amount of time that people spend in public parks thus increasing the risk of infection 235 However no published research has proven this to be so North America edit Many studies in North America have examined ecological and environmental correlates of the number of people affected by Lyme disease A 2005 study using climate suitability modelling of I scapularis projected that climate change would cause an overall 213 increase in suitable vector habitat by 2080 with northward expansions in Canada increased suitability in the central U S and decreased suitable habitat and vector retraction in the southern U S 236 A 2008 review of published studies concluded that the presence of forests or forested areas was the only variable that consistently elevated the risk of Lyme disease whereas other environmental variables showed little or no concordance between studies 237 The authors argued that the factors influencing tick density and human risk between sites are still poorly understood and that future studies should be conducted over longer time periods become more standardized across regions and incorporate existing knowledge of regional Lyme disease ecology 237 Canada edit The range of ticks able to carry Lyme disease has expanded from a limited area of Ontario to include areas of southern Quebec Manitoba northern Ontario southern New Brunswick southwest Nova Scotia and limited parts of Saskatchewan and Alberta as well as British Columbia Cases have been reported as far east as the island of Newfoundland 109 238 239 240 A model based prediction by Leighton et al 2012 suggests that the range of the I scapularis tick will expand into Canada by 46 km year over the next decade with warming climatic temperatures as the main driver of increased speed of spread 241 Mexico edit A 2007 study suggests Borrelia burgdorferi infections are endemic to Mexico from four cases reported between 1999 and 2000 242 United States edit nbsp CDC map showing the risk of Lyme disease in the United States particularly its concentration in the Northeast Megalopolis and western WisconsinLyme disease is the most common tick borne disease in North America and Europe and one of the fastest growing infectious diseases in the United States Of cases reported to the United States CDC the ratio of Lyme disease infection is 7 9 cases for every 100 000 persons In the ten states where Lyme disease is most common the average was 31 6 cases for every 100 000 persons for the year 2005 243 244 245 Although Lyme disease has been reported in all states due to travel associated infections about 99 of all reported cases are confined to just five geographic areas New England Mid Atlantic East North Central South Atlantic and West North Central 83 246 247 CDC implemented national surveillance of Lyme disease cases in 1991 Since then reporting criteria has been modified multiple times 248 The 2022 surveillance case definition classifies cases as confirmed probable and suspect 249 CDC emphasizes that while surveillance data has limitations it is useful due to uniformity simplicity and timeliness While cases are under reported in high incidence areas over reporting is likely in low incidence areas Additionally surveillance cases are reported by county of residence and not where an infection was necessarily contracted 250 83 The number of reported cases of the disease has been increasing as are endemic regions in North America For example B burgdorferi sensu lato was previously thought to be hindered in its ability to be maintained in an enzootic cycle in California because it was assumed the large lizard population would dilute the number of people affected by B burgdorferi in local tick populations this has since been brought into question as some evidence has suggested lizards can become infected 251 Except for one study in Europe 252 much of the data implicating lizards is based on DNA detection of the spirochete and has not demonstrated that lizards are able to infect ticks feeding upon them 251 253 254 255 As some experiments suggest lizards are refractory to infection with Borrelia it appears likely their involvement in the enzootic cycle is more complex and species specific 77 While B burgdorferi is most associated with ticks hosted by white tailed deer and white footed mice Borrelia afzelii is most frequently detected in rodent feeding vector ticks and Borrelia garinii and Borrelia valaisiana appear to be associated with birds Both rodents and birds are competent reservoir hosts for B burgdorferi sensu stricto The resistance of a genospecies of Lyme disease spirochetes to the bacteriolytic activities of the alternative complement pathway of various host species may determine its reservoir host association 252 Several similar but apparently distinct conditions may exist caused by various species or subspecies of Borrelia in North America A regionally restricted condition that may be related to Borrelia infection is southern tick associated rash illness STARI also known as Masters disease Amblyomma americanum known commonly as the lone star tick is recognized as the primary vector for STARI In some parts of the geographical distribution of STARI Lyme disease is quite rare e g Arkansas so people in these regions experiencing Lyme like symptoms especially if they follow a bite from a lone star tick should consider STARI as a possibility It is generally a milder condition than Lyme and typically responds well to antibiotic treatment 256 In recent years there have been 5 to 10 cases a year of a disease similar to Lyme occurring in Montana It occurs primarily in pockets along the Yellowstone River in central Montana People have developed a red bull s eye rash around a tick bite followed by weeks of fatigue and a fever 247 Lyme disease effects are comparable among males and females A wide range of age groups is affected though the number of cases is highest among 10 to 19 year olds For unknown reasons Lyme disease is seven times more common among Asians 257 South America edit In Brazil a Lyme like disease known as Baggio Yoshinari syndrome was identified caused by microorganisms that do not belong to the B burgdorferi sensu lato complex and transmitted by ticks of the Amblyomma and Rhipicephalus genera 258 The first reported case of BYS in Brazil was made in 1992 in Cotia Sao Paulo 259 Etymology editLyme disease was diagnosed as a separate condition for the first time in 1975 in Lyme Connecticut 260 History editThe evolutionary history of Borrelia burgdorferi genetics has been the subject of recent studies One study has found that prior to the reforestation that accompanied post colonial farm abandonment in New England and the wholesale migration into the mid west that occurred during the early 19th century Lyme disease was present for thousands of years in America and had spread along with its tick hosts from the Northeast to the Midwest 261 John Josselyn who visited New England in 1638 and again from 1663 to 1670 wrote there be infinite numbers of ticks hanging upon the bushes in summertime that will cleave to man s garments and creep into his breeches eating themselves in a short time into the very flesh of a man I have seen the stockings of those that have gone through the woods covered with them 262 This is also confirmed by the writings of Peter Kalm a Swedish botanist who was sent to