fbpx
Wikipedia

Neurotoxin

Neurotoxins are toxins that are destructive to nerve tissue (causing neurotoxicity).[3] Neurotoxins are an extensive class of exogenous chemical neurological insults[4] that can adversely affect function in both developing and mature nervous tissue.[5] The term can also be used to classify endogenous compounds, which, when abnormally contacted, can prove neurologically toxic.[4] Though neurotoxins are often neurologically destructive, their ability to specifically target neural components is important in the study of nervous systems.[6] Common examples of neurotoxins include lead,[7] ethanol (drinking alcohol),[8] glutamate,[9] nitric oxide,[10] botulinum toxin (e.g. Botox),[11] tetanus toxin,[12] and tetrodotoxin.[6] Some substances such as nitric oxide and glutamate are in fact essential for proper function of the body and only exert neurotoxic effects at excessive concentrations.

Neurotoxins can be found in a number of organisms, including some strains of cyanobacteria,[1] that can be found in algal blooms or washed up on shore in a green scum.[2]

Neurotoxins inhibit neuron control over ion concentrations across the cell membrane,[6] or communication between neurons across a synapse.[13] Local pathology of neurotoxin exposure often includes neuron excitotoxicity or apoptosis[14] but can also include glial cell damage.[15] Macroscopic manifestations of neurotoxin exposure can include widespread central nervous system damage such as intellectual disability,[5] persistent memory impairments,[16] epilepsy, and dementia.[17] Additionally, neurotoxin-mediated peripheral nervous system damage such as neuropathy or myopathy is common. Support has been shown for a number of treatments aimed at attenuating neurotoxin-mediated injury, such as antioxidant[8] and antitoxin[18] administration.

Background

 
Illustration of typical multipolar neuron

Exposure to neurotoxins in society is not new,[19] as civilizations have been exposed to neurologically destructive compounds for thousands of years. One notable example is the possible significant lead exposure during the Roman Empire resulting from the development of extensive plumbing networks and the habit of boiling vinegared wine in lead pans to sweeten it, the process generating lead acetate, known as "sugar of lead".[20] In part, neurotoxins have been part of human history because of the fragile and susceptible nature of the nervous system, making it highly prone to disruption.

The nervous tissue found in the brain, spinal cord, and periphery comprises an extraordinarily complex biological system that largely defines many of the unique traits of individuals. As with any highly complex system, however, even small perturbations to its environment can lead to significant functional disruptions. Properties leading to the susceptibility of nervous tissue include a high surface area of neurons, a high lipid content which retains lipophilic toxins, high blood flow to the brain inducing increased effective toxin exposure, and the persistence of neurons through an individual's lifetime, leading to compounding of damages.[21] As a result, the nervous system has a number of mechanisms designed to protect it from internal and external assaults, including the blood brain barrier.

The blood–brain barrier (BBB) is one critical example of protection which prevents toxins and other adverse compounds from reaching the brain.[22] As the brain requires nutrient entry and waste removal, it is perfused by blood flow. Blood can carry a number of ingested toxins, however, which would induce significant neuron death if they reach nervous tissue. Thus, protective cells termed astrocytes surround the capillaries in the brain and absorb nutrients from the blood and subsequently transport them to the neurons, effectively isolating the brain from a number of potential chemical insults.[22]

 
Astrocytes surrounding capillaries in the brain to form the blood brain barrier

This barrier creates a tight hydrophobic layer around the capillaries in the brain, inhibiting the transport of large or hydrophilic compounds. In addition to the BBB, the choroid plexus provides a layer of protection against toxin absorption in the brain. The choroid plexuses are vascularized layers of tissue found in the third, fourth, and lateral ventricles of the brain, which through the function of their ependymal cells, are responsible for the synthesis of cerebrospinal fluid (CSF).[23] Importantly, through selective passage of ions and nutrients and trapping heavy metals such as lead, the choroid plexuses maintain a strictly regulated environment which contains the brain and spinal cord.[22][23]

 
Choroid plexus

By being hydrophobic and small, or inhibiting astrocyte function, some compounds including certain neurotoxins are able to penetrate into the brain and induce significant damage. In modern times, scientists and physicians have been presented with the challenge of identifying and treating neurotoxins, which has resulted in a growing interest in both neurotoxicology research and clinical studies.[24] Though clinical neurotoxicology is largely a burgeoning field, extensive inroads have been made in the identification of many environmental neurotoxins leading to the classification of 750 to 1000 known potentially neurotoxic compounds.[21] Due to the critical importance of finding neurotoxins in common environments, specific protocols have been developed by the United States Environmental Protection Agency (EPA) for testing and determining neurotoxic effects of compounds (USEPA 1998). Additionally, in vitro systems have increased in use as they provide significant improvements over the more common in vivo systems of the past. Examples of improvements include tractable, uniform environments, and the elimination of contaminating effects of systemic metabolism.[24] In vitro systems, however, have presented problems as it has been difficult to properly replicate the complexities of the nervous system, such as the interactions between supporting astrocytes and neurons in creating the BBB.[25] To even further complicate the process of determining neurotoxins when testing in-vitro, neurotoxicity and cytotoxicity may be difficult to distinguish as exposing neurons directly to compounds may not be possible in-vivo, as it is in-vitro. Additionally, the response of cells to chemicals may not accurately convey a distinction between neurotoxins and cytotoxins, as symptoms like oxidative stress or skeletal modifications may occur in response to either.[26]

In an effort to address this complication, neurite outgrowths (either axonal or dendritic) in response to applied compounds have recently been proposed as a more accurate distinction between true neurotoxins and cytotoxins in an in-vitro testing environment. Due to the significant inaccuracies associated with this process, however, it has been slow in gaining widespread support.[27] Additionally, biochemical mechanisms have become more widely used in neurotoxin testing, such that compounds can be screened for sufficiency to induce cell mechanism interference, like the inhibition of acetylcholinesterase capacity of organophosphates (includes DDT and sarin gas).[28] Though methods of determining neurotoxicity still require significant development, the identification of deleterious compounds and toxin exposure symptoms has undergone significant improvement.

Applications in neuroscience

Though diverse in chemical properties and functions, neurotoxins share the common property that they act by some mechanism leading to either the disruption or destruction of necessary components within the nervous system. Neurotoxins, however, by their very design can be very useful in the field of neuroscience. As the nervous system in most organisms is both highly complex and necessary for survival, it has naturally become a target for attack by both predators and prey. As venomous organisms often use their neurotoxins to subdue a predator or prey very rapidly, toxins have evolved to become highly specific to their target channels such that the toxin does not readily bind other targets[29] (see Ion Channel toxins). As such, neurotoxins provide an effective means by which certain elements of the nervous system may be accurately and efficiently targeted. An early example of neurotoxin based targeting used radiolabeled tetrodotoxin to assay sodium channels and obtain precise measurements about their concentration along nerve membranes.[29] Likewise through isolation of certain channel activities, neurotoxins have provided the ability to improve the original Hodgkin-Huxley model of the neuron in which it was theorized that single generic sodium and potassium channels could account for most nervous tissue function.[29] From this basic understanding, the use of common compounds such as tetrodotoxin, tetraethylammonium, and bungarotoxins have led to a much deeper understanding of the distinct ways in which individual neurons may behave.

