fbpx
Wikipedia

Cockayne syndrome

Cockayne syndrome (CS), also called Neill-Dingwall syndrome, is a rare and fatal autosomal recessive neurodegenerative disorder characterized by growth failure, impaired development of the nervous system, abnormal sensitivity to sunlight (photosensitivity), eye disorders and premature aging.[1][2][3] Failure to thrive and neurological disorders are criteria for diagnosis, while photosensitivity, hearing loss, eye abnormalities, and cavities are other very common features.[3] Problems with any or all of the internal organs are possible. It is associated with a group of disorders called leukodystrophies, which are conditions characterized by degradation of neurological white matter. There are two primary types of Cockayne syndrome: Cockayne syndrome type A (CSA), arising from mutations in the ERCC8 gene, and Cockayne syndrome type B (CSB), resulting from mutations in the ERCC6 gene.[4]

Cockayne syndrome
Other namesNeill-Dingwall syndrome
SpecialtyMedical genetics, neurology, dermatology 

The underlying disorder is a defect in a DNA repair mechanism.[5] Unlike other defects of DNA repair, patients with CS are not predisposed to cancer or infection.[6] Cockayne syndrome is a rare but destructive disease usually resulting in death within the first or second decade of life. The mutation of specific genes in Cockayne syndrome is known, but the widespread effects and its relationship with DNA repair is yet to be well understood.[6]

It is named after English physician Edward Alfred Cockayne (1880–1956) who first described it in 1936 and re-described in 1946.[7] Neill-Dingwall syndrome was named after Mary M. Dingwall and Catherine A. Neill.[7] These two scientists described the case of two brothers with Cockayne syndrome and asserted it was the same disease described by Cockayne. In their article, the two contributed to the signs of the disease through their discovery of calcifications in the brain. They also compared Cockayne syndrome to what is now known as Hutchinson–Gilford progeria syndrome (HGPS), then called progeria, due to the advanced aging that characterizes both disorders.[7]

Types edit

  • CS Type I, the "classic" form, is characterized by normal fetal growth with the onset of abnormalities in the first two years of life. Vision and hearing gradually decline.[8] The central and peripheral nervous systems progressively degenerate until death in the first or second decade of life as a result of serious neurological degradation. Cortical atrophy is less severe in CS Type I.[9]
  • CS Type II is present from birth (congenital) and is much more severe than CS Type 1.[8] It involves very little neurological development after birth. Death usually occurs by age seven. This specific type has also been designated as cerebro-oculo-facio-skeletal (COFS) syndrome or Pena-Shokeir syndrome Type II.[8] COFS syndrome is named so due to the effects it has on the brain, eyes, face, and skeletal system, as the disease frequently causes brain atrophy, cataracts, loss of fat in the face, and osteoporosis. COFS syndrome can be further subdivided into several conditions (COFS types 1, 2, 3 (associated with xeroderma pigmentosum) and 4).[10] Typically patients with this early-onset form of the disorder show more severe brain damage, including reduced myelination of white matter, and more widespread calcifications, including in the cortex and basal ganglia.[9]
  • CS Type III, characterized by late-onset, is typically milder than Types I and II.[8] Often patients with Type III will live into adulthood.
  • Xeroderma pigmentosum-Cockayne syndrome (XP-CS) occurs when an individual also has xeroderma pigmentosum, another DNA repair disease. Some symptoms of each disease are expressed. For instance, freckling and pigment abnormalities characteristic of XP are present. The neurological disorder, spasticity, and underdevelopment of sexual organs characteristic of CS are seen. However, hypomyelination and the facial features of typical CS patients are not present.[11]

Causes edit

If hyperoxia or excess oxygen occurs in the body, the cellular metabolism produces several highly reactive forms of oxygen called free radicals. This can cause oxidative damage to cellular components including the DNA. In normal cells, our body repairs the damaged sections. In the case of this disease, due to subtle defects in transcription, children's genetic machinery for synthesizing proteins needed by the body does not operate at normal capacity. Over time, went this theory, results in developmental failure and death. Every minute, the body pumps 10 to 20 liters of oxygen through the blood, carrying it to billions of cells in our bodies. In its normal molecular form, oxygen is harmless. However, cellular metabolism involving oxygen can generate several highly reactive free radicals. These free radicals can cause oxidative damage to cellular components including the DNA. In an average human cell, several thousand lesions occur in the DNA every day. Many of these lesions result from oxidative damage. Each lesion—a damaged section of DNA—must be snipped out and the DNA repaired to preserve its normal function. Unrepaired DNA can lose its ability to code for proteins. Mutations also can result. These mutations can activate oncogenes or silence tumor suppressor genes. According to research, oxidative damage to active genes is not preferentially repaired, and in the most severe cases, the repair is slowed throughout the whole genome. The resulting accumulation of oxidative damage could impair the normal functions of the DNA and may even result in triggering a program of cell death (apoptosis). The children with this disease do not repair the active genes where oxidative damage occurs. Normally, oxidative damage repair is faster in the active genes (which make up less than five percent of the genome) than in inactive regions of the DNA. The resulting accumulation of oxidative damage could impair the normal functions of the DNA and may even result in triggering a program of cell death (apoptosis).[12]

Genetics edit

 
Cockayne syndrome has an autosomal recessive pattern of inheritance.