America by Linnaeus and who found the forests of New York abound with ticks when he visited in 1749 When Kalm s journey was retraced 100 years later the forests were gone and the Lyme bacterium had probably become isolated to a few pockets along the northeast coast Wisconsin and Minnesota 263 Perhaps the first detailed description of what is now known as Lyme disease appeared in the writings of John Walker after a visit to the island of Jura Deer Island off the west coast of Scotland in 1764 264 He gives a good description both of the symptoms of Lyme disease with exquisite pain in the interior parts of the limbs and of the tick vector itself which he describes as a worm with a body which is of a reddish color and of a compressed shape with a row of feet on each side that penetrates the skin Many people from this area of Great Britain emigrated to North America between 1717 and the end of the 18th century citation needed The examination of preserved museum specimens has found Borrelia DNA in an infected Ixodes ricinus tick from Germany that dates back to 1884 and from an infected mouse from Cape Cod that died in 1894 263 The 2010 autopsy of Otzi the Iceman a 5 300 year old mummy revealed the presence of the DNA sequence of Borrelia burgdorferi making him the earliest known human with Lyme disease 265 The early European studies of what is now known as Lyme disease described its skin manifestations The first study dates to 1883 in Breslau Germany now Wroclaw Poland where physician Alfred Buchwald described a man who for 16 years had had a degenerative skin disorder now known as acrodermatitis chronica atrophicans 266 At a 1909 research conference Swedish dermatologist Arvid Afzelius presented a study about an expanding ring like lesion he had observed in an older woman following the bite of a sheep tick He named the lesion erythema migrans 266 The skin condition now known as borrelial lymphocytoma was first described in 1911 267 The modern history of medical understanding of the disease including its cause diagnosis and treatment has been difficult 268 Neurological problems following tick bites were recognized starting in the 1920s French physicians Garin and Bujadoux described a farmer with a painful sensory radiculitis accompanied by mild meningitis following a tick bite A large ring shaped rash was also noted although the doctors did not relate it to the meningoradiculitis In 1930 the Swedish dermatologist Sven Hellerstrom was the first to propose EM and neurological symptoms following a tick bite were related 269 In the 1940s German neurologist Alfred Bannwarth described several cases of chronic lymphocytic meningitis and polyradiculoneuritis some of which were accompanied by erythematous skin lesions Carl Lennhoff who worked at the Karolinska Institute in Sweden believed many skin conditions were caused by spirochetes In 1948 he used a special stain to microscopically observe what he believed were spirochetes in various types of skin lesions including EM 270 Although his conclusions were later shown to be erroneous interest in the study of spirochetes was sparked In 1949 Nils Thyresson who also worked at the Karolinska Institute was the first to treat ACA with penicillin 271 In the 1950s the relationship among tick bite lymphocytoma EM and Bannwarth s syndrome was recognized throughout Europe leading to the widespread use of penicillin for treatment in Europe 272 273 In 1970 a dermatologist in Wisconsin named Rudolph Scrimenti recognized an EM lesion in a person after recalling a paper by Hellerstrom that had been reprinted in an American science journal in 1950 This was the first documented case of EM in the United States Based on the European literature he treated the person with penicillin 274 The full syndrome now known as Lyme disease was not recognized until a cluster of cases originally thought to be juvenile rheumatoid arthritis was identified in three towns in southeastern Connecticut in 1975 including the towns Lyme and Old Lyme which gave the disease its popular name 275 This was investigated by physicians David Snydman and Allen Steere of the Epidemic Intelligence Service and by others from Yale University including Stephen Malawista who is credited as a co discover of the disease 276 The recognition that the people in the United States had EM led to the recognition that Lyme arthritis was one manifestation of the same tick borne condition known in Europe 277 Before 1976 the elements of B burgdorferi sensu lato infection were called or known as tick borne meningopolyneuritis Garin Bujadoux syndrome Bannwarth syndrome Afzelius s disease 278 Montauk Knee or sheep tick fever Since 1976 the disease is most often referred to as Lyme disease 279 280 Lyme borreliosis or simply borreliosis 281 282 In 1980 Steere et al began to test antibiotic regimens in adults with Lyme disease 283 In the same year New York State Health Dept epidemiologist Jorge Benach provided Willy Burgdorfer a researcher at the Rocky Mountain Biological Laboratory with collections of I dammini scapularis from Shelter Island New York a known Lyme endemic area as part of an ongoing investigation of Rocky Mountain spotted fever In examining the ticks for rickettsiae Burgdorfer noticed poorly stained rather long irregularly coiled spirochetes Further examination revealed spirochetes in 60 of the ticks Burgdorfer credited his familiarity with the European literature for his realization that the spirochetes might be the long sought cause of ECM and Lyme disease Benach supplied him with more ticks from Shelter Island and sera from people diagnosed with Lyme disease University of Texas Health Science Center researcher Alan Barbour offered his expertise to culture and immunochemically characterize the organism Burgdorfer subsequently confirmed his discovery by isolating from people with Lyme disease spirochetes identical to those found in ticks 284 In June 1982 he published his findings in Science and the spirochete was named Borrelia burgdorferi in his honor 285 After the identification of B burgdorferi as the causative agent of Lyme disease antibiotics were selected for testing guided by in vitro antibiotic sensitivities including tetracycline antibiotics amoxicillin cefuroxime axetil intravenous and intramuscular penicillin and intravenous ceftriaxone 286 287 The mechanism of tick transmission was also the subject of much discussion B burgdorferi spirochetes were identified in tick saliva in 1987 confirming the hypothesis that transmission occurred via tick salivary glands 288 Society culture amp controversy editLandscape changes amp urbanizationUrbanization and other anthropogenic factors can be implicated in the spread of Lyme disease to humans In many areas expansion of suburban neighborhoods has led to gradual deforestation of surrounding wooded areas and increased border contact between humans and tick dense areas Human expansion has also resulted in a reduction of predators that hunt deer as well as mice chipmunks and other small rodents the primary reservoirs for Lyme disease As a consequence of increased human contact with host and vector the likelihood of transmission of the disease has greatly increased 289 290 Researchers are investigating possible links between global warming and the spread of vector borne diseases including Lyme disease 291 The dilution