Mechanisms of activity

As neurotoxins are compounds which adversely affect the nervous system, a number of mechanisms through which they function are through the inhibition of neuron cellular processes. These inhibited processes can range from membrane depolarization mechanisms to inter-neuron communication. By inhibiting the ability for neurons to perform their expected intracellular functions, or pass a signal to a neighboring cell, neurotoxins can induce systemic nervous system arrest as in the case of botulinum toxin,[13] or even nervous tissue death.[30] The time required for the onset of symptoms upon neurotoxin exposure can vary between different toxins, being on the order of hours for botulinum toxin[18] and years for lead.[31]

Neurotoxin classification Neurotoxins
Na channel inhibitors Tetrodotoxin[6]
K channel inhibitors Tetraethylammonium[32]
Cl channel inhibitors Chlorotoxin,[33]
Ca channel inhibitors Conotoxin[34]
Inhibitors of synaptic vesicle release Botulinum toxin,[35]

Tetanus toxin[36]

Blood brain barrier inhibitors Aluminium,[37]

Mercury[38]

Receptor inhibitors/antagonists Bungarotoxin,[39]

Curare[40]

Receptor agonists Anatoxin-a,[41][42]

Caramboxin,[43]

25I-NBOMe,[44]

JWH-018,[45]

5-MEO-DiPT

Cytoskeleton interference Ammonia,[46]

Arsenic[47]

Ca-mediated cytotoxicity Lead[48]
Multiple effects Ethanol,[49][50]

N-Hexane,[51]

Methanol[49][50]

Receptor-selective neurotoxins MPP+
Endogenous neurotoxin sources Nitric oxide,[52]

Glutamate,[53]

Dopamine[54]

Inhibitors

Sodium channel

Tetrodotoxin
 
The puffer fish is known for carrying lethal amounts of tetrodotoxin.

Tetrodotoxin (TTX) is a poison produced by organisms belonging to the Tetraodontiformes order, which includes the puffer fish, ocean sunfish, and porcupine fish.[55] Within the puffer fish, TTX is found in the liver, gonads, intestines, and skin.[6][56] TTX can be fatal if consumed, and has become a common form of poisoning in many countries. Common symptoms of TTX consumption include paraesthesia (often restricted to the mouth and limbs), muscle weakness, nausea, and vomiting[55] and often manifest within 30 minutes of ingestion.[57] The primary mechanism by which TTX is toxic is through the inhibition of sodium channel function, which reduces the functional capacity of neuron communication. This inhibition largely affects a susceptible subset of sodium channels known as TTX-sensitive (TTX-s), which also happens to be largely responsible for the sodium current that drives the depolarization phase of neuron action potentials.[6]

 
Inhibited signaling response resulting from neuron exposure to tetrodotoxin.

TTX-resistant (TTX-r) is another form of sodium channel which has limited sensitivity to TTX, and is largely found in small diameter axons such as those found in nociception neurons.[6] When a significant level of TTX is ingested, it will bind sodium channels on neurons and reduce their membrane permeability to sodium. This results in an increased effective threshold of required excitatory signals in order to induce an action potential in a postsynaptic neuron.[6] The effect of this increased signaling threshold is a reduced excitability of postsynaptic neurons, and subsequent loss of motor and sensory function which can result in paralysis and death. Though assisted ventilation may increase the chance of survival after TTX exposure, there is currently no antitoxin. The use of the acetylcholinesterase inhibitor Neostigmine or the muscarinic acetylcholine antagonist atropine (which will inhibit parasympathetic activity), however, can increase sympathetic nerve activity enough to improve the chance of survival after TTX exposure.[55]

Potassium channel

Tetraethylammonium

Tetraethylammonium (TEA) is a compound that, like a number of neurotoxins, was first identified through its damaging effects to the nervous system and shown to have the capacity of inhibiting the function of motor nerves and thus the contraction of the musculature in a manner similar to that of curare.[58] Additionally, through chronic TEA administration, muscular atrophy would be induced.[58] It was later determined that TEA functions in-vivo primarily through its ability to inhibit both the potassium channels responsible for the delayed rectifier seen in an action potential and some population of calcium-dependent potassium channels.[32] It is this capability to inhibit potassium flux in neurons that has made TEA one of the most important tools in neuroscience. It has been hypothesized that the ability for TEA to inhibit potassium channels is derived from its similar space-filling structure to potassium ions.[58] What makes TEA very useful for neuroscientists is its specific ability to eliminate potassium channel activity, thereby allowing the study of neuron response contributions of other ion channels such as voltage gated sodium channels.[59] In addition to its many uses in neuroscience research, TEA has been shown to perform as an effective treatment of Parkinson's disease through its ability to limit the progression of the disease.[60]

Chloride channel

Chlorotoxin

Chlorotoxin (Cltx) is the active compound found in scorpion venom, and is primarily toxic because of its ability to inhibit the conductance of chloride channels.[33] Ingestion of lethal volumes of Cltx results in paralysis through this ion channel disruption. Similar to botulinum toxin, Cltx has been shown to possess significant therapeutic value. Evidence has shown that Cltx can inhibit the ability for gliomas to infiltrate healthy nervous tissue in the brain, significantly reducing the potential invasive harm caused by tumors.[61][62]

Calcium channel

Conotoxin

Conotoxins represent a category of poisons produced by the marine cone snail, and are capable of inhibiting the activity of a number of ion channels such as calcium, sodium, or potassium channels.[63][64] In many cases, the toxins released by the different types of cone snails include a range of different types of conotoxins, which may be specific for different ion channels, thus creating a venom capable of widespread nerve function interruption.[63] One of the unique forms of conotoxins, ω-conotoxin (ω-CgTx) is highly specific for Ca channels and has shown usefulness in isolating them from a system.[65] As calcium flux is necessary for proper excitability of a cell, any significant inhibition could prevent a large amount of functionality. Significantly, ω-CgTx is capable of long term binding to and inhibition of voltage-dependent calcium channels located in the membranes of neurons but not those of muscle cells.[66]