Cockayne syndrome is classified genetically as follows:[13]

Type OMIM Gene
A 216400 ERCC8 (also called CSA)
B 133540 ERCC6 (also called CSB)
C 216411 none known
  • Mutations in the ERCC8 (also known as CSA) gene or the ERCC6 (also known as CSB) gene are the cause of Cockayne syndrome type A and type B.[8] Mutations in the ERCC6 gene mutation makes up ~70% of cases. The proteins made by these genes are involved in repairing damaged DNA via the transcription-coupled repair mechanism, particularly the DNA in active genes. DNA damage is caused by ultraviolet rays from sunlight, radiation, or free radicals in the body. A normal cell can repair DNA damage before it accumulates. If either the ERCC6 or the ERCC8 gene is altered (as in Cockayne Syndrome), DNA damage encountered during transcription isn't repaired, causing RNA polymerase to stall at that location, interfering with gene expression. As the unrepaired DNA damage accumulates, progressively more active gene expression is impeded, leading to malfunctioning cells or cell death, which likely contributes to the signs of Cockayne Syndrome such as premature aging and neuronal hypomyelination.[8]

Mechanism edit

In contrast to cells with normal repair capability, CSA and CSB deficient cells are unable to preferentially repair cyclobutane pyrimidine dimers induced by the action of ultraviolet (UV) light on the template strand of actively transcribed genes.[14] This deficiency reflects the loss of ability to perform the DNA repair process known as transcription coupled nucleotide excision repair (TC-NER).[15]

Within the damaged cell, the CSA protein normally localizes to sites of DNA damage, particularly inter-strand cross-links, double-strand breaks and some monoadducts.[16] CSB protein is also normally recruited to DNA damaged sites, and its recruitment is most rapid and robust as follows: interstrand crosslinks > double-strand breaks > monoadducts > oxidative damage.[16] CSB protein forms a complex with another DNA repair protein, SNM1A (DCLRE1A), a 5' – 3' exonuclease, that localizes to inter-strand cross-links in a transcription dependent manner.[17] The accumulation of CSB protein at sites of DNA double-strand breaks occurs in a transcription dependent manner and facilitates homologous recombinational repair of the breaks.[18] During the G0/G1 phase of the cell cycle, DNA damage can trigger a CSB-dependent recombinational repair process that uses an RNA (rather than DNA) template.[19]

The premature aging features of CS are likely due, at least in part, to the deficiencies in DNA repair (see DNA damage theory of aging).[15]

Diagnosis edit

People with this syndrome have smaller than normal head sizes (microcephaly), are of short stature (dwarfism), their eyes appear sunken, and they have an "aged" look. They often have long limbs with joint contractures (inability to relax the muscle at a joint), a hunched back (kyphosis), and they may be very thin (cachetic), due to a loss of subcutaneous fat. Their small chin, large ears, and pointy, thin nose often give an aged appearance.[9] The skin of those with Cockayne syndrome is also frequently affected: hyperpigmentation, varicose or spider veins (telangiectasia),[9] and serious sensitivity to sunlight are common, even in individuals without XP-CS. Often patients with Cockayne Syndrome will severely burn or blister with very little heat exposure. The eyes of patients can be affected in various ways and eye abnormalities are common in CS. Cataracts and cloudiness of the cornea (corneal opacity) are common. The loss of and damage to the nerves of the optic nerve, causing optic atrophy, can occur.[3] Nystagmus, or involuntary eye movement, and pupils that fail to dilate demonstrate a loss of control of voluntary and involuntary muscle movement.[9] A salt and pepper retinal pigmentation is also a typical sign. Diagnosis is determined by a specific test for DNA repair, which measures the recovery of RNA after exposure to UV radiation. Despite being associated with genes involved in nucleotide excision repair (NER), unlike xeroderma pigmentosum, CS is not associated with an increased risk of cancer.[6]

Laboratory Studies edit

In Cockayne syndrome patients, UV-irradiated cells show decreased DNA and RNA synthesis. https://emedicine.medscape.com/article/1115866-workup#c5 Laboratory studies are mainly useful to eliminate other disorders. For example, skeletal radiography, endocrinologic tests, and chromosomal breakage studies can help in excluding disorders included in the differential diagnosis.[citation needed]

Imaging Studies edit

Brain CT scanning in Cockayne syndrome patients may reveal calcifications and cortical atrophy.[15]

Other Tests edit

Prenatal evaluation is possible. Amniotic fluid cell culturing is used to demonstrate that fetal cells are deficient in RNA synthesis after UV irradiation.[citation needed]

Neurology edit

Imaging studies reveal a widespread absence of the myelin sheaths of the neurons in the white matter of the brain and general atrophy of the cortex.[6] Calcifications have also been found in the putamen, an area of the forebrain that regulates movements and aids in some forms of learning,[9] along with the cortex.[7] Additionally, atrophy of the central area of the cerebellum found in patients with Cockayne syndrome could also result in the lack of muscle control, particularly involuntary, and poor posture typically seen.[citation needed]

Treatment edit

There is no permanent cure for this syndrome, although patients can be symptomatically treated. Treatment usually involves physical therapy and minor surgeries to the affected organs, such as cataract removal.[3] Also wearing high-factor sunscreen and protective clothing is recommended because Cockayne Syndrome patients are very sensitive to UV radiation.[20] Optimal nutrition can also help. Genetic counseling for the parents is recommended, as the disorder has a 25% chance of being passed to any future children, and prenatal testing is also a possibility.[3] Another important aspect is the prevention of recurrence of CS in other siblings. Identification of gene defects involved makes it possible to offer genetic counseling and antenatal diagnostic testing to the parents who already have one affected child.[21]

Currently, there are two ongoing projects focused on the development of gene therapy for Cockayne syndrome. The first project, led by the Viljem Julijan Association for Children with Rare Diseases, aims to develop gene therapy specifically for Cockayne syndrome type B.[22] The second project, led by the Riaan Research Initiative, is dedicated to the development of gene therapy for Cockayne syndrome type A.[23]

Prognosis edit

The prognosis for those with Cockayne syndrome is poor, as death typically occurs by the age of 12.[24] The prognosis for Cockayne syndrome varies by disease type. There are three types of Cockayne syndrome according to the severity and onset of the symptoms. However, the differences between the types are not always clear-cut, and some researchers believe the signs and symptoms reflect a spectrum instead of distinct types: Cockayne syndrome Type A (CSA) is marked by normal development until a child is 1 or 2 years old, at which point growth slows and developmental delays are noticed. Symptoms are not apparent until they are 1 year. Life expectancy for type A is approximately 10 to 20 years. These symptoms are seen in CS type 1 children. Cockayne syndrome type B (CSB), also known as "cerebro-oculo-facio-skeletal (COFS) syndrome" (or "Pena-Shokeir syndrome type B"), is the most severe subtype. Symptoms are present at birth and normal brain development stops after birth. The average lifespan for children with type B is up to 7 years of age. These symptoms are seen in CS type 2 children. Cockayne syndrome type C (CSC) appears later in childhood with milder symptoms than the other types and a slower progression of the disorder. People with this type of Cockayne syndrome live into adulthood, with an average lifespan of 40 to 50 years. These symptoms are seen in CS type 3.[15]