effectGiven these habitat host dynamics some researchers have begun to postulate whether the dilution effect could mitigate the spread of Lyme disease 289 The dilution effect is a hypothesis that predicts that an increase in host biodiversity will result in a decrease in the number of vectors infected with B burgdorferi 289 Scientific research has shown that nymphal infection prevalence NIP decreases as the number of host species increases supporting the dilution effect 289 That said these findings should not be misinterpreted to suggest that there is a direct relationship between decreased NIP and decreased epidemiological risk as this has yet to be proven 289 Additionally it is important to note that thus far the dilution effect is only supported in the Northeastern United States and has been disproved in other parts of the world that also experience high Lyme disease incidence rates 292 Chronic Lyme disease Main article Chronic Lyme disease The term chronic Lyme disease is controversial and not recognized in the medical literature 293 and most medical authorities advise against long term antibiotic treatment for Lyme disease 35 117 294 Studies have shown that most people diagnosed with chronic Lyme disease either have no objective evidence of previous or current infection with B burgdorferi or are people who should be classified as having post treatment Lyme disease syndrome PTLDS which is defined as continuing or relapsing non specific symptoms such as fatigue musculoskeletal pain and cognitive complaints in a person previously treated for Lyme disease 295 The 2008 documentary Under Our Skin is known for promoting controversial and unrecognized theories about chronic Lyme disease 296 Other animals editDogs edit Prevention of Lyme disease is an important step in keeping dogs safe in endemic areas Prevention education and a number of preventive measures are available First for dog owners who live near or who often frequent tick infested areas routine vaccinations of their dogs is an important step 297 Another crucial preventive measure is the use of persistent acaricides such as topical repellents or pesticides that contain triazapentadienes Amitraz phenylpyrazoles Fipronil or permethrin pyrethroids 298 These acaricides target primarily the adult stages of Lyme carrying ticks and reduce the number of reproductively active ticks in the environment 297 Formulations of these ingredients are available in a variety of topical forms including spot ons sprays powders impregnated collars solutions and shampoos 298 Examination of a dog for ticks after being in a tick infested area is an important precautionary measure to take in the prevention of Lyme disease Key spots to examine include the head neck and ears 299 In dogs a serious long term prognosis may result in glomerular disease 300 which is a category of kidney damage that may cause chronic kidney disease 181 Dogs may also experience chronic joint disease if the disease is left untreated However the majority of cases of Lyme disease in dogs result in complete recovery with and sometimes without treatment with antibiotics 301 verification needed In rare cases Lyme disease can be fatal to both humans and dogs 302 Cats edit Unlike dogs it is very rare for a cat to be infected with Lyme disease However cats are nevertheless capable of being infected with B burgdorferi following a bite from an infected tick Cats who are infected with Lyme Disease may show symptoms including but not limited to lameness fatigue or loss of appetite 303 In two cases the infected cats experienced cardiac irregularities similar to symptoms of Lyme in both dogs and humans 304 However cats who are infected with Lyme disease are likely to be asymptomatic and show no noticeable signs of the disease 304 303 Cats with Lyme are often treated with antibiotics much like other animals In some cases additional treatment or therapy may be required 303 Horses edit While Lyme disease can occur in horses not every infection with B burgdorferi is associated with symptoms 305 Especially detection of specific antibodies against B burgdorferi alone is not sufficient for a diagnosis of equine Lyme disease and unspecific testing for antibodies is not recommended 306 References edit a b c d e f g h i j k Signs and Symptoms of Lyme Disease cdc gov 11 January 2013 Archived from the original on 16 January 2013 Retrieved 2 March 2015 Lyme disease Symptoms and causes Mayo Clinic Retrieved 7 June 2022 a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae Shapiro ED May 2014 Clinical practice Lyme disease PDF The New England Journal of Medicine 370 18 1724 1731 doi 10 1056 NEJMcp1314325 PMC 4487875 PMID 24785207 Archived from the original PDF on 21 August 2016 Retrieved 5 July 2016 a b Lyme Disease Diagnosis and Testing cdc gov 10 January 2013 Archived from the original on 2 March 2015 Retrieved 2 March 2015 a b How many people get Lyme disease Centers for Disease Control and Prevention 13 January 2021 Retrieved 11 January 2023 a b Kugeler KJ Schwartz AM Delorey MJ Mead PS Hinckley AF February 2021 Estimating the Frequency of Lyme Disease Diagnoses United States 2010 2018 Emerging Infectious Diseases 27 2 616 619 doi 10 3201 eid2702 202731 PMC 7853543 PMID 33496229 a b c d Marques AR Strle F Wormser GP August 2021 Comparison of Lyme Disease in the United States and Europe Emerging Infectious Diseases 27 8 2017 2024 doi 10 3201 eid2708 204763 PMC 8314816 PMID 34286689 a b c Wenner M 11 June 2021 Let s Do a Tick Check These pervasive bloodsuckers can give you more than just Lyme disease Here s how to protect yourself Interactive The New York Times Retrieved 19 June 2021 a b Wolcott KA Margos G Fingerle V Becker NS September 2021 Host association of Borrelia burgdorferi sensu lato A review Ticks and Tick Borne Diseases 12 5 101766 doi 10 1016 j ttbdis 2021 101766 PMID 34161868 Aucott J Morrison C Munoz B Rowe PC Schwarzwalder A West SK June 2009 Diagnostic challenges of early Lyme disease lessons from a community case series BMC Infectious Diseases 9 79 doi 10 1186 1471 2334 9 79 PMC 2698836 PMID 19486523 Aucott JN June 2015 Posttreatment Lyme disease syndrome Infectious Disease Clinics of North America 29 2 309 323 doi 10 1016 j idc 2015 02 012 PMID 25999226 Johnson RC 1996 Borrelia In Baron S et al eds Baron s Medical Microbiology 4th ed Univ of Texas Medical Branch ISBN 978 0 9631172 1 2 PMID 21413339 Archived from the original on 7 February 2009 a b Lyme disease transmission cdc gov 11 January 2013 Archived from the original on 3 March 2015 Retrieved 2 March 2015 a b Steere AC Strle F Wormser GP Hu LT Branda JA Hovius JW et al December 2016 Lyme borreliosis Nature Reviews Disease Primers 2 16090 doi 10 1038 nrdp 2016 90 PMC 5539539 PMID 27976670 Lyme borreliosis PDF ECDC Archived PDF from the original on 29 September 2018 Retrieved 29 September 2018 a b Pritt BS Mead PS Johnson DK Neitzel DF Respicio Kingry LB Davis JP et al May 2016 Identification of a novel pathogenic Borrelia species causing Lyme borreliosis with unusually high spirochaetaemia a descriptive study The Lancet Infectious Diseases 16 5 556 564 doi 10 1016 S1473 3099 15 00464 8 PMC 4975683 PMID 26856777 Two step Laboratory Testing Process cdc gov 15 November 2011 Archived from the original on 12 March 2015 Retrieved 2 March 2015 Testing of Ticks cdc gov 4 June 2013 