Synaptic vesicle release

Botulinum toxin
 
Mechanism of Botulinum Toxin neurotoxicity

Botulinum toxin (BTX) is a group of neurotoxins consisting of eight distinct compounds, referred to as BTX-A,B,C,D,E,F,G,H, which are produced by the bacterium Clostridium botulinum and lead to muscular paralysis.[67] A notably unique feature of BTX is its relatively common therapeutic use in treating dystonia and spasticity disorders,[67] as well as in inducing muscular atrophy[11] despite being the most poisonous substance known.[18] BTX functions peripherally to inhibit acetylcholine (ACh) release at the neuromuscular junction through degradation of the SNARE proteins required for ACh vesicle-membrane fusion.[35] As the toxin is highly biologically active, an estimated dose of 1μg/kg body weight is sufficient to induce an insufficient tidal volume and resultant death by asphyxiation.[13] Due to its high toxicity, BTX antitoxins have been an active area of research. It has been shown that capsaicin (active compound responsible for heat in chili peppers) can bind the TRPV1 receptor expressed on cholinergic neurons and inhibit the toxic effects of BTX.[18]

Tetanus toxin

Tetanus neurotoxin (TeNT) is a compound that functionally reduces inhibitory transmissions in the nervous system resulting in muscular tetany. TeNT is similar to BTX, and is in fact highly similar in structure and origin; both belonging to the same category of clostridial neurotoxins.[12] Like BTX, TeNT inhibits inter-neuron communication by means of vesicular neurotransmitter (NT) release.[36] One notable difference between the two compounds is that while BTX inhibits muscular contractions, TeNT induces them. Though both toxins inhibit vesicle release at neuron synapses, the reason for this different manifestation is that BTX functions mainly in the peripheral nervous system (PNS) while TeNT is largely active in the central nervous system (CNS).[68] This is a result of TeNT migration through motor neurons to the inhibitory neurons of the spinal cord after entering through endocytosis.[69] This results in a loss of function in inhibitory neurons within the CNS resulting in systemic muscular contractions. Similar to the prognosis of a lethal dose of BTX, TeNT leads to paralysis and subsequent suffocation.[69]

Blood brain barrier

Aluminium

Neurotoxic behavior of Aluminium is known to occur upon entry into the circulatory system, where it can migrate to the brain and inhibit some of the crucial functions of the blood brain barrier (BBB).[37] A loss of function in the BBB can produce significant damage to the neurons in the CNS, as the barrier protecting the brain from other toxins found in the blood will no longer be capable of such action. Though the metal is known to be neurotoxic, effects are usually restricted to patients incapable of removing excess ions from the blood, such as those experiencing renal failure.[70] Patients experiencing aluminium toxicity can exhibit symptoms such as impaired learning and reduced motor coordination.[71] Additionally, systemic aluminium levels are known to increase with age, and have been shown to correlate with Alzheimer's Disease, implicating it as a neurotoxic causative compound of the disease.[72] Despite its known toxicity in its ionic form, studies are divided on the potential toxicity of using aluminium in packaging and cooking appliances.

Mercury

Mercury is capable of inducing CNS damage by migrating into the brain by crossing the BBB.[38] Mercury exists in a number of different compounds, though methylmercury (MeHg+), dimethylmercury and diethylmercury are the only significantly neurotoxic forms. Diethylmercury and dimethylmercury are considered some of the most potent neurotoxins ever discovered.[38] MeHg+ is usually acquired through consumption of seafood, as it tends to concentrate in organisms high on the food chain.[73] It is known that the mercuric ion inhibits amino acid (AA) and glutamate (Glu) transport, potentially leading to excitotoxic effects.[74]

Receptor agonists and antagonists

Anatoxin-a

External video
  Very Fast Death Factor
University of Nottingham

Investigations into anatoxin-a, also known as "Very Fast Death Factor", began in 1961 following the deaths of cows that drank from a lake containing an algal bloom in Saskatchewan, Canada.[41][42] It is a cyanotoxin produced by at least four different genera of cyanobacteria, and has been reported in North America, Europe, Africa, Asia, and New Zealand.[75]

Toxic effects from anatoxin-a progress very rapidly because it acts directly on the nerve cells (neurons). The progressive symptoms of anatoxin-a exposure are loss of coordination, twitching, convulsions and rapid death by respiratory paralysis. The nerve tissues which communicate with muscles contain a receptor called the nicotinic acetylcholine receptor. Stimulation of these receptors causes a muscular contraction. The anatoxin-a molecule is shaped so it fits this receptor, and in this way it mimics the natural neurotransmitter normally used by the receptor, acetylcholine. Once it has triggered a contraction, anatoxin-a does not allow the neurons to return to their resting state, because it is not degraded by cholinesterase which normally performs this function. As a result, the muscle cells contract permanently, the communication between the brain and the muscles is disrupted and breathing stops.[76][77]

When it was first discovered, the toxin was called the Very Fast Death Factor (VFDF) because when it was injected into the body cavity of mice it induced tremors, paralysis and death within a few minutes. In 1977, the structure of VFDF was determined as a secondary, bicyclic amine alkaloid, and it was renamed anatoxin-a.[78][79] Structurally, it is similar to cocaine.[80] There is continued interest in anatoxin-a because of the dangers it presents to recreational and drinking waters, and because it is a particularly useful molecule for investigating acetylcholine receptors in the nervous system.[81] The deadliness of the toxin means that it has a high military potential as a toxin weapon.[82]

Bungarotoxin

Bungarotoxin is a compound with known interaction with nicotinic acetylcholine receptors (nAChRs), which constitute a family of ion channels whose activity is triggered by neurotransmitter binding.[83] Bungarotoxin is produced in a number of different forms, though one of the commonly used forms is the long chain alpha form, α-bungarotoxin, which is isolated from the banded krait snake.[39] Though extremely toxic if ingested, α-bungarotoxin has shown extensive usefulness in neuroscience as it is particularly adept at isolating nAChRs due to its high affinity to the receptors.[39] As there are multiple forms of bungarotoxin, there are different forms of nAChRs to which they will bind, and α-bungarotoxin is particularly specific for α7-nAChR.[84] This α7-nAChR functions to allow calcium ion influx into cells, and thus when blocked by ingested bungarotoxin will produce damaging effects, as ACh signaling will be inhibited.[84] Likewise, the use of α-bungarotoxin can be very useful in neuroscience if it is desirable to block calcium flux in order to isolate effects of other channels. Additionally, different forms of bungarotoxin may be useful for studying inhibited nAChRs and their resultant calcium ion flow in different systems of the body. For example, α-bungarotoxin is specific for nAChRs found in the musculature and κ-bungarotoxin is specific for nAChRs found in neurons.[85]