Epidemiology edit

Cockayne syndrome is rare worldwide. No racial predilection is reported for Cockayne syndrome. No sexual predilection is described for Cockayne syndrome; the male-to-female ratio is equal. Cockayne syndrome I (CS-A) manifests in childhood. Cockayne syndrome II (CS-B) manifests at birth or in infancy, and it has a worse prognosis.[15]

Recent research edit

The recent research on Jan 2018 mentions different CS features that are seen globally with similarities and differences: CS has an incidence of 1 in 250,000 live births, and a prevalence of approximately 1 per 2.5 million, which is remarkably consistent across various regions globally:[25][26]

Affected parts Clinical features pathology
Face Wizened faceies. Sunken eyes, large ears, thin pointy nose. Small chin. Dental caries, enamel hypoplasia
Skin, hair, nails Photosensitivity. Wrinkled and aged appearing skin. Thin dry hair, prematurely gray hair. Poor venous access.
Central nervous system Microcephaly usually beginning at age 2. Mental retardation with low IQ. Delayed milestones.Tremors, ataxia, seizures, strokes, and subdural hemorrhages. Demyelination – is patchy and segmental– "Metachromatic leukodystrophy". Both oligodendroglia and Schwann cells are affected. Affects cerebral white matter, corpus callosum, brainstem, spinal cord, and peripheral nerves. Neuronal loss at multiple sites, especially the cerebellum. Loss of anterior horn cells due to anterograde and/or retrograde degeneration.

Calcification [55–95%] of the cerebral cortex (especially depths of sulci, basal ganglia, cerebellum, thalamus; also of the arteries, arterioles, and capillaries).

Vascular changes - String vessels, especially in areas of Metachromatic leukodystrophy, calcification in leptomeningeal vessels, accelerated atherosclerosis and arteriolosclerosis. Gliosis is present.

Astrocytes and microglia may show irregular cytoplasm, multiple nuclei. May be seen as a high-intensity white matter on FLAIR MRI sequences signals. No major brain malformations. Relative sparing of the cerebral cortex, slight thinning of cortical ribbon may be seen. Normal gyral pattern with widening of sulci. Lamination, neuronal size, and configuration of the neocortex are preserved. May show parietal occipital dominance. Severe cerebellar atrophy. Loss of Purkinje, granular neurons, and in some cases neurons in the dentate nucleus. Dendrites of Purkinje cells may be grossly deformed ("cactus flowers"), ferruginated dendrites. Dendrites have fewer higher order branches. Purkinje "axonal torpedoes" may be present. Ventricular enlargement, enlarged cisterna magna are seen. Amyloid plaques, neurofibrillary tangles, Hirano bodies not commonly seen, although ubiquitin reactivity of axons present

Hearing and vestibular systems Sensorineural, high tone hearing loss [60–90%]. Mixed conductive and sensorineural hearing loss (44%) Most commonly bilateral, rarely unilateral Loss of hair cells in the cochlea, particularly in the basal turn. Loss of neurons in spiral ganglion. Atrophy of auditory pathways. Scala communis, thickened stapes curare, widened prototympanum. Loss of hair cells in pars superior. Loss of neurons in vestibular ganglion. Collapse of the endolymphatic duct of pars inferior
Vision Corneal opacification.

Cataracts [36–86%]. Usually bilateral, most develop by 4 years of age. Pigmentary retinopathy ("salt and pepper")[43–89%]. Miotic pupils, Optic disk pallor, Enophthalmos, Narrow palpebral fissures.

Patchy loss of melanin pigment granules. Lipofuscin deposition, large pigment laden cells in a perivascular distribution. Retinal pigment epithelial atrophy and hyperplasia. Loss of cells in ganglion and outer nuclear cell layers. Both the outer and inner segments of photoreceptors are affected. Optic nerve atrophy, with partial demyelination, axonal loss, and gliosis
Musculoskeletal system Cachectic dwarfism. Contractures. Kyphosis, scoliosis. Stooped posture. Muscle wasting. Denervation myopathy, disuse atrophy
Cardiovascular system Accelerated hypertension. Aortic root dilatation. Cardiomyopathy. Increased intima medial thickening. Atherosclerosis, arteriosclerosis.
Gastrointestinal system Severe reflux. Abnormal gastrointestinal motility. Many have percutaneous gastrostomy tubes. Hepatomegaly, splenomegaly, elevated liver enzymes. Altered metabolism of drugs -
Renal system Renal failure Renal arteries show changes in advanced atherosclerosis and arteriolosclerosis. Unilateral or hypoplastic kidneys.
Reproductive system - -
Males Micropenis, smaller testicular size -
Females Ovarian atrophy. A successful pregnancy has been reported. -
Endocrine systems Normal secondary sexual characteristics. Normal growth hormone, thyroid-stimulating hormone, calcium levels Normal pituitary gland and thyroid gland
Eccrine systems Decreased production of sweat, tears, saliva -