Archived from the original on 19 February 2015 Retrieved 2 March 2015 Although some commercial groups offer testing in general it is not recommended Tick Removal cdc gov 23 June 2014 Archived from the original on 10 March 2015 Retrieved 2 March 2015 a b c Post Treatment Lyme Disease Syndrome cdc gov 1 December 2017 Archived from the original on 27 February 2015 Retrieved 20 June 2018 Regional Disease Vector Ecology Profile Central Europe DIANE Publishing April 2001 p 136 ISBN 978 1 4289 1143 7 Archived from the original on 8 September 2017 Williams C 2007 Infectious disease epidemiology theory and practice 2nd ed Sudbury Mass Jones and Bartlett Publishers p 447 ISBN 978 0 7637 2879 3 Archived from the original on 8 September 2017 Willy Burgdorfer obituary Daily Telegraph 1 December 2014 Archived from the original on 1 December 2014 Retrieved 1 December 2014 Baker PJ November 2010 Chronic Lyme disease in defense of the scientific enterprise FASEB Journal 24 11 4175 4177 doi 10 1096 fj 10 167247 PMID 20631327 S2CID 36141950 Lantos PM June 2015 Chronic Lyme disease Infectious Disease Clinics of North America 29 2 325 340 doi 10 1016 j idc 2015 02 006 PMC 4477530 PMID 25999227 a b Lyme disease vaccine Centers for Disease Control and Prevention 11 August 2022 Retrieved 7 June 2023 a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac Wright WF Riedel DJ Talwani R Gilliam BL June 2012 Diagnosis and management of Lyme disease American Family Physician 85 11 1086 1093 PMID 22962880 Archived from the original on 27 September 2013 a b c d Tibbles CD Edlow JA June 2007 Does this patient have erythema migrans JAMA 297 23 2617 2627 doi 10 1001 jama 297 23 2617 PMID 17579230 a b c Lyme disease rashes and look alikes Lyme Disease Centers for Disease Control and Prevention 21 December 2018 Archived from the original on 2 April 2019 Retrieved 18 April 2019 a b c Lyme disease erythema migrans Lyme disease NICE guideline NG95 National Institute for Health and Care Excellence Archived from the original on 9 May 2019 Retrieved 8 May 2019 Lyme Disease The Lecturio Medical Concept Library Retrieved 9 July 2021 a b Lyme disease at eMedicine Steere AC Sikand VK Schoen RT Nowakowski J August 2003 Asymptomatic infection with Borrelia burgdorferi Clinical Infectious Diseases 37 4 528 532 doi 10 1086 376914 PMID 12905137 primary source a b Biesiada G Czepiel J Lesniak MR Garlicki A Mach T December 2012 Lyme disease review Archives of Medical Science 8 6 978 982 doi 10 5114 aoms 2012 30948 PMC 3542482 PMID 23319969 a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae af ag ah Wormser GP Dattwyler RJ Shapiro ED Halperin JJ Steere AC Klempner MS et al November 2006 The clinical assessment treatment and prevention of lyme disease human granulocytic anaplasmosis and babesiosis clinical practice guidelines by the Infectious Diseases Society of America Clinical Infectious Diseases 43 9 1089 1134 doi 10 1086 508667 PMID 17029130 a b Halperin JJ June 2008 Nervous system Lyme disease Infectious Disease Clinics of North America 22 2 261 74 vi doi 10 1016 j idc 2007 12 009 PMID 18452800 S2CID 10590435 a b c d e f g h i j Mygland A Ljostad U Fingerle V Rupprecht T Schmutzhard E Steiner I January 2010 EFNS guidelines on the diagnosis and management of European Lyme neuroborreliosis PDF European Journal of Neurology 17 1 8 16 doi 10 1111 j 1468 1331 2009 02862 x PMID 19930447 S2CID 14166137 Archived PDF from the original on 8 August 2017 Retrieved 30 April 2019 a b c d e Pachner AR Steere AC July 1984 Neurological findings of Lyme disease The Yale Journal of Biology and Medicine 57 4 481 483 PMC 2590042 PMID 6516450 Moscatello AL Worden DL Nadelman RB Wormser G Lucente F June 1991 Otolaryngologic aspects of Lyme disease The Laryngoscope 101 6 Pt 1 592 595 doi 10 1288 00005537 199106000 00004 PMID 2041438 S2CID 35345898 a b c d e f g h Ogrinc K Lusa L Lotric Furlan S Bogovic P Stupica D Cerar T et al August 2016 Course and Outcome of Early European Lyme Neuroborreliosis Bannwarth Syndrome Clinical and Laboratory Findings Clinical Infectious Diseases 63 3 346 353 doi 10 1093 cid ciw299 PMID 27161773 a b c d e f g h i Garro A Nigrovic LE May 2018 Managing Peripheral Facial Palsy Annals of Emergency Medicine 71 5 618 624 doi 10 1016 j annemergmed 2017 08 039 PMID 29110887 a b Halperin JJ September 2010 Nervous system Lyme disease The Journal of the Royal College of Physicians of Edinburgh 40 3 248 255 doi 10 4997 JRCPE 2010 314 PMID 21127770 a b c d e f g h i j k l m n o Fish AE Pride YB Pinto DS June 2008 Lyme carditis PDF Infectious Disease Clinics of North America 22 2 275 88 vi doi 10 1016 j idc 2007 12 008 PMID 18452801 Archived PDF from the original on 29 August 2017 Retrieved 9 May 2019 a b Stanek G Wormser GP Gray J Strle F February 2012 Lyme borreliosis Lancet 379 9814 461 473 doi 10 1016 S0140 6736 11 60103 7 PMID 21903253 S2CID 31461047 a b Stanek G Strle F June 2008 Lyme disease European perspective Infectious Disease Clinics of North America 22 2 327 39 vii doi 10 1016 j idc 2008 01 001 PMID 18452805 Guardado KE Sergent S January 2022 Pediatric unilateral knee swelling a case report of a complicated differential diagnosis and often overlooked cause Journal of Osteopathic Medicine 122 2 105 109 doi 10 1515 jom 2020 0332 PMID 34989214 S2CID 245705028 Puius YA Kalish RA June 2008 Lyme arthritis pathogenesis clinical presentation and management Infectious Disease Clinics of North America 22 2 289 300 vi vii doi 10 1016 j idc 2007 12 014 PMID 18452802 a b Wormser GP Strle F Shapiro ED Dattwyler RJ Auwaerter PG February 2017 A critical appraisal of the mild axonal peripheral neuropathy of late neurologic Lyme disease Diagnostic Microbiology and Infectious Disease 87 2 163 167 doi 10 1016 j diagmicrobio 2016 11 003 PMC 5924701 PMID 27914746 Bratton RL Whiteside JW Hovan MJ Engle RL Edwards FD May 2008 Diagnosis and treatment of Lyme disease Mayo Clinic Proceedings 83 5 566 571 doi 10 4065 83 5 566 PMID 18452688 Shadick NA Phillips CB Sangha O Logigian EL Kaplan RF Wright EA et al December 1999 Musculoskeletal and neurologic outcomes in patients with previously treated Lyme disease Annals of Internal Medicine 131 12 919 926 doi 10 7326 0003 4819 131 12 199912210 00003 PMID 10610642 S2CID 20746489 Seltzer EG Gerber MA Cartter ML Freudigman K Shapiro ED February 2000 Long term outcomes of persons with Lyme disease JAMA 283 5 609 616 doi 10 1001 jama 283 5 609 PMID 10665700 a b c d e f Lantos PM Rumbaugh J Bockenstedt LK Falck Ytter YT Aguero Rosenfeld ME Auwaerter PG et al January 2021 Clinical Practice Guidelines by the Infectious Diseases Society of America IDSA American Academy of Neurology AAN and American College of Rheumatology ACR 2020 Guidelines for the Prevention Diagnosis and Treatment of Lyme Disease Clinical Infectious Diseases 72 1 e1 e48 doi 10 1093 cid ciaa1215 PMID 33417672 Mullegger RR 2004 Dermatological manifestations of Lyme borreliosis European Journal of Dermatology 14 5 296 309 PMID 15358567 Samuels DS Radolf JD eds 2010 Chapter 6 Structure Function and Biogenesis of the Borrelia Cell Envelope Borrelia Molecular Biology Host Interaction and Pathogenesis Caister Academic Press ISBN 978 1 904455 58 5 Radolf JD Samuels DS eds 2021 Lyme Disease and Relapsing Fever Spirochetes Genomics Molecular Biology Host Interactions and Disease Pathogenesis Caister