Caramboxin

Caramboxin (CBX) is a toxin found in star fruit (Averrhoa carambola). Individuals with some types of kidney disease are susceptible to adverse neurological effects including intoxication, seizures and even death after eating star fruit or drinking juice made of this fruit. Caramboxin is a new nonpeptide amino acid toxin that stimulate the glutamate receptors in neurons. Caramboxin is an agonist of both NMDA and AMPA glutamatergic ionotropic receptors with potent excitatory, convulsant, and neurodegenerative properties.[43]

Curare

The term "curare" is ambiguous because it has been used to describe a number of poisons which at the time of naming were understood differently from present day understandings. In the past the characterization has meant poisons used by South American tribes on arrows or darts, though it has matured to specify a specific categorization of poisons which act on the neuromuscular junction to inhibit signaling and thus induce muscle relaxation.[86] The neurotoxin category contains a number of distinct poisons, though all were originally purified from plants originating in South America.[86] The effect with which injected curare poison is usually associated is muscle paralysis and resultant death.[87] Curare notably functions to inhibit nicotinic acetylcholine receptors at the neuromuscular junction. Normally, these receptor channels allow sodium ions into muscle cells to initiate an action potential that leads to muscle contraction. By blocking the receptors, the neurotoxin is capable of significantly reducing neuromuscular junction signaling, an effect which has resulted in its use by anesthesiologists to produce muscular relaxation.[88]

Cytoskeleton interference

Ammonia

 
An Astrocyte, a cell notable for maintaining the blood brain barrier

Ammonia toxicity is often seen through two routes of administration, either through consumption or through endogenous ailments such as liver failure.[89][90] One notable case in which ammonia toxicity is common is in response to cirrhosis of the liver which results in hepatic encephalopathy, and can result in cerebral edema (Haussinger 2006). This cerebral edema can be the result of nervous cell remodeling. As a consequence of increased concentrations, ammonia activity in-vivo has been shown to induce swelling of astrocytes in the brain through increased production of cGMP (Cyclic Guanosine Monophosphate) within the cells which leads to Protein Kinase G-mediated (PKG) cytoskeletal modifications.[46] The resultant effect of this toxicity can be reduced brain energy metabolism and function. Importantly, the toxic effects of ammonia on astrocyte remodeling can be reduced through administration of L-carnitine.[89] This astrocyte remodeling appears to be mediated through ammonia-induced mitochondrial permeability transition. This mitochondrial transition is a direct result of glutamine activity a compound which forms from ammonia in-vivo.[91] Administration of antioxidants or glutaminase inhibitor can reduce this mitochondrial transition, and potentially also astrocyte remodeling.[91]

Arsenic

Arsenic is a neurotoxin commonly found concentrated in areas exposed to agricultural runoff, mining, and smelting sites (Martinez-Finley 2011). One of the effects of arsenic ingestion during the development of the nervous system is the inhibition of neurite growth[92] which can occur both in PNS and the CNS.[93] This neurite growth inhibition can often lead to defects in neural migration, and significant morphological changes of neurons during development,[94]) often leading to neural tube defects in neonates.[95] As a metabolite of arsenic, arsenite is formed after ingestion of arsenic and has shown significant toxicity to neurons within about 24 hours of exposure. The mechanism of this cytotoxicity functions through arsenite-induced increases in intracellular calcium ion levels within neurons, which may subsequently reduce mitochondrial transmembrane potential which activates caspases, triggering cell death.[94] Another known function of arsenite is its destructive nature towards the cytoskeleton through inhibition of neurofilament transport.[47] This is particularly destructive as neurofilaments are used in basic cell structure and support. Lithium administration has shown promise, however, in restoring some of the lost neurofilament motility.[96] Additionally, similar to other neurotoxin treatments, the administration of certain antioxidants has shown some promise in reducing neurotoxicity of ingested arsenic.[94]

Calcium-mediated cytotoxicity

Lead

 
Lead pipes and solder are common sources of ingested lead.

Lead is a potent neurotoxin whose toxicity has been recognized for at least thousands of years.[97] Though neurotoxic effects for lead are found in both adults and young children, the developing brain is particularly susceptible to lead-induced harm, effects which can include apoptosis and excitotoxicity.[97] An underlying mechanism by which lead is able to cause harm is its ability to be transported by calcium ATPase pumps across the BBB, allowing for direct contact with the fragile cells within the central nervous system.[98] Neurotoxicity results from lead's ability to act in a similar manner to calcium ions, as concentrated lead will lead to cellular uptake of calcium which disrupts cellular homeostasis and induces apoptosis.[48] It is this intracellular calcium increase that activates protein kinase C (PKC), which manifests as learning deficits in children as a result of early lead exposure.[48] In addition to inducing apoptosis, lead inhibits interneuron signaling through the disruption of calcium-mediated neurotransmitter release.[99]

Neurotoxins with multiple effects

Ethanol

 
Male baby exhibiting Fetal Alcohol Syndrome (FAS).

As a neurotoxin, ethanol has been shown to induce nervous system damage and affect the body in a variety of ways. Among the known effects of ethanol exposure are both transient and lasting consequences. Some of the lasting effects include long-term reduced neurogenesis in the hippocampus,[100][101] widespread brain atrophy,[102] and induced inflammation in the brain.[103] Of note, chronic ethanol ingestion has additionally been shown to induce reorganization of cellular membrane constituents, leading to a lipid bilayer marked by increased membrane concentrations of cholesterol and saturated fat.[50] This is important as neurotransmitter transport can be impaired through vesicular transport inhibition, resulting in diminished neural network function. One significant example of reduced inter-neuron communication is the ability for ethanol to inhibit NMDA receptors in the hippocampus, resulting in reduced long-term potentiation (LTP) and memory acquisition.[49] NMDA has been shown to play an important role in LTP and consequently memory formation.[104] With chronic ethanol intake, however, the susceptibility of these NMDA receptors to induce LTP increases in the mesolimbic dopamine neurons in an inositol 1,4,5-triphosphate (IP3) dependent manner.[105] This reorganization may lead to neuronal cytotoxicity both through hyperactivation of postsynaptic neurons and through induced addiction to continuous ethanol consumption. It has, additionally, been shown that ethanol directly reduces intracellular calcium ion accumulation through inhibited NMDA receptor activity, and thus reduces the capacity for the occurrence of LTP.[106]