See also edit

References edit

  1. ^ Bertola; Cao, H; Albano, Lm; Oliveira, Dp; Kok, F; Marques-Dias, Mj; Kim, Ca; Hegele, Ra (2006). "Cockayne syndrome type A: novel mutations in eight typical patients". Journal of Human Genetics. 51 (8): 701–5. doi:10.1007/s10038-006-0011-7. PMID 16865293.
  2. ^ James, William; Berger, Timothy; Elston, Dirk (2005). Andrews' Diseases of the Skin: Clinical Dermatology (10th ed.). Saunders. p. 575. ISBN 978-0-7216-2921-6.
  3. ^ a b c d e Bender M, Potocki L, Metry D. What syndrome is this? Cockayne syndrome. Pediatric Dermatology [serial online]. November 2003;20(6):538-540. Available from: MEDLINE with Full Text, Ipswich, MA. Accessed April 30, 2015.
  4. ^ Laugel, Vincent (2013). "Cockayne syndrome: the expanding clinical and mutational spectrum". Mechanisms of Ageing and Development. 134 (5–6): 161–170. doi:10.1016/j.mad.2013.02.006. ISSN 1872-6216. PMID 23428416. S2CID 19137836.
  5. ^ Hoeijmakers JH (October 2009). "DNA damage, aging, and cancer". N. Engl. J. Med. 361 (15): 1475–85. doi:10.1056/NEJMra0804615. PMID 19812404.
  6. ^ a b c d Nance M, Berry S (1 January 1992). "Cockayne syndrome: review of 140 cases". American Journal of Medical Genetics. 42 (1): 68–84. doi:10.1002/ajmg.1320420115. PMID 1308368.
  7. ^ a b c d Neill CA, Dingwall MM. A Syndrome Resembling Progeria: A Review of Two Cases. Archives of Disease in Childhood. 1950;25(123):213-223.
  8. ^ a b c d e f Cockayne Syndrome. Genetics Home Reference http://ghr.nlm.nih.gov/condition/cockayne-syndrome Published April 28, 2015. Reviewed May 2010. Accessed April 30, 2015.
  9. ^ a b c d e f Javadzadeh M. Cockayne Syndrome. Iran J Child Neurol. Autumn 2014;8;4(Suppl.1):18-19.
  10. ^ Cerebrooculofacioskeletal Syndrome 2. Online Mendelian Inheritance in Man. https://omim.org/entry/610756. Published 2/12/2007.
  11. ^ Laugel V. Cockayne Syndrome. 2000 Dec 28 [Updated 2012 Jun 14]. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2015. Available from: [1]
  12. ^ "Cell Injury and Death". The Lecturio Medical Concept Library. Retrieved 7 July 2021.
  13. ^ Vessoni, Alexandre Teixeira; Guerra, Camila Chaves Coelho; Kajitani, Gustavo Satoru; Nascimento, Livia Luz Souza; Garcia, Camila Carrião Machado (2020). "Cockayne Syndrome: The many challenges and approaches to understand a multifaceted disease". Genetics and Molecular Biology. 43 (1 suppl. 1): e20190085. doi:10.1590/1678-4685-GMB-2019-0085. ISSN 1415-4757. PMC 7250278. PMID 32453336.
  14. ^ van Hoffen A, Natarajan AT, Mayne LV, van Zeeland AA, Mullenders LH, Venema J (1993). "Deficient repair of the transcribed strand of active genes in Cockayne's syndrome cells". Nucleic Acids Res. 21 (25): 5890–5. doi:10.1093/nar/21.25.5890. PMC 310470. PMID 8290349.
  15. ^ a b c d e Karikkineth A, Scheibye-Knudsen M, Fivenson E, Croteau D, Bohr B (January 2017). "Cockayne syndrome: Clinical features, model systems and pathways". Ageing Research Reviews. 33: 3–17. doi:10.1016/j.arr.2016.08.002. PMC 5195851. PMID 27507608.
  16. ^ a b Iyama T, Wilson DM (2016). "Elements That Regulate the DNA Damage Response of Proteins Defective in Cockayne Syndrome". J. Mol. Biol. 428 (1): 62–78. doi:10.1016/j.jmb.2015.11.020. PMC 4738086. PMID 26616585.
  17. ^ Iyama T, Lee SY, Berquist BR, Gileadi O, Bohr VA, Seidman MM, McHugh PJ, Wilson DM (2015). "CSB interacts with SNM1A and promotes DNA interstrand crosslink processing". Nucleic Acids Res. 43 (1): 247–58. doi:10.1093/nar/gku1279. PMC 4288174. PMID 25505141.
  18. ^ Batenburg NL, Thompson EL, Hendrickson EA, Zhu XD (2015). "Cockayne syndrome group B protein regulates DNA double-strand break repair and checkpoint activation". EMBO J. 34 (10): 1399–416. doi:10.15252/embj.201490041. PMC 4491999. PMID 25820262.
  19. ^ Wei L, Nakajima S, Böhm S, Bernstein KA, Shen Z, Tsang M, Levine AS, Lan L (2015). "DNA damage during the G0/G1 phase triggers RNA-templated, Cockayne syndrome B-dependent homologous recombination". Proc. Natl. Acad. Sci. U.S.A. 112 (27): E3495–504. Bibcode:2015PNAS..112E3495W. doi:10.1073/pnas.1507105112. PMC 4500203. PMID 26100862.
  20. ^ Kyllermen, Marten. Cockayne Syndrome. Swedish Information Centre for Rare Diseases. 2012: 4.0. http://www.socialstyrelsen.se/rarediseases/cockaynesyndrome#anchor_17 2015-09-24 at the Wayback Machine
  21. ^ Title: Cockayne Syndrome Authors: Dr Nita R Sutay, Dr Md Ashfaque Tinmaswala, Dr Manjiri Karlekar, Dr Swati Jhahttp://jmscr.igmpublication.org/v3-i7/35%20jmscr.pdf
  22. ^ admin (2020-10-23). "Cockayne syndrome type b | Viljem Julijan Association". Cure for Cockayne Syndrome – type B. Retrieved 2023-07-06.
  23. ^ "Riaan Research Initiative — Cockayne Syndrome". Retrieved 2023-07-06.
  24. ^ "Cockayne syndrome | Genetic and Rare Diseases Information Center (GARD) – an NCATS Program".
  25. ^ Karikkineth, A. C.; Scheibye-Knudsen, M.; Fivenson, E.; Croteau, D. L.; Bohr, V. A. (2016). "Cockayne syndrome: Clinical features, model systems and pathways". Ageing Research Reviews. 33: 3–17. doi:10.1016/j.arr.2016.08.002. PMC 5195851. PMID 27507608.
  26. ^ Kubota, Masaya; Ohta, Sayaka; Ando, Aki; Koyama, Akiko; Terashima, Hiroshi; Kashii, Hirofumi; Hoshino, Hideki; Sugita, Katsuo; Hayashi, Masaharu (June 2015). "Nationwide survey of Cockayne syndrome in Japan: Incidence, clinical course and prognosis: Cockayne syndrome in Japan". Pediatrics International. 57 (3): 339–347. doi:10.1111/ped.12635. PMID 25851792. S2CID 5311897.
  27. ^ "Orphanet: CAMFAK syndrome".