Academic Press ISBN 978 1 913652 61 6 Cutler SJ Ruzic Sabljic E Potkonjak A February 2017 Emerging borreliae Expanding beyond Lyme borreliosis PDF Molecular and Cellular Probes 31 22 27 doi 10 1016 j mcp 2016 08 003 PMID 27523487 Stanek G Reiter M April 2011 The expanding Lyme Borrelia complex clinical significance of genomic species Clinical Microbiology and Infection 17 4 487 493 doi 10 1111 j 1469 0691 2011 03492 x PMID 21414082 Schneider BS Schriefer ME Dietrich G Dolan MC Morshed MG Zeidner NS October 2008 Borrelia bissettii isolates induce pathology in a murine model of disease Vector Borne and Zoonotic Diseases 8 5 623 633 doi 10 1089 vbz 2007 0251 PMID 18454594 Archived from the original on 29 January 2020 Retrieved 29 January 2020 Rudenko N Golovchenko M Mokracek A Piskunova N Ruzek D Mallatova N Grubhoffer L October 2008 Detection of Borrelia bissettii in cardiac valve tissue of a patient with endocarditis and aortic valve stenosis in the Czech Republic Journal of Clinical Microbiology 46 10 3540 3543 doi 10 1128 JCM 01032 08 PMC 2566110 PMID 18650352 UpToDate www uptodate com Retrieved 10 July 2021 a b Tilly K Rosa PA Stewart PE June 2008 Biology of infection with Borrelia burgdorferi Infectious Disease Clinics of North America 22 2 217 34 v doi 10 1016 j idc 2007 12 013 PMC 2440571 PMID 18452798 a b c d Ryan KJ Ray CG eds 2004 Sherris Medical Microbiology 4th ed McGraw Hill pp 434 37 ISBN 978 0 8385 8529 0 Smith Fiola D Hamilton GC March 2005 2005 Fact Sheet Prevent Tick Bites Prevent Lyme Disease PDF Rutgers University Rutgers Cooperative Research amp Extension New Jersey Agricultural Experiment Station Archived from the original PDF on 26 June 2013 Scribner H 31 May 2019 People are freaking out over the CDC s photo of a muffin with ticks in it Deseret News Utah Archived from the original on 25 January 2020 The U S CDC published a picture of a poppy seed muffin in which some of the poppy seeds were actually ticks a b Lo Re V Occi JL MacGregor RR April 2004 Identifying the vector of Lyme disease American Family Physician 69 8 1935 1937 PMID 15117014 Westport Weston Health District 2004 Archived from the original on 29 September 2013 Retrieved 26 September 2013 a b Lyme Disease Data and surveillance Lyme Disease Centers for Disease Control and Prevention 5 February 2019 Archived from the original on 13 April 2019 Retrieved 12 April 2019 Hovius JW van Dam AP Fikrig E September 2007 Tick host pathogen interactions in Lyme borreliosis PDF Trends in Parasitology 23 9 434 438 doi 10 1016 j pt 2007 07 001 PMID 17656156 Archived PDF from the original on 16 February 2019 Retrieved 9 September 2019 a b c Steere AC Coburn J Glickstein L April 2004 The emergence of Lyme disease The Journal of Clinical Investigation 113 8 1093 1101 doi 10 1172 JCI21681 PMC 385417 PMID 15085185 Hu L 13 May 2019 Patient education What to do after a tick bite to prevent Lyme disease Beyond the Basics www uptodate com UpToDate Archived from the original on 1 January 2020 Retrieved 30 January 2020 de Mik EL van Pelt W Docters van Leeuwen BD van der Veen A Schellekens JF Borgdorff MW April 1997 The geographical distribution of tick bites and erythema migrans in general practice in The Netherlands International Journal of Epidemiology 26 2 451 457 doi 10 1093 ije 26 2 451 PMID 9169184 Sun Y Xu R 2003 Ability of Ixodes persulcatus Haemaphysalis concinna and Dermacentor silvarum ticks to acquire and transstadially transmit Borrelia garinii Experimental amp Applied Acarology 31 1 2 151 160 doi 10 1023 B APPA 0000005119 30172 43 PMID 14756409 S2CID 19214181 Ledin KE Zeidner NS Ribeiro JM Biggerstaff BJ Dolan MC Dietrich G et al March 2005 Borreliacidal activity of saliva of the tick Amblyomma americanum Medical and Veterinary Entomology 19 1 90 95 doi 10 1111 j 0269 283X 2005 00546 x PMID 15752182 S2CID 270178 Masters EJ Grigery CN Masters RW June 2008 STARI or Masters disease Lone Star tick vectored Lyme like illness Infectious Disease Clinics of North America 22 2 361 76 viii doi 10 1016 j idc 2007 12 010 PMID 18452807 Clark K November 2004 Borrelia species in host seeking ticks and small mammals in northern Florida Journal of Clinical Microbiology 42 11 5076 5086 doi 10 1128 JCM 42 11 5076 5086 2004 PMC 525154 PMID 15528699 Eisen L Eisen RJ Lane RS December 2004 The roles of birds lizards and rodents as hosts for the western black legged tick Ixodes pacificus Journal of Vector Ecology 29 2 295 308 PMID 15709249 a b Lane RS Mun J Eisen L Eisen RJ August 2006 Refractoriness of the western fence lizard Sceloporus occidentalis to the Lyme disease group spirochete Borrelia bissettii The Journal of Parasitology 92 4 691 696 doi 10 1645 GE 738R1 1 PMID 16995383 S2CID 24200639 Walsh CA Mayer EW Baxi LV January 2007 Lyme disease in pregnancy case report and review of the literature Obstetrical amp Gynecological Survey 62 1 41 50 doi 10 1097 01 ogx 0000251024 43400 9a PMID 17176487 S2CID 31929160 Dotters Katz SK Kuller J Heine RP September 2013 Arthropod borne bacterial diseases in pregnancy Obstetrical amp Gynecological Survey 68 9 635 649 doi 10 1097 OGX 0b013e3182a5ed46 PMID 25102120 S2CID 26801402 Lakos A Solymosi N June 2010 Maternal Lyme borreliosis and pregnancy outcome International Journal of Infectious Diseases 14 6 e494 e498 doi 10 1016 j ijid 2009 07 019 PMID 19926325 Steere AC August 1989 Lyme disease The New England Journal of Medicine 321 9 586 596 doi 10 1056 NEJM198908313210906 PMID 2668764 Tick Attachment and Tickborne Diseases www maine gov Archived from the original on 1 October 2018 Retrieved 1 October 2018 a b c CDC Lyme disease FAQ Centers for Disease Control and Prevention 8 January 2021 Retrieved 11 January 2023 Swanson SJ Neitzel D Reed KD Belongia EA October 2006 Coinfections acquired from ixodes ticks Clinical Microbiology Reviews 19 4 708 727 doi 10 1128 CMR 00011 06 PMC 1592693 PMID 17041141 Lindgren E Gustafson R July 2001 Tick borne encephalitis in Sweden and climate change Lancet 358 9275 16 18 doi 10 1016 S0140 6736 00 05250 8 PMID 11454371 S2CID 336323 Pachner AR Steiner I June 2007 Lyme neuroborreliosis infection immunity and inflammation The Lancet Neurology 6 6 544 552 doi 10 1016 S1474 4422 07 70128 X PMID 17509489 S2CID 11600373 Does Borrelia burgdorferi produce a neurotoxin American Lyme Disease Foundation Retrieved 30 July 2022 Auwaerter PG Aucott J Dumler JS January 2004 Lyme borreliosis Lyme disease molecular and cellular pathobiology and prospects for prevention diagnosis and treatment Expert Reviews in Molecular Medicine 6 2 1 22 doi 10 1017 S1462399404007276 PMID 14987414 S2CID 24311718 Fikrig E Narasimhan S April 2006 Borrelia burgdorferi traveling incognito Microbes and Infection 8 5 1390 1399 doi 10 1016 j micinf 2005 12 022 PMID 16698304 Xu Q Seemanapalli SV Reif KE Brown CR Liang FT April 2007 Increasing the recruitment of neutrophils to the site of infection dramatically attenuates Borrelia burgdorferi infectivity Journal of Immunology 178 8 5109 5115 doi 10 4049 jimmunol 178 8 5109 PMID 17404293 Coleman JL Gebbia JA Piesman J Degen JL Bugge TH Benach JL June 1997 Plasminogen is required for efficient dissemination of B burgdorferi in ticks and for enhancement of spirochetemia in mice Cell 89 7 1111 1119 doi 10 1016 S0092 8674 00 80298 6 PMID 9215633 S2CID 18750257 Steere AC July 2001 Lyme disease The New England Journal