In addition to the neurotoxic effects of ethanol in mature organisms, chronic ingestion is capable of inducing severe developmental defects. Evidence was first shown in 1973 of a connection between chronic ethanol intake by mothers and defects in their offspring.[107] This work was responsible for creating the classification of fetal alcohol syndrome, a disease characterized by common morphogenesis aberrations such as defects in craniofacial formation, limb development, and cardiovascular formation. The magnitude of ethanol neurotoxicity in fetuses leading to fetal alcohol syndrome has been shown to be dependent on antioxidant levels in the brain such as vitamin E.[108] As the fetal brain is relatively fragile and susceptible to induced stresses, severe deleterious effects of alcohol exposure can be seen in important areas such as the hippocampus and cerebellum. The severity of these effects is directly dependent upon the amount and frequency of ethanol consumption by the mother, and the stage in development of the fetus.[109] It is known that ethanol exposure results in reduced antioxidant levels, mitochondrial dysfunction (Chu 2007), and subsequent neuronal death, seemingly as a result of increased generation of reactive oxidative species (ROS).[30] This is a plausible mechanism, as there is a reduced presence in the fetal brain of antioxidant enzymes such as catalase and peroxidase.[110] In support of this mechanism, administration of high levels of dietary vitamin E results in reduced or eliminated ethanol-induced neurotoxic effects in fetuses.[8]

n-Hexane

n-Hexane is a neurotoxin which has been responsible for the poisoning of several workers in Chinese electronics factories in recent years.[111][112][113][51]

Receptor-selective neurotoxins

MPP+

MPP+, the toxic metabolite of MPTP is a selective neurotoxin which interferes with oxidative phosphorylation in mitochondria by inhibiting complex I, leading to the depletion of ATP and subsequent cell death. This occurs almost exclusively in dopaminergic neurons of the substantia nigra, resulting in the presentation of permanent parkinsonism in exposed subjects 2–3 days after administration.

Endogenous neurotoxin sources

Unlike most common sources of neurotoxins which are acquired by the body through ingestion, endogenous neurotoxins both originate from and exert their effects in-vivo. Additionally, though most venoms and exogenous neurotoxins will rarely possess useful in-vivo capabilities, endogenous neurotoxins are commonly used by the body in useful and healthy ways, such as nitric oxide which is used in cell communication.[114] It is often only when these endogenous compounds become highly concentrated that they lead to dangerous effects.[9]

Nitric oxide

Though nitric oxide (NO) is commonly used by the nervous system in inter-neuron communication and signaling, it can be active in mechanisms leading to ischemia in the cerebrum (Iadecola 1998). The neurotoxicity of NO is based on its importance in glutamate excitotoxicity, as NO is generated in a calcium-dependent manner in response to glutamate mediated NMDA activation, which occurs at an elevated rate in glutamate excitotoxicity.[52] Though NO facilitates increased blood flow to potentially ischemic regions of the brain, it is also capable of increasing oxidative stress,[115] inducing DNA damage and apoptosis.[116] Thus an increased presence of NO in an ischemic area of the CNS can produce significantly toxic effects.

Glutamate

Glutamate, like nitric oxide, is an endogenously produced compound used by neurons to perform normally, being present in small concentrations throughout the gray matter of the CNS.[9] One of the most notable uses of endogenous glutamate is its functionality as an excitatory neurotransmitter.[53] When concentrated, however, glutamate becomes toxic to surrounding neurons. This toxicity can be both a result of direct lethality of glutamate on neurons and a result of induced calcium flux into neurons leading to swelling and necrosis.[53] Support has been shown for these mechanisms playing significant roles in diseases and complications such as Huntington's disease, epilepsy, and stroke.[9]