External links edit

  • This article incorporates some public domain text from The U.S. National Library of Medicine

cockayne, syndrome, cocaine, syndrome, redirects, here, substance, cocaine, other, uses, cockayne, disambiguation, also, called, neill, dingwall, syndrome, rare, fatal, autosomal, recessive, neurodegenerative, disorder, characterized, growth, failure, impaired. Cocaine syndrome redirects here For the substance see cocaine For other uses see Cockayne disambiguation Cockayne syndrome CS also called Neill Dingwall syndrome is a rare and fatal autosomal recessive neurodegenerative disorder characterized by growth failure impaired development of the nervous system abnormal sensitivity to sunlight photosensitivity eye disorders and premature aging 1 2 3 Failure to thrive and neurological disorders are criteria for diagnosis while photosensitivity hearing loss eye abnormalities and cavities are other very common features 3 Problems with any or all of the internal organs are possible It is associated with a group of disorders called leukodystrophies which are conditions characterized by degradation of neurological white matter There are two primary types of Cockayne syndrome Cockayne syndrome type A CSA arising from mutations in the ERCC8 gene and Cockayne syndrome type B CSB resulting from mutations in the ERCC6 gene 4 Cockayne syndromeOther namesNeill Dingwall syndromeSpecialtyMedical genetics neurology dermatology The underlying disorder is a defect in a DNA repair mechanism 5 Unlike other defects of DNA repair patients with CS are not predisposed to cancer or infection 6 Cockayne syndrome is a rare but destructive disease usually resulting in death within the first or second decade of life The mutation of specific genes in Cockayne syndrome is known but the widespread effects and its relationship with DNA repair is yet to be well understood 6 It is named after English physician Edward Alfred Cockayne 1880 1956 who first described it in 1936 and re described in 1946 7 Neill Dingwall syndrome was named after Mary M Dingwall and Catherine A Neill 7 These two scientists described the case of two brothers with Cockayne syndrome and asserted it was the same disease described by Cockayne In their article the two contributed to the signs of the disease through their discovery of calcifications in the brain They also compared Cockayne syndrome to what is now known as Hutchinson Gilford progeria syndrome HGPS then called progeria due to the advanced aging that characterizes both disorders 7 Contents 1 Types 2 Causes 3 Genetics 4 Mechanism 5 Diagnosis 5 1 Laboratory Studies 5 2 Imaging Studies 5 3 Other Tests 5 4 Neurology 6 Treatment 7 Prognosis 8 Epidemiology 9 Recent research 10 See also 11 References 12 External linksTypes editCS Type I the classic form is characterized by normal fetal growth with the onset of abnormalities in the first two years of life Vision and hearing gradually decline 8 The central and peripheral nervous systems progressively degenerate until death in the first or second decade of life as a result of serious neurological degradation Cortical atrophy is less severe in CS Type I 9 CS Type II is present from birth congenital and is much more severe than CS Type 1 8 It involves very little neurological development after birth Death usually occurs by age seven This specific type has also been designated as cerebro oculo facio skeletal COFS syndrome or Pena Shokeir syndrome Type II 8 COFS syndrome is named so due to the effects it has on the brain eyes face and skeletal system as the disease frequently causes brain atrophy cataracts loss of fat in the face and osteoporosis COFS syndrome can be further subdivided into several conditions COFS types 1 2 3 associated with xeroderma pigmentosum and 4 10 Typically patients with this early onset form of the disorder show more severe brain damage including reduced myelination of white matter and more widespread calcifications including in the cortex and basal ganglia 9 CS Type III characterized by late onset is typically milder than Types I and II 8 Often patients with Type III will live into adulthood Xeroderma pigmentosum Cockayne syndrome XP CS occurs when an individual also has xeroderma pigmentosum another DNA repair disease Some symptoms of each disease are expressed For instance freckling and pigment abnormalities characteristic of XP are present The neurological disorder spasticity and underdevelopment of sexual organs characteristic of CS are seen However hypomyelination and the facial features of typical CS patients are not present 11 Causes editIf hyperoxia or excess oxygen occurs in the body the cellular metabolism produces several highly reactive forms of oxygen called free radicals This can cause oxidative damage to cellular components including the DNA In normal cells our body repairs the damaged sections In the case of this disease due to subtle defects in transcription children s genetic machinery for synthesizing proteins needed by the body does not operate at normal capacity Over time went this theory results in developmental failure and death Every minute the body pumps 10 to 20 liters of oxygen through the blood carrying it to billions of cells in our bodies In its normal molecular form oxygen is harmless However cellular metabolism involving oxygen can generate several highly reactive free radicals These free radicals can cause oxidative damage to cellular components including the DNA In an average human cell several thousand lesions occur in the DNA every day Many of these lesions result from oxidative damage Each lesion a damaged section of DNA must be snipped out and the DNA repaired to preserve its normal function Unrepaired DNA can lose its ability to code for proteins Mutations also can result These mutations can activate oncogenes or silence tumor suppressor genes According to research oxidative damage to active genes is not preferentially repaired and in the most severe cases the repair is slowed throughout the whole genome The resulting accumulation of oxidative damage could impair the normal functions of the DNA and may even result in