of Medicine 345 2 115 125 doi 10 1056 NEJM200107123450207 PMID 11450660 Rupprecht TA Koedel U Fingerle V Pfister HW 2008 The pathogenesis of lyme neuroborreliosis from infection to inflammation Molecular Medicine 14 3 4 205 212 doi 10 2119 2007 00091 Rupprecht PMC 2148032 PMID 18097481 Archived from the original on 3 April 2015 Cabello FC Godfrey HP Newman SA August 2007 Hidden in plain sight Borrelia burgdorferi and the extracellular matrix Trends in Microbiology 15 8 350 354 doi 10 1016 j tim 2007 06 003 PMID 17600717 Archived from the original on 7 July 2019 Retrieved 5 July 2019 Ramesh G Alvarez AL Roberts ED Dennis VA Lasater BL Alvarez X Philipp MT September 2003 Pathogenesis of Lyme neuroborreliosis Borrelia burgdorferi lipoproteins induce both proliferation and apoptosis in rhesus monkey astrocytes European Journal of Immunology 33 9 2539 2550 doi 10 1002 eji 200323872 PMID 12938230 S2CID 25448372 Halperin JJ Heyes MP January 1992 Neuroactive kynurenines in Lyme borreliosis Neurology 42 1 43 50 doi 10 1212 WNL 42 1 43 PMID 1531156 S2CID 37495839 Fallon BA Keilp J Prohovnik I Heertum RV Mann JJ 2003 Regional cerebral blood flow and cognitive deficits in chronic lyme disease The Journal of Neuropsychiatry and Clinical Neurosciences 15 3 326 332 doi 10 1176 appi neuropsych 15 3 326 PMID 12928508 Gasse T Murr C Meyersbach P Schmutzhard E Wachter H Fuchs D September 1994 Neopterin production and tryptophan degradation in acute Lyme neuroborreliosis versus late Lyme encephalopathy European Journal of Clinical Chemistry and Clinical Biochemistry Submitted manuscript 32 9 685 689 doi 10 1515 cclm 1994 32 9 685 PMID 7865624 S2CID 22214184 Zajkowska J Grygorczuk S Kondrusik M Pancewicz S Hermanowska Szpakowicz T 2006 New aspects of pathogenesis of Lyme borreliosis Przeglad Epidemiologiczny in Polish 60 Suppl 1 167 170 PMID 16909797 Ercolini AM Miller SD January 2009 The role of infections in autoimmune disease Clinical and Experimental Immunology 155 1 1 15 doi 10 1111 j 1365 2249 2008 03834 x PMC 2665673 PMID 19076824 Singh SK Girschick HJ July 2004 Lyme borreliosis from infection to autoimmunity Clinical Microbiology and Infection 10 7 598 614 doi 10 1111 j 1469 0691 2004 00895 x PMID 15214872 Oldstone MB October 1998 Molecular mimicry and immune mediated diseases FASEB Journal 12 13 1255 1265 doi 10 1096 fasebj 12 13 1255 PMC 7164021 PMID 9761770 S2CID 16089636 Raveche ES Schutzer SE Fernandes H Bateman H McCarthy BA Nickell SP Cunningham MW February 2005 Evidence of Borrelia autoimmunity induced component of Lyme carditis and arthritis Journal of Clinical Microbiology 43 2 850 856 doi 10 1128 JCM 43 2 850 856 2005 PMC 548028 PMID 15695691 Weinstein A Britchkov M July 2002 Lyme arthritis and post Lyme disease syndrome Current Opinion in Rheumatology 14 4 383 387 doi 10 1097 00002281 200207000 00008 PMID 12118171 Bolz DD Weis JJ August 2004 Molecular mimicry to Borrelia burgdorferi pathway to autoimmunity Autoimmunity 37 5 387 392 doi 10 1080 08916930410001713098 PMID 15621562 S2CID 43045224 Brown SL Hansen SL Langone JJ July 1999 Role of serology in the diagnosis of Lyme disease JAMA 282 1 62 66 doi 10 1001 jama 282 1 62 PMID 10404913 Hofmann H 1996 Lyme borreliosis problems of serological diagnosis Infection 24 6 470 472 doi 10 1007 BF01713052 PMID 9007597 S2CID 19701481 Wormser GP Masters E Nowakowski J McKenna D Holmgren D Ma K et al October 2005 Prospective clinical evaluation of patients from Missouri and New York with erythema migrans like skin lesions Clinical Infectious Diseases 41 7 958 965 doi 10 1086 432935 PMID 16142659 a b Lyme Disease risk areas map Risk of Lyme disease to Canadians Government of Canada 27 January 2015 Archived from the original on 10 May 2019 Retrieved 8 May 2019 a b c d e f Aucott J Morrison C Munoz B Rowe PC Schwarzwalder A West SK June 2009 Diagnostic challenges of early Lyme disease lessons from a community case series BMC Infectious Diseases 9 79 79 doi 10 1186 1471 2334 9 79 PMC 2698836 PMID 19486523 Lyme disease diagnosis Centers for Disease Control and Prevention CDC 7 October 2008 Archived from the original on 28 August 2009 Retrieved 6 July 2009 a b Lyme disease NICE guideline NG95 National Institute for Health and Care Excellence 11 April 2018 Archived from the original on 11 May 2019 Retrieved 24 May 2019 Wilske B 2005 Epidemiology and diagnosis of Lyme borreliosis Annals of Medicine 37 8 568 579 doi 10 1080 07853890500431934 PMID 16338759 S2CID 30818459 a b c DePietropaolo DL Powers JH Gill JM Foy AJ July 2005 Diagnosis of lyme disease American Family Physician 72 2 297 304 doi 10 1093 cid cir464 PMID 16050454 Are serological tests of any value in the diagnosis of Lyme disease American Lyme Disease Foundation Archived from the original on 19 December 2019 Retrieved 2 December 2019 Coyle PK Schutzer SE Deng Z Krupp LB Belman AL Benach JL Luft BJ November 1995 Detection of Borrelia burgdorferi specific antigen in antibody negative cerebrospinal fluid in neurologic Lyme disease Neurology 45 11 2010 2015 doi 10 1212 WNL 45 11 2010 PMID 7501150 S2CID 23797801 a b c d Halperin JJ Shapiro ED Logigian E Belman AL Dotevall L Wormser GP et al July 2007 Practice parameter treatment of nervous system Lyme disease an evidence based review report of the Quality Standards Subcommittee of the American Academy of Neurology Neurology 69 1 91 102 doi 10 1212 01 wnl 0000265517 66976 28 PMID 17522387 Molloy PJ Persing DH Berardi VP August 2001 False positive results of PCR testing for Lyme disease Clinical Infectious Diseases 33 3 412 413 doi 10 1086 321911 PMID 11438915 a b Aguero Rosenfeld ME Wang G Schwartz I Wormser GP July 2005 Diagnosis of lyme borreliosis Clinical Microbiology Reviews 18 3 484 509 doi 10 1128 CMR 18 3 484 509 2005 PMC 1195970 PMID 16020686 Nocton JJ Dressler F Rutledge BJ Rys PN Persing DH Steere AC January 1994 Detection of Borrelia burgdorferi DNA by polymerase chain reaction in synovial fluid from patients with Lyme arthritis The New England Journal of Medicine 330 4 229 234 doi 10 1056 NEJM199401273300401 PMID 8272083 Hyde FW Johnson RC White TJ Shelburne CE January 1989 Detection of antigens in urine of mice and humans infected with Borrelia burgdorferi etiologic agent of Lyme disease Journal of Clinical Microbiology 27 1 58 61 doi 10 1128 JCM 27 1 58 61 1989 PMC 267232 PMID 2913036 Shafagati N Patanarut A Luchini A Lundberg L Bailey C Petricoin E et al July 2014 The use of Nanotrap particles for biodefense and emerging infectious disease diagnostics Pathogens and Disease 71 2 164 176 doi 10 1111 2049 632X 12136 PMC 7108521 PMID 24449537 Shafagati N Patanarut A Luchini A Lundberg L Bailey C Petricoin E et al July 2014 The use of Nanotrap particles for biodefense and emerging infectious disease diagnostics Pathogens and Disease 71 2 164 176 doi 10 1111 2049 632x 12136 PMC 7108521 PMID 24449537 Burdash N Fernandes J June 1991 Lyme borreliosis detecting the great imitator The Journal of the American Osteopathic Association 91 6 573 4 577 8 doi 10 1515 jom 1991 910610 PMID 1874654 S2CID 245118295 Hildenbrand P Craven DE Jones R Nemeskal P June 2009 Lyme neuroborreliosis manifestations of a rapidly emerging zoonosis AJNR American Journal of Neuroradiology 30 6 1079 1087 doi 10 3174 ajnr A1579 PMC 7051319 PMID 19346313 Westervelt HJ McCaffrey RJ September 2002 Neuropsychological functioning in chronic Lyme disease Neuropsychology Review 12 3 153 177 doi 10 1023 A 1020381913563 PMID 12428915 S2CID 2807397 Dworkin RH Johnson RW Breuer J Gnann JW Levin MJ Backonja M et al January 2007 Recommendations for the management of herpes zoster Clinical Infectious Diseases 44 Suppl 1 S1 26 doi 10 1086 510206 PMID 17143845 S2CID 10894629 Halperin JJ May 2015 Chronic Lyme disease misconceptions and challenges for patient management Infection and Drug Resistance 8 119 128 doi 10 2147 IDR S66739 PMC 4440423 PMID 26028977 a b Tarulli AW Raynor EM May 2007 Lumbosacral radiculopathy PDF Neurologic Clinics 25 2 387 405 doi 10 1016 j ncl 2007 01 008 PMID 17445735 S2CID 15518713 Archived from the original PDF on 20 February 2019 Pachner AR 1989 Neurologic manifestations of Lyme disease the new great imitator Reviews of Infectious Diseases 11 Suppl 6 S1482 S1486 doi 10 1093 clinids 11 Supplement 6 S1482 PMID 2682960 S2CID 3862308 Branda JA Linskey K Kim YA Steere AC Ferraro MJ September 2011 Two tiered antibody testing for Lyme disease with use of 2 enzyme immunoassays a whole cell sonicate enzyme immunoassay followed by a VlsE C6 peptide enzyme immunoassay Clinical Infectious Diseases 53 6 541 547 doi 10 1093 cid cir464 PMID 21865190 a b c d e f g h i j Preventing tick bites on people Lyme Disease Centers for Disease Control and Prevention 8 March 2019 Archived from the original on 15 June 2019 Retrieved 21 May 2019 a b About ticks preventative measures Tick borne diseases Companion Vector Borne Diseases Archived from the original on 7 June 2019 Retrieved 21 May 2019 a b c d e f Eisen L Eisen RJ September 2016 Critical Evaluation of the Linkage Between Tick Based Risk Measures and the Occurrence of Lyme Disease Cases Journal of Medical Entomology 53 5 1050 1062 doi 10 1093 jme tjw092 PMC 5777907 PMID 27330093 Host seeking CVBD Companion Vector Borne Diseases Archived from the original on 16 October 2016 Retrieved 8 December 2016 a b c Tips to prevent tick bytes Insect repellents Environmental Protection Agency 15 July 2013 Archived from the original on 15 April 2019 Retrieved 21 May 2019 a b c d e f g h Protection against Mosquitoes Ticks amp Other Arthropods Travelers Health Centers for Disease Control and Prevention Archived from the original on 9 May 2019 Retrieved 21 May 2019 a b c Miller NJ Rainone EE Dyer MC Gonzalez ML Mather TN March 2011 Tick bite protection with permethrin treated summer weight clothing Journal of Medical Entomology 48 2 327 333 doi 10 1603 me10158 PMID 21485369 a b Eisen L Rose D Prose R Breuner NE Dolan MC Thompson K Connally N October 2017 Bioassays to evaluate non contact spatial repellency contact irritancy and acute toxicity of permethrin treated clothing against nymphal Ixodes scapularis ticks Ticks and Tick Borne Diseases 8 6 837 849 doi 10 1016 j ttbdis 2017 06 010 PMC 5665650 PMID 28754599 Dymond NL Swift IM June 2008 Permethrin toxicity in cats a retrospective study of 20 cases Australian Veterinary Journal 86 6 219 223 doi 10 1111 j 1751 0813 2008 00298 x PMID 18498556 a b Repellents protection against mosquitoes ticks and other arthropods Insect repellents Environmental Protection Agency 9 July 2013 Archived from the original on 20 May 2019 Retrieved 21 May 2019 a b c Nguyen QD Vu MN Hebert AA January 2023 Insect repellents An updated review for the clinician Journal of the American Academy of Dermatology 88 1 123 130 doi 10 1016 j jaad 2018 10 053 PMID 30395919 S2CID 53246686 a b c d e Stafford KC 2007 Tick management handbook rev ed PDF Bulletins Connecticut Agricultural Experiment Station Archived PDF from the original on 3 April 2019 Retrieved 21 May 2019 Piesman J Dolan MC May 2002 Protection against lyme disease spirochete transmission provided by prompt removal of nymphal Ixodes scapularis Acari Ixodidae Journal of Medical Entomology 39 3 509 512 doi 10 1603 0022 2585 39 3 509 PMID 12061448 S2CID 39805040 a b c d e f Tick removal and testing Lyme Disease Centers for Disease Control and Prevention 22 April 2019 Archived from the original on 23 November 2017 Retrieved 21 May 2019 a b Tick Allergy 2014 Archived from the original on 14 May 2015 Retrieved 30 April 2015 Roggla G 2002 Death of unknown origin in alpine medicine Wilderness amp Environmental Medicine 13 2 181 doi 10 1580 1080 6032 2002 013 0181 ltte 2 0 co 2 PMID 12092975 Ghirga G Ghirga P September 2010 Effective tick removal with a fishing line knot Wilderness amp Environmental Medicine 21 3 270 271 doi 10 1016 j wem 2010 04 005 PMID 20832708 a b c d e Preventing tick bites in the yard Lyme Disease Centers for Disease Control and Prevention 8 March 2019 Archived from the original on 15 June 2019 Retrieved 21 May 2019 CDC Lyme Disease NIOSH Workplace Safety and Health Topic www cdc gov 3 August 2017 Archived from the original on 13 November 2015 Retrieved 3 November 2015 Risks Lyme Disease NIOSH CDC www cdc gov 14 November 2018 Archived from the original on 26 April 2019 Retrieved 26 April 2019 Lyme disease The Centers for Disease Control and Prevention 12 October 2016 Archived from the original on 17 June 2017 Retrieved 22 June 2017 nbsp This article incorporates public domain material from websites or documents of the Centers for Disease Control and Prevention a b c Chrobak U 3 February 2022 Lyme and other tick borne diseases are on the rise But why Knowable Magazine doi 10 1146 knowable 020222 1 Retrieved 4 March 2022 Rand PW Lubelczyk C Holman MS Lacombe EH Smith RP July 2004 Abundance of Ixodes scapularis Acari Ixodidae after the complete removal of deer from an isolated offshore island endemic for Lyme Disease Journal of Medical Entomology 41 4 779 784 doi 10 1603 0022 2585 41 4 779 PMID 15311475 S2CID 5853201 Figure 2 Changes in deer density and cases of Lyme disease in Mumford Cove Connecticut 1996 2004 CT DEP data PDF Managing Urban Deer in Connecticut 2nd ed Connecticut Department of Environmental Protection Wildlife Division June 2007 p 4 Archived PDF from the original on 2 June 2013 Stafford KC 2004 Tick Management Handbook PDF Connecticut Agricultural Experiment Station and Connecticut Department of Public Health p 46 Archived PDF from the original on 25 October 2007 Retrieved 21 August 2007 Rochlin I Toledo A June 2020 Emerging tick borne pathogens of public health importance a mini review Journal of Medical Microbiology 69 6 781 791 doi 10 1099 jmm 0 001206 PMC 7451033 PMID 32478654 Rajput ZI Hu SH Chen WJ Arijo AG Xiao CW November 2006 Importance of ticks and their chemical and immunological control in livestock Journal of Zhejiang University Science B 7 11 912 921 doi 10 1631 jzus 2006 B0912 PMC 1635821 PMID 17048307 a b Gern L Estrada Pena A Frandsen F Gray JS Jaenson TG Jongejan F et al March 1998 European reservoir hosts of Borrelia burgdorferi sensu lato Zentralblatt Fur Bakteriologie 287 3 196 204 doi 10 1016 S0934 8840 98 80121 7 PMID 9580423 a b c d Wodecka B Rymaszewska A Skotarczak B April 2014 Host and pathogen DNA identification in blood meals of nymphal Ixodes ricinus ticks from forest parks and rural forests of Poland Experimental amp Applied Acarology 62 4 543 555 doi 10 1007 s10493 013 9763 x PMC 3933768 PMID 24352572 Jaenson TG Talleklint L September 1992 Incompetence of roe deer as reservoirs of the Lyme borreliosis spirochete Journal of Medical Entomology 29 5 813 817 doi 10 1093 jmedent 29 5 813 PMID 1404260 Talleklint L Jaenson TG November 1994 Transmission of Borrelia burgdorferi s l from mammal reservoirs to the primary vector of Lyme borreliosis Ixodes ricinus Acari Ixodidae in Sweden Journal of Medical Entomology 31 6 880 886 doi 10 1093 jmedent 31 6 880 PMID 7815401 Overzier E Pfister K Herb I Mahling M Bock G Silaghi C June 2013 Detection of tick borne pathogens in roe deer Capreolus capreolus in questing ticks Ixodes ricinus and in ticks infesting roe deer in southern Germany Ticks and Tick Borne Diseases 4 4 320 328 doi 10 1016 j ttbdis 2013 01 004 PMID 23571115 permanent dead link a b O Bier NS Hatke AL Camire AC Marconi RT 2021 Human and Veterinary Vaccines for Lyme Disease Current Issues in Molecular Biology 42 191 222 doi 10 21775 cimb 042 191 PMC 7946718 PMID 33289681 Poland GA Jacobson RM March 2001 The prevention of Lyme disease with vaccine Vaccine 19 17 19 2303 2308 doi 10 1016 S0264 410X 00 00520 X PMID 11257352 a b Poland GA February 2011 Vaccines against Lyme disease What happened and what lessons can we learn Clinical Infectious Diseases 52 Suppl 3 s253 s258 doi 10 1093 cid ciq116 PMID 21217172 Rowe C 13 June 1999 Lukewarm Response To New Lyme Vaccine The New York Times Archived from the original on 10 February 2012 Retrieved 11 July 2008 a b c Abbott A February 2006 Lyme disease uphill struggle Nature 439 7076 524 525 doi 10 1038 439524a PMID 16452949 S2CID 4315588 Sole Lyme Vaccine Is Pulled Off Market The New York Times 28 February 2002 Archived from the original on 30 August 2010 Retrieved 11 July 2008 a b Nigrovic LE Thompson KM January 2007 The Lyme vaccine a cautionary tale Epidemiology and Infection 135 1 1 8 doi 10 1017 S0950268806007096 PMC 2870557 PMID 16893489 When a vaccine is safe Nature 439 7076 509 February 2006 Bibcode 2006Natur 439Q 509 doi 10 1038 439509a PMID 16452935 Aronowitz RA June 2012 The rise and fall of the lyme disease vaccines a cautionary tale for risk interventions in American medicine and public health The Milbank Quarterly 90 2 250 277 doi 10 1111 j 1468 0009 2012 00663 x PMC 3460208 PMID 22709388 Anti vaxxers stopped the last Lyme disease vaccine The FDA has just fast tracked a new one Newsweek 25 July 2017 Archived from the original on 25 December 2017 Retrieved 25 December 2017 Brooks D 6 August 2018 A Lyme vaccine for humans is getting closer says French biotech firm Concord Monitor Retrieved 20 July 2021 Taylor NP 4 February 2022 Pfizer s 130M Lyme disease vaccine advances to phase 3 after 3rd shot shown to increase antibodies Fierce Biotech Retrieved 5 June 2022 Neergaard L Lum S 8 August 2022 Major test of first possible Lyme vaccine in 20 years begins Associated Press Retrieved 11 August 2022 a b An Efficacy Safety Tolerability Immunogenicity and Lot Consistency Clinical Trial of a 6 Valent OspA Based Lyme Disease Vaccine VLA15 VALOR ClinicalTrials gov National Institute of Health 28 July 2022 Retrieved 11 August 2022 Klein A 17 November 2021 An mRNA vaccine that causes a red itchy skin rash in response to bites by ticks may allow them to be removed before they transmit Lyme disease causing bacteria New Scientist Hathaway B Novel Lyme vaccine shows promise Yale University Retrieved 13 December 2021 Compared to non immunized guinea pigs vaccinated animals exposed to infected ticks quickly developed redness at the tick bite site None of the immunized animals developed Lyme disease if ticks were removed when redness developed In contrast about half of the control group became infected with B burgdorferi after tick removal When a single infected tick was attached to immunized guinea pigs and not removed none of vaccinated animals were infected compared to 60 percent of control animals However protection waned in immunized guinea pigs if three ticks remained attached to the animal Ticks in immunized animals were unable to feed aggressively and dislodged more quickly than those on guinea pigs in the control group Sajid A Matias J Arora G Kurokawa C DePonte K Tang X et al November 2021 mRNA vaccination induces tick resistance and prevents transmission of the Lyme disease agent Science Translational Medicine 13 620 eabj9827 doi 10 1126 scitranslmed abj9827 PMID 34788080 S2CID 244375227 a b Brooks WC Lyme Disease Veterinary Information Network Archived from the original on 4 April 2015 Retrieved 10 February 2012 Pace EJ O Reilly M May 2020 Tickborne Diseases Diagnosis and Management American Family Physician 101 9 530 540 PMID 32352736 Berende A ter Hofstede HJ Vos FJ van Middendorp H Vogelaar ML Tromp M et al March 2016 Randomized Trial of Longer Term Therapy for Symptoms Attributed to Lyme Disease The New England Journal of Medicine 374 13 1209 1220 doi 10 1056 NEJMoa1505425 PMID 27028911 Stephenson M 4 October 2012 OTC Drops Telling the Tears Apart Review of Ophtalmology Jobson Medical Information LLC Archived from the original on 17 April 2019 Retrieved 16 April 2019 a b c d e Arvikar SL Steere AC June 2015 Diagnosis and treatment of Lyme arthritis Infectious Disease Clinics of North America 29 2 269 280 doi 10 1016 j idc 2015 02 004 PMC 4443866 PMID 25999223 a b c Wormser GP Ramanathan R Nowakowski J McKenna D Holmgren D Visintainer P et al May 2003 Duration of antibiotic therapy for early Lyme disease A randomized double blind placebo controlled trial Annals of Internal Medicine 138 9 697 704 doi 10 7326 0003 4819 138 9 200305060 00005 PMID 12729423 S2CID 3083800 Stupica D Lusa L Ruzic Sabljic E Cerar T Strle F August 2012 Treatment of erythema migrans with doxycycline for 10 days versus 15 days Clinical Infectious Diseases 55 3 343 350 doi 10 1093 cid cis402 PMID 22523260 a b Dattwyler RJ Luft BJ Kunkel MJ Finkel MF Wormser GP Rush TJ et al July 1997 Ceftriaxone compared with doxycycline for the treatment of acute disseminated Lyme disease The New England Journal of Medicine 337 5 289 294 doi 10 1056 NEJM199707313370501 PMID 9233865 Vrethem M Hellblom L Widlund M Ahl M Danielsson O Ernerudh J Forsberg P October 2002 Chronic symptoms are common in patients with neuroborreliosis a questionnaire follow up study Acta Neurologica Scandinavica 106 4 205 208 doi 10 1034 j 1600 0404 2002 01358 x PMID 12225315 S2CID 42290158 Berglund J Stjernberg L Ornstein K Tykesson Joelsson K Walter H 2002 5 y Follow up study of patients with neuroborreliosis Scandinavian Journal of Infectious Diseases 34 6 421 425 doi 10 1080 00365540110080421 PMID 12160168 S2CID 28306612 a b c Clark JR Carlson RD Sasaki CT Pachner AR Steere AC November 1985 Facial paralysis in Lyme disease The Laryngoscope 95 11 1341 1345 doi 10 1288 00005537 198511000 00009 PMID 4058212 S2CID 85021 a b c Dotevall L Hagberg L March 1999 Successful oral doxycycline treatment of Lyme disease associated facial palsy and meningitis Clinical Infectious Diseases 28 3 569 574 doi 10 1086 515145 PMID 10194080 Kalish RA Kaplan RF Taylor E Jones Woodward L Workman K Steere AC February 2001 Evaluation of study patients with Lyme disease 10 20 year follow up The Journal of Infectious Diseases 183 3 453 460 doi 10 1086 318082 PMID 11133377 a b c Jowett N Gaudin RA Banks CA Hadlock TA June 2017 Steroid use in Lyme disease associated facial palsy is associated with worse long term outcomes The Laryngoscope 127 6 1451 1458 doi 10 1002 lary 26273 PMID 27598389 S2CID 25596860 span, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.