See also

Notes

  1. ^ Sivonen, K (1999). "Toxins produced by cyanobacteria". Vesitalous. 5: 11–18.
  2. ^ Scottish Government Blue-Green Algae (Cyanobacteria) in Inland Waters: Assessment and Control of Risks to Public Health Retrieved 15 December 2011.
  3. ^ Dorland's Medical Dictionary for Health Consumers
  4. ^ a b Spencer 2000
  5. ^ a b Olney 2002
  6. ^ a b c d e f g h Kiernan 2005
  7. ^ Lidsky 2003
  8. ^ a b c Heaton; Barrow, Marieta; Mitchell, J. Jean; Paiva, Michael (2000). "Amelioration of Ethanol-Induced Neurotoxicity in the Neonatal Rat Central Nervous System by Antioxidant Therapy". Alcoholism: Clinical and Experimental Research. 24 (4): 512–18. doi:10.1111/j.1530-0277.2000.tb02019.x. PMID 10798588.
  9. ^ a b c d Choi 1987
  10. ^ Zhang 1994
  11. ^ a b Rosales, Raymond L.; Arimura, Kimiyoshi; Takenaga, Satoshi; Osame, Mitsuhiro (1996). "Extrafusal and Intrafusal Muscle Effects in Experimental Botulinum Toxin-A Injection". Muscle & Nerve. 19 (4): 488–96. doi:10.1002/(sici)1097-4598(199604)19:4<488::aid-mus9>3.0.co;2-8. PMID 8622728. S2CID 20849034.
  12. ^ a b Simpson 1986
  13. ^ a b c Arnon 2001
  14. ^ Dikranian 2001
  15. ^ Deng 2003
  16. ^ Jevtovic-Todorovic 2003
  17. ^ Nadler 1978
  18. ^ a b c d Thyagarajan 2009
  19. ^ Neurotoxins: Definition, Epidemiology, Etiology
  20. ^ Hodge 2002
  21. ^ a b Dobbs 2009
  22. ^ a b c Widmaier, Eric P., Hershel Raff, Kevin T. Strang, and Arthur J. Vander (2008) Vander's Human Physiology: the Mechanisms of Body Function.' Boston: McGraw-Hill Higher Education.
  23. ^ a b Martini 2009
  24. ^ a b Costa 2011
  25. ^ Harry 1998
  26. ^ Gartlon 2006
  27. ^ Radio, Nicholas M.; Mundy, William R. (2008). "Developmental Neurotoxicity Testing in Vitro: Models for Assessing Chemical Effects on Neurite Out-growth". NeuroToxicology. 29 (3): 361–376. doi:10.1016/j.neuro.2008.02.011. PMID 18403021.
  28. ^ Lotti 2005
  29. ^ a b c Adams 2003
  30. ^ a b Brocardo 2011
  31. ^ Lewendon 2001
  32. ^ a b Haghdoost-Yazdi 2011
  33. ^ a b DeBin 1993
  34. ^ McClesky 1987
  35. ^ a b Garcia-Rodriguez 2011
  36. ^ a b Williamson 1996
  37. ^ a b Banks 1988
  38. ^ a b c Aschner 1990
  39. ^ a b c Dutertre 2006
  40. ^ Koller 1988
  41. ^ a b Carmichael 1978
  42. ^ a b Carmichael 1975
  43. ^ a b Garcia-Cairasco, N.; Moyses-Neto, M.; Del Vecchio, F.; Oliveira, J. A. C.; Dos Santos, F. L.; Castro, O. W.; Arisi, G. M.; Dantas, M. R.; Carolino, R. O. G.; Coutinho-Netto, J.; Dagostin, A. L. A.; Rodrigues, M. C. A.; Leão, R. M.; Quintiliano, S. A. P.; Silva, L. F.; Gobbo-Neto, L.; Lopes, N. P. (2013). "Elucidating the Neurotoxicity of the Star Fruit". Angewandte Chemie International Edition. 52 (49): 13067–13070. doi:10.1002/anie.201305382. PMID 24281890.
  44. ^ Rutgrere 2012
  45. ^ Roller 1994
  46. ^ a b Konopacka 2009
  47. ^ a b DeFuria 2006
  48. ^ a b c Bressler 1999
  49. ^ a b c Lovinger, D.; White, G.; Weight, F. (1989). "Ethanol Inhibits NMDA-activated Ion Current in Hippocampal Neurons". Science. 243 (4899): 1721–724. Bibcode:1989Sci...243.1721L. doi:10.1126/science.2467382. PMID 2467382.
  50. ^ a b c Leonard, B. E. (1986). "Is Ethanol a Neurotoxin?: the Effects of Ethanol on Neuronal Structure and Function". Alcohol and Alcoholism. 21 (4): 325–38. doi:10.1093/oxfordjournals.alcalc.a044638. PMID 2434114.
  51. ^ a b Occupational Safety and Health Guideline for n-Hexane 2011-12-18 at the Wayback Machine, OSHA.gov
  52. ^ a b Garthwaite 1988
  53. ^ a b c Choi 1990
  54. ^ Ben-Shachar D, Zuk R, Glinka Y (1995). "Dopamine neurotoxicity: inhibition of mitochondrial respiration". J. Neurochem. 64 (2): 718–23. doi:10.1046/j.1471-4159.1995.64020718.x. PMID 7830065. S2CID 9060404.
  55. ^ a b c Chowdhury, F. R.; Ahasan, H A M. Nazmul; Rashid, A K M. Mamunur; Mamun, A. Al; Khaliduzzaman, S. M. (2007). "Tetrodotoxin Poisoning: a Clinical Analysis, Role of Neostigmine and Short-term Outcome of 53 Cases". Singapore Medical Journal. 48 (9): 830–33. PMID 17728964.
  56. ^ Ahasan 2004
  57. ^ Lau 1995
  58. ^ a b c Standfield 1983
  59. ^ Roed 1989
  60. ^ Haghdoost-Yasdi 2011
  61. ^ Deshane 2003
  62. ^ Soroceanu 1998
  63. ^ a b Jacob 2010
  64. ^ Olivera 1987
  65. ^ Cruz 1986
  66. ^ McCleskey 1987
  67. ^ a b Brin, Mitchell F (1997) "Botulinum Toxin: Chemistry, Pharmacology, Toxicity, and Immunology." Muscle & Nerve, 20 (S6): 146–68.
  68. ^ Montecucco 1986
  69. ^ a b Pirazzini 2011
  70. ^ King 1981
  71. ^ Rabe 1982
  72. ^ Walton 2006
  73. ^ Chan 2011
  74. ^ Brookes 1988
  75. ^ Yang 2007
  76. ^ Wood 2007
  77. ^ National Center for Environmental Assessment
  78. ^ Devlin 1977
  79. ^ Moore 1977
  80. ^ Metcalf 2009
  81. ^ Stewart 2008
  82. ^ Dixit 2005
  83. ^ Tsetlin 2003
  84. ^ a b Liu 2008
  85. ^ Hue 2007
  86. ^ a b Bisset 1992
  87. ^ Schlesinger 1946
  88. ^ Griffith, Harold R.; Johnson, G. Enid (1942). "The Use Of Curare In General Anesthesia". Anesthesiology. 3 (4): 418–420. doi:10.1097/00000542-194207000-00006. S2CID 71400545.
  89. ^ a b Matsuoka 1991
  90. ^ Buzanska (2000)
  91. ^ a b Norenberg 2004
  92. ^ Liu 2009[full citation needed]
  93. ^ Vahidnia 2007
  94. ^ a b c Rocha 2011
  95. ^ Brender 2005
  96. ^ DeFuria 2007
  97. ^ a b Lidskey 2003
  98. ^ Bradbury 1993
  99. ^ Lasley 1999
  100. ^ Taffe 2010
  101. ^ Morris 2009
  102. ^ Bleich 2003
  103. ^ Blanco 2005
  104. ^ Davis 1992
  105. ^ Bernier 2011
  106. ^ Takadera 1990
  107. ^ Jones 1973
  108. ^ Mitchell 1999
  109. ^ Gil-Mohapel 2010
  110. ^ Bergamini 2004
  111. ^ Workers poisoned while making iPhones ABC News, October 25, 2010
  112. ^ Dirty Secrets 2017-05-25 at the Wayback Machine ABC Foreign Correspondent, 2010-Oct-26
  113. ^ Mr Daisey and the Apple Factory, This American Life, January 6, 2012
  114. ^ Iadecola 1998
  115. ^ Beckman 1990
  116. ^ Bonfoco 1995