triggering a program of cell death apoptosis The children with this disease do not repair the active genes where oxidative damage occurs Normally oxidative damage repair is faster in the active genes which make up less than five percent of the genome than in inactive regions of the DNA The resulting accumulation of oxidative damage could impair the normal functions of the DNA and may even result in triggering a program of cell death apoptosis 12 Genetics edit nbsp Cockayne syndrome has an autosomal recessive pattern of inheritance Cockayne syndrome is classified genetically as follows 13 Type OMIM GeneA 216400 ERCC8 also called CSA B 133540 ERCC6 also called CSB C 216411 none knownMutations in the ERCC8 also known as CSA gene or the ERCC6 also known as CSB gene are the cause of Cockayne syndrome type A and type B 8 Mutations in the ERCC6 gene mutation makes up 70 of cases The proteins made by these genes are involved in repairing damaged DNA via the transcription coupled repair mechanism particularly the DNA in active genes DNA damage is caused by ultraviolet rays from sunlight radiation or free radicals in the body A normal cell can repair DNA damage before it accumulates If either the ERCC6 or the ERCC8 gene is altered as in Cockayne Syndrome DNA damage encountered during transcription isn t repaired causing RNA polymerase to stall at that location interfering with gene expression As the unrepaired DNA damage accumulates progressively more active gene expression is impeded leading to malfunctioning cells or cell death which likely contributes to the signs of Cockayne Syndrome such as premature aging and neuronal hypomyelination 8 Mechanism editIn contrast to cells with normal repair capability CSA and CSB deficient cells are unable to preferentially repair cyclobutane pyrimidine dimers induced by the action of ultraviolet UV light on the template strand of actively transcribed genes 14 This deficiency reflects the loss of ability to perform the DNA repair process known as transcription coupled nucleotide excision repair TC NER 15 Within the damaged cell the CSA protein normally localizes to sites of DNA damage particularly inter strand cross links double strand breaks and some monoadducts 16 CSB protein is also normally recruited to DNA damaged sites and its recruitment is most rapid and robust as follows interstrand crosslinks gt double strand breaks gt monoadducts gt oxidative damage 16 CSB protein forms a complex with another DNA repair protein SNM1A DCLRE1A a 5 3 exonuclease that localizes to inter strand cross links in a transcription dependent manner 17 The accumulation of CSB protein at sites of DNA double strand breaks occurs in a transcription dependent manner and facilitates homologous recombinational repair of the breaks 18 During the G0 G1 phase of the cell cycle DNA damage can trigger a CSB dependent recombinational repair process that uses an RNA rather than DNA template 19 The premature aging features of CS are likely due at least in part to the deficiencies in DNA repair see DNA damage theory of aging 15 Diagnosis editPeople with this syndrome have smaller than normal head sizes microcephaly are of short stature dwarfism their eyes appear sunken and they have an aged look They often have long limbs with joint contractures inability to relax the muscle at a joint a hunched back kyphosis and they may be very thin cachetic due to a loss of subcutaneous fat Their small chin large ears and pointy thin nose often give an aged appearance 9 The skin of those with Cockayne syndrome is also frequently affected hyperpigmentation varicose or spider veins telangiectasia 9 and serious sensitivity to sunlight are common even in individuals without XP CS Often patients with Cockayne Syndrome will severely burn or blister with very little heat exposure The eyes of patients can be affected in various ways and eye abnormalities are common in CS Cataracts and cloudiness of the cornea corneal opacity are common The loss of and damage to the nerves of the optic nerve causing optic atrophy can occur 3 Nystagmus or involuntary eye movement and pupils that fail to dilate demonstrate a loss of control of voluntary and involuntary muscle movement 9 A salt and pepper retinal pigmentation is also a typical sign Diagnosis is determined by a specific test for DNA repair which measures the recovery of RNA after exposure to UV radiation Despite being associated with genes involved in nucleotide excision repair NER unlike xeroderma pigmentosum CS is not associated with an increased risk of cancer 6 Laboratory Studies edit In Cockayne syndrome patients UV irradiated cells show decreased DNA and RNA synthesis https emedicine medscape com article 1115866 workup c5 Laboratory studies are mainly useful to eliminate other disorders For example skeletal radiography endocrinologic tests and chromosomal breakage studies can help in excluding disorders included in the differential diagnosis citation needed Imaging Studies edit Brain CT scanning in Cockayne syndrome patients may reveal calcifications and cortical atrophy 15 Other Tests edit Prenatal evaluation is possible Amniotic fluid cell culturing is used to demonstrate that fetal cells are deficient in RNA synthesis after UV irradiation citation needed Neurology edit Imaging studies reveal a widespread absence of the myelin sheaths of the neurons in the white matter of the brain and general atrophy of the cortex 6 Calcifications have also been found in the putamen an area of the forebrain that regulates movements and aids in some forms of learning 9 along with the cortex 7 Additionally atrophy of the central area of the cerebellum found in patients with Cockayne syndrome could also result in the lack of muscle control particularly involuntary and poor posture typically seen citation needed Treatment editThere is no permanent cure for this syndrome although patients can be symptomatically treated Treatment usually involves physical therapy and minor surgeries to the affected organs such as cataract removal 3 Also wearing high factor sunscreen and protective clothing is recommended because Cockayne Syndrome patients are very sensitive to UV radiation 20 Optimal nutrition can also help Genetic counseling for the parents is recommended as the disorder has a 25 chance of being passed to any future children and prenatal testing is also a possibility 3 Another important aspect is the prevention of recurrence of CS in other siblings Identification of gene defects involved makes it possible to offer genetic counseling and antenatal diagnostic testing to the parents who already have one affected child 21 Currently there are two ongoing projects focused on the development of gene therapy for Cockayne syndrome The first project led by the Viljem Julijan Association for Children with Rare Diseases aims to develop gene therapy specifically for Cockayne syndrome type B 22 The second project led by the Riaan Research Initiative is dedicated to the development of gene therapy for Cockayne syndrome type A 23 Prognosis editThe prognosis for those with Cockayne syndrome is poor as death typically occurs by the age of 12 24 The prognosis for Cockayne syndrome varies by disease type There are three types of Cockayne syndrome according to the severity and onset of the symptoms However the differences between the types are not always clear cut and some researchers believe the signs and symptoms reflect a spectrum instead of distinct types Cockayne syndrome Type A CSA is marked by normal development until a child is 1 or 2 years old at which point growth slows and developmental delays are noticed Symptoms are not apparent until they are 1 year Life expectancy for type A is approximately 10 to 20 years These symptoms are seen in CS type 1 children Cockayne syndrome type B CSB also known as cerebro oculo facio skeletal COFS syndrome or Pena Shokeir syndrome type B is the most severe subtype Symptoms are present at birth and normal brain development stops after birth The average lifespan for children with type B is up to 7 years of age These symptoms are seen in CS type 2 children Cockayne syndrome type C CSC appears later in childhood with milder symptoms than the other types and a slower progression of the disorder People with this type of Cockayne syndrome live into adulthood with an average lifespan of 40 to 50 years These symptoms are seen in CS type 3 15 Epidemiology editCockayne syndrome is rare worldwide No racial predilection is reported for Cockayne syndrome No sexual predilection is described for Cockayne syndrome the male to female ratio is equal Cockayne syndrome I CS A manifests in childhood Cockayne syndrome II CS B manifests at birth or in infancy and it has a worse prognosis 15 Recent research editThe recent research on Jan 2018 mentions different CS features that are seen globally with similarities and differences CS has an incidence of 1 in 250 000 live births and a prevalence of approximately 1 per 2 5 million which is remarkably consistent across various regions globally 25 26 Affected parts Clinical features pathologyFace Wizened faceies Sunken eyes large ears thin pointy nose Small chin Dental caries enamel hypoplasiaSkin hair nails Photosensitivity Wrinkled and aged appearing skin Thin dry hair prematurely gray hair Poor venous access Central nervous system Microcephaly usually beginning at age 2 Mental retardation with low IQ Delayed milestones Tremors ataxia seizures strokes and subdural hemorrhages Demyelination is patchy and segmental Metachromatic leukodystrophy Both oligodendroglia and Schwann cells are affected Affects cerebral white matter corpus callosum brainstem spinal cord and peripheral nerves Neuronal loss at multiple sites especially the cerebellum Loss of anterior horn cells due to anterograde and or retrograde degeneration Calcification 55 95 of the cerebral cortex especially depths of sulci basal ganglia cerebellum thalamus also of the arteries arterioles and capillaries Vascular changes String vessels especially in areas of Metachromatic leukodystrophy calcification in leptomeningeal vessels accelerated atherosclerosis and arteriolosclerosis Gliosis is present Astrocytes and microglia may show irregular cytoplasm multiple nuclei May be seen as a high intensity white matter on FLAIR MRI sequences signals No major brain malformations Relative sparing of the cerebral cortex slight thinning of cortical ribbon may be seen Normal gyral pattern with widening of sulci Lamination neuronal size and configuration of the neocortex are preserved May show parietal occipital dominance Severe cerebellar atrophy Loss of Purkinje granular neurons and in some cases neurons in the dentate nucleus Dendrites of Purkinje cells may be grossly deformed cactus flowers ferruginated dendrites Dendrites have fewer higher order branches Purkinje axonal torpedoes may be present Ventricular enlargement enlarged cisterna magna are seen Amyloid plaques neurofibrillary tangles Hirano bodies not commonly seen although ubiquitin reactivity of axons presentHearing and vestibular systems Sensorineural high tone hearing loss 60 90 Mixed conductive and sensorineural hearing loss 44 Most commonly bilateral rarely unilateral Loss of hair cells in the cochlea particularly in the basal turn Loss of neurons in spiral ganglion Atrophy of auditory pathways Scala communis thickened stapes curare widened prototympanum Loss of hair cells in pars superior Loss of neurons in vestibular ganglion Collapse of the endolymphatic duct of pars inferiorVision Corneal opacification Cataracts 36 86 Usually bilateral most develop by 4 years of age Pigmentary retinopathy salt and pepper 43 89 Miotic pupils Optic disk pallor Enophthalmos Narrow palpebral fissures Patchy loss of melanin pigment granules Lipofuscin deposition large pigment laden cells in a perivascular distribution Retinal pigment epithelial atrophy and hyperplasia Loss of cells in ganglion and outer nuclear cell layers Both the outer and inner segments of photoreceptors are affected Optic nerve atrophy with partial demyelination axonal loss and gliosisMusculoskeletal system Cachectic dwarfism Contractures Kyphosis scoliosis Stooped posture Muscle wasting Denervation myopathy disuse atrophyCardiovascular system Accelerated hypertension Aortic root dilatation Cardiomyopathy Increased intima medial thickening Atherosclerosis arteriosclerosis Gastrointestinal system Severe reflux Abnormal gastrointestinal motility Many have percutaneous gastrostomy tubes Hepatomegaly splenomegaly elevated liver enzymes Altered metabolism of drugs Renal system Renal failure Renal arteries show changes in advanced atherosclerosis and arteriolosclerosis Unilateral or hypoplastic kidneys Reproductive system Males Micropenis smaller testicular size Females Ovarian atrophy A successful pregnancy has been reported Endocrine systems Normal secondary sexual characteristics Normal growth hormone thyroid stimulating hormone calcium levels Normal pituitary gland and thyroid glandEccrine systems Decreased production of sweat tears saliva See also editAccelerated aging disease Biogerontology Degenerative disease Genetic disorder CAMFAK syndrome thought to be a form or subset of Cockayne syndrome 27 References edit Bertola Cao H Albano Lm Oliveira Dp Kok F Marques Dias Mj Kim Ca Hegele Ra 2006 Cockayne syndrome type A novel mutations in eight typical patients Journal of Human Genetics 51 8 701 5 doi 10 1007 s10038 006 0011 7 PMID 16865293 James William Berger Timothy Elston Dirk 2005 Andrews Diseases of the Skin Clinical Dermatology 10th ed Saunders p 575 ISBN 978 0 7216 2921 6 a b c d e Bender M Potocki L Metry D What syndrome is this Cockayne syndrome Pediatric Dermatology serial online November 2003 20 6 538 540 Available from MEDLINE with Full Text Ipswich MA Accessed April 30 2015 Laugel Vincent 2013 Cockayne syndrome the expanding clinical and mutational spectrum Mechanisms of Ageing and Development 134 5 6 161 170 doi 10 1016 j mad 2013 02 006 ISSN 1872 6216 PMID 23428416 S2CID 19137836 Hoeijmakers JH October 2009 DNA damage aging and cancer N Engl J Med 361 15 1475 85 doi 10 1056 NEJMra0804615 PMID 19812404 a b c d Nance M Berry S 1 January 1992 Cockayne syndrome review of 140 cases American Journal of Medical Genetics 42 1 68 84 doi 10 1002 ajmg 1320420115 PMID 1308368 a b c d Neill CA Dingwall MM A Syndrome Resembling Progeria A Review of Two Cases Archives of Disease in Childhood 1950 25 123 213 223 a b c d e f Cockayne Syndrome Genetics Home Reference http ghr nlm nih gov condition cockayne syndrome Published April 28 2015 Reviewed May 2010 Accessed April 30 2015 a b c d e f Javadzadeh M Cockayne Syndrome Iran J Child Neurol Autumn 2014 8 4 Suppl 1 18 19 Cerebrooculofacioskeletal Syndrome 2 Online Mendelian Inheritance in Man https omim org entry 610756 Published 2 12 2007 Laugel V Cockayne Syndrome 2000 Dec 28 Updated 2012 Jun 14 In Pagon RA Adam MP Ardinger HH et al editors GeneReviews Internet Seattle WA University of Washington Seattle 1993 2015 Available from 1 Cell Injury and Death The Lecturio Medical Concept Library Retrieved 7 July 2021 Vessoni Alexandre Teixeira Guerra Camila Chaves Coelho Kajitani Gustavo Satoru Nascimento Livia Luz Souza Garcia Camila Carriao Machado 2020 Cockayne Syndrome The many challenges and approaches to understand a multifaceted disease Genetics and Molecular Biology 43 1 suppl 1 e20190085 doi 10 1590 1678 4685 GMB 2019 0085 ISSN 1415 4757 PMC 7250278 PMID 32453336 van Hoffen A Natarajan AT Mayne LV van Zeeland AA Mullenders LH Venema J 1993 Deficient repair of the transcribed strand of active genes in Cockayne s syndrome cells Nucleic Acids Res 21 25 5890 5 doi 10 1093 nar 21 25 5890 PMC 310470 PMID 8290349 a b c d e Karikkineth A Scheibye Knudsen M Fivenson E Croteau D Bohr B January 2017 Cockayne syndrome Clinical features model systems and pathways Ageing Research Reviews 33 3 17 doi 10 1016 j arr 2016 08 002 PMC 5195851 PMID 27507608 a b Iyama T Wilson DM 2016 Elements That Regulate the DNA Damage Response of Proteins Defective in Cockayne Syndrome J Mol Biol 428 1 62 78 doi 10 1016 j jmb 2015 11 020 PMC 4738086 PMID 26616585 Iyama T Lee SY Berquist BR Gileadi O Bohr VA Seidman MM McHugh PJ Wilson DM 2015 CSB interacts with SNM1A and promotes DNA interstrand crosslink processing Nucleic Acids Res 43 1 247 58 doi 10 1093 nar gku1279 PMC 4288174 PMID 25505141 Batenburg NL Thompson EL Hendrickson EA Zhu XD 2015 Cockayne syndrome group B protein regulates DNA double strand break repair and checkpoint activation EMBO J 34 10 1399 416 doi 10 15252 embj 201490041 PMC 4491999 PMID 25820262 Wei L Nakajima S Bohm S Bernstein KA Shen Z Tsang M Levine AS Lan L 2015 DNA damage during the G0 G1 phase triggers RNA templated Cockayne syndrome B dependent homologous recombination Proc Natl Acad Sci U S A 112 27 E3495 504 Bibcode 2015PNAS 112E3495W doi 10 1073 pnas 1507105112 PMC 4500203 PMID 26100862 Kyllermen Marten Cockayne Syndrome Swedish Information Centre for Rare Diseases 2012 4 0 http www socialstyrelsen se rarediseases cockaynesyndrome anchor 17 Archived 2015 09 24 at the Wayback Machine Title Cockayne Syndrome Authors Dr Nita R Sutay Dr Md Ashfaque Tinmaswala Dr Manjiri Karlekar Dr Swati Jhahttp jmscr igmpublication org v3 i7 35 20jmscr pdf admin 2020 10 23 Cockayne syndrome type b Viljem Julijan Association Cure for Cockayne Syndrome type B Retrieved 2023 07 06 Riaan Research Initiative Cockayne Syndrome Retrieved 2023 07 06 Cockayne syndrome Genetic and Rare Diseases Information Center GARD an NCATS Program Karikkineth A C Scheibye Knudsen M Fivenson E Croteau D L Bohr V A 2016 Cockayne syndrome Clinical features model systems and pathways Ageing Research Reviews 33 3 17 doi 10 1016 j arr 2016 08 002 PMC 5195851 PMID 27507608 Kubota Masaya Ohta Sayaka Ando Aki Koyama Akiko Terashima Hiroshi Kashii Hirofumi Hoshino Hideki Sugita Katsuo Hayashi Masaharu June 2015 Nationwide survey of Cockayne syndrome in Japan Incidence clinical course and prognosis Cockayne syndrome in Japan Pediatrics International 57 3 339 347 doi 10 1111 ped 12635 PMID 25851792 S2CID 5311897 Orphanet CAMFAK syndrome External links editThis article incorporates some public domain text from The U S National Library of Medicine Retrieved from https en wikipedia org w index php title Cockayne syndrome amp oldid 1193072940, 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.