References

  • Adams, Michael E.; Olivera, Baldomero M. (1994). "Neurotoxins: Overview of an Emerging Research Technology". Trends in Neurosciences. 17 (4): 151–55. doi:10.1016/0166-2236(94)90092-2. PMID 7517594. S2CID 3984106.
  • Arnon, Stephen S.; Schechter, Robert; Inglesby, Thomas V.; Henderson, Donald A.; Bartlett, John G.; Ascher, Michael S.; Eitzen, Edward; Fine, Anne D.; Hauer, Jerome; Layton, Marcelle; Lillibridge, Scott; Osterholm, Michael T.; O'Toole, Tara; Parker, Gerald; Perl, Trish M.; Russell, Philip K.; Swerdlow, David L.; Tonat, Kevin (2001). "Botulinum Toxin as a Biological Weapon". The Journal of the American Medical Association. 285 (8): 1059–069. doi:10.1001/jama.285.8.1059. PMID 11209178.
  • Aschner, M.; Aschner, J. (1990). "Mercury Neurotoxicity: Mechanisms of Blood–brain barrier Transport". Neuroscience & Biobehavioral Reviews. 14 (2): 169–76. doi:10.1016/s0149-7634(05)80217-9. PMID 2190116. S2CID 8777931.
  • Banks, William A.; Kastin, Abba J. (1989). "Aluminum-Induced Neurotoxicity: Alterations in Membrane Function at the Blood–brain barrier". Neuroscience & Biobehavioral Reviews. 13 (1): 47–53. doi:10.1016/s0149-7634(89)80051-x. PMID 2671833. S2CID 46507895.
  • Baum-Baicker, Cynthia (1985). "The Health Benefits of Moderate Alcohol Consumption: A Review of the Literature". Drug and Alcohol Dependence. 15 (3): 207–27. doi:10.1016/0376-8716(85)90001-8. PMID 4028954.
  • Beckman, J. S. (1990). "Apparent Hydroxyl Radical Production by Peroxynitrite: Implications for Endothelial Injury from Nitric Oxide and Superoxide". Proceedings of the National Academy of Sciences. 87 (4): 1620–624. Bibcode:1990PNAS...87.1620B. doi:10.1073/pnas.87.4.1620. PMC 53527. PMID 2154753.
  • Bergamini, Carlo M.; Gambetti, Stefani; Dondi, Alessia; Cervellati, Carlo (2004). "Oxygen, Reactive Oxygen Species and Tissue Damage". Current Pharmaceutical Design. 10 (14): 1611–626. doi:10.2174/1381612043384664. PMID 15134560.
  • Bernier Brian E., Whitaker Leslie R., Morikawa Hitoshi (2011). "Previous Ethanol Experience Enhances Synaptic Plasticity of NMDA Receptors in the Ventral Tegmental Area". The Journal of Neuroscience. 31 (14): 5305–212. doi:10.1523/JNEUROSCI.5282-10.2011. PMC 3086894. PMID 21471355.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  • Bisset, Norman G (1992). "War and Hunting Poisons of the New World. Part 1. Notes on the Early History of Curare". Journal of Ethnopharmacology. 36 (1): 1–26. doi:10.1016/0378-8741(92)90056-w. PMID 1501489.
  • Blanco, Ana M.; Valles, Soraya L.; Pascual, Maria; Guerri, Consuelo (2005). "Involvement of TLR4/Type I IL-1 Receptor Signaling in the Induction of Inflammatory Mediators and Cell Death Induced by Ethanol in Cultured Astrocytes". The Journal of Immunology. 175 (10): 6893–899. doi:10.4049/jimmunol.175.10.6893. PMID 16272348.
  • Bleich, S (2003). "Hyperhomocysteinemia as a New Risk Factor for Brain Shrinkage in Patients with Alcoholism". Neuroscience Letters. 335 (3): 179–82. doi:10.1016/s0304-3940(02)01194-1. PMID 12531462. S2CID 33032529.
  • Bonfoco E (1995). "Apoptosis and Necrosis: Two Distinct Events Induced, Respectively, by Mild and Intense Insults with N-Methyl-D-Aspartate or Nitric Oxide/Superoxide in Cortical Cell Cultures". Proceedings of the National Academy of Sciences. 92 (16): 7162–166. Bibcode:1995PNAS...92.7162B. doi:10.1073/pnas.92.16.7162. PMC 41299. PMID 7638161.
  • Bradbury, MW; Deane, R (1993). "Permeability of the blood±brain barrier to lead. [Review]". Neurotoxicology. 14 (2–3): 131–6. PMID 8247388.
  • Brender, J.; Suarez, L.; Felkner, M.; Gilani, Z.; Stinchcomb, D.; Moody, K.; Henry, J.; Hendricks, K. (2006). "Maternal Exposure to Arsenic, Cadmium, Lead, and Mercury and Neural Tube Defects in Offspring". Environmental Research. 101 (1): 132–39. Bibcode:2006ER....101..132B. doi:10.1016/j.envres.2005.08.003. PMID 16171797.
  • Bressler, J; Kim, KA; Chakraborti, T; Goldstein, G (1999). "Molecular mechanisms of lead neurotoxicity. [Review]". Neurochem Res. 24 (4): 595–600. doi:10.1023/A:1022596115897. PMID 10227691. S2CID 3017703.
  • Brocardo, Patricia S.; Gil-Mohapel, Joana; Christie, Brian R. (2011). "The Role of Oxidative Stress in Fetal Alcohol Spectrum Disorders". Brain Research Reviews. 67 (1–2): 209–25. doi:10.1016/j.brainresrev.2011.02.001. PMID 21315761. S2CID 1213492.
  • Brookes, N (1988). "Specificity and Reversibility of the Inhibition by HgCl of Glutamate Transport in Astrocyte Cultures". Journal of Neurochemistry. 50 (4): 1117–122. doi:10.1111/j.1471-4159.1988.tb10581.x. PMID 2894409. S2CID 27522735.
  • Buzanska, L.; Zablocka, B.; Dybel, A.; Domanska-Janik, K.; Albrecht, J. (2000). "Delayed Induction of Apoptosis by Ammonia in C6 Glioma Cells". Neurochemistry International. 37 (2–3): 287–97. doi:10.1016/s0197-0186(00)00030-9. PMID 10812214. S2CID 33351533.
  • Carmichael WW, Biggs DF, Gorham PR (1975). "Toxicology and pharmacological action of Anabaena flos-aquae toxin". Science. 187 (4176): 542–544. Bibcode:1975Sci...187..542C. doi:10.1126/science.803708. PMID 803708.
  • Carmichael WW, Gorham PR (1978). "Anatoxins from clones of Anabaena flos-aquae isolated from lakes of western Canada". Mitt. Infernal. Verein. Limnol. 21: 285–295.
  • Chan, H. M. (2011) "Mercury in Fish: Human Health Risks." Encyclopedia of Environmental Health: 697–704.
  • Choi, D (1988). "Calcium-mediated Neurotoxicity: Relationship to Specific Channel Types and Role in Ischemic Damage". Trends in Neurosciences. 11 (10): 465–69. doi:10.1016/0166-2236(88)90200-7. PMID 2469166. S2CID 36909560.
  • Choi, D. W.; Rothman, S. M. (1990). "The Role of Glutamate Neurotoxicity in Hypoxic-Ischemic Neuronal Death". Annual Review of Neuroscience. 13 (1): 171–82. doi:10.1146/annurev.neuro.13.1.171. PMID 1970230.
  • Choi, Dennis W (1987). "Ionic Dependence of Glutamate Neurotoxicity". The Journal of Neuroscience. 7 (2): 369–79. doi:10.1523/JNEUROSCI.07-02-00369.1987. PMC 6568907. PMID 2880938.
  • Choi, Dennis W.; Maulucci-Gedde, Margaret; Kriegstein, Arnold R. (1987). "Glutamate Neurotoxicity in Cortical Cell Culture". The Journal of Neuroscience. 7 (2): 357–68. doi:10.1523/JNEUROSCI.07-02-00357.1987. PMC 6568898. PMID 2880937.
  • Chu, Jennifer; Tong, Ming; Monte, Suzanne M. (2007). "Chronic Ethanol Exposure Causes Mitochondrial Dysfunction and Oxidative Stress in Immature Central Nervous System Neurons". Acta Neuropathologica. 113 (6): 659–73. doi:10.1007/s00401-007-0199-4. PMID 17431646. S2CID 25418731.
  • Clancy, Barbara; Finlay, Barbara L.; Darlington, Richard B.; Anand, K.J.S. (2007). "Extrapolating Brain Development from Experimental Species to Humans". NeuroToxicology. 28 (5): 931–37. doi:10.1016/j.neuro.2007.01.014. PMC 2077812. PMID 17368774.
  • Costa, Lucio G., Gennaro Giordano, and Marina Guizzetti (2011) In Vitro Neurotoxicology: Methods and Protocols. New York: Humana.
  • Coyle, Joseph T.; Schwarcz, Robert (1976). "Lesion of Striatal Neurons with Kainic Acid Provides a Model for Huntington's Chorea". Nature. 246 (5574): 244–46. Bibcode:1976Natur.263..244C. doi:10.1038/263244a0. PMID 8731. S2CID 4271762.
  • Cruz, Lourdes J.; Olivera, Baldomero M. (1987). "Calcium Channel Antagonists ω-Conotoxin Defines a New High Affinity Site". The Journal of Biological Chemistry. 14 (261): 6230–233.
  • Davis, S.; Butcher, S. P.; Morris, R. (1992). "The NMDA Receptor Antagonist D-2-amino-5phosphonopentanoate (D-AP5) Impairs Spatial Learning and LTP in Vivo at Intracerebral Concentrations Comparable to Those That Block LTP in Vitro". The Journal of Neuroscience. 12 (1): 21–34. doi:10.1523/JNEUROSCI.12-01-00021.1992. PMC 6575679. PMID 1345945.
  • Dawson, V. L. (1991). "Nitric Oxide Mediates Glutamate Neurotoxicity in Primary Cortical Cultures". Proceedings of the National Academy of Sciences. 88 (14): 6368–371. Bibcode:1991PNAS...88.6368D. doi:10.1073/pnas.88.14.6368. PMC 52084. PMID 1648740.
  • Debin, John A., John E. Maggio, and Gary R. Strichartz (1993) "Purification and Characterization of Chlorotoxin, a Chloride Channel Ligand from the Venom of the Scorpion." The American Physiological Society, pp. 361–69.
  • DeFuria, Jason; Shea, Thomas B. (2007). "Arsenic Inhibits Neurofilament Transport and Induces Perikaryal Accumulation of Phosphorylated Neurofilaments: Roles of JNK and GSK-3β". Brain Research. 1181: 74–82. doi:10.1016/j.brainres.2007.04.019. PMID 17961518. S2CID 37068398.
  • Defuria, Jason (2006) "The Environmental Neurotoxin Arsenic Impairs Neurofilament Dynamics by Overactivation of C-JUN Terminal Kinase: Potential Role for Amyotrophic Lateral Sclerosis." UMI, pp. 1–16.
  • Deng Wenbin, Poretz Ronald D (2003). "Oligodendroglia in Developmental Neurotoxicity". NeuroToxicology. 24 (2): 161–78. doi:10.1016/s0161-813x(02)00196-1. PMID 12606289.
  • Deshane, Jessy; Garner, Craig C.; Sontheimer, Harald (2003). "Chlorotoxin Inhibits Glioma Cell Invasion via Matrix Metalloproteinase-2". The Journal of Biological Chemistry. 278 (6): 4135–144. doi:10.1074/jbc.m205662200. PMID 12454020.
  • Devlin JP, Edwards OE, Gorham PR, Hunter NR, Pike RK, Stavric B (1977). "Anatoxin-a, a toxic alkaloid from Anabaena flos-aquae NRC-44h". Can. J. Chem. 55 (8): 1367–1371. doi:10.1139/v77-189. Archived from the original on 2012-07-09.
  • Dikranian, K (2001). "Apoptosis in the in Vivo Mammalian Forebrain". Neurobiology of Disease. 8 (3): 359–79. doi:10.1006/nbdi.2001.0411. PMID 11447994. S2CID 21965033.
  • Dixit A, Dhaked RK, Alam SI, Singh L (2005). "Military potential of biological neurotoxins". Toxin Reviews. 24 (2): 175–207. doi:10.1081/TXR-200057850. S2CID 85651107.
  • Dobbs, Michael R (2009) Clinical Neurotoxicology. Philadelphia: Saunders-Elsevier.
  • Dutertre, S.; Lewis, R. (2006). "Toxin Insights into Nicotinic Acetylcholine Receptors". Biochemical Pharmacology. 72 (6): 661–70. doi:10.1016/j.bcp.2006.03.027. PMID 16716265.
  • Garcia-Rodriguez, C.; Geren, I. N.; Lou, J.; Conrad, F.; Forsyth, C.; Wen, W.; Chakraborti, S.; Zao, H.; Manzanarez, G.; Smith, T. J.; Brown, J.; Tepp, W. H.; Liu, N.; Wijesuriya, S.; Tomic, M. T.; Johnson, E. A.; Smith, L. A.; Marks, J. D. (2011). "Response Re: 'Neutralizing Human Monoclonal Antibodies Binding Multiple Serotypes of Botulinum Neurotoxin' by Garcia-Rodriguez Et Al., PEDS, 2011;24:321–331". Protein Engineering Design and Selection. 24 (9): 633–34. doi:10.1093/protein/gzr012.
  • Garthwaite, John; Charles, Sarah L.; Chess-Williams, Russel (1988). "Endothelim-derived Relaxing Factor Release on Activation of NMDA Receptors Suggests Role as Intercellular Messenger in the Brain". Nature. 336 (24): 385–88. Bibcode:1988Natur.336..385G. doi:10.1038/336385a0. PMID 2904125. S2CID 4361381.
  • Gartlon, J.; Kinsner, A.; Balprice, A.; Coecke, S.; Clothier, R. (2006). "Evaluation of a Proposed in Vitro Test Strategy Using Neuronal and Non-neuronal Cell Systems for Detecting Neurotoxicity". Toxicology in Vitro. 20 (8): 1569–581. doi:10.1016/j.tiv.2006.07.009. PMID 16959468.
  • Gil-Mohapel, Joana; Boehme, Fanny; Kainer, Leah; Christie, Brian R. (2010). "Hippocampal Cell Loss and Neurogenesis after Fetal Alcohol Exposure: Insights from Different Rodent Models". Brain Research Reviews. 64 (2): 283–303. doi:10.1016/j.brainresrev.2010.04.011. PMID 20471420. S2CID 10159395.
  • Haghdoost-Yazdi, Hashem; Faraji, Ayda; Fraidouni, Negin; Movahedi, Mohadeseh; Hadibeygi, Elham; Vaezi, Fatemeh (2011). "Significant Effects of 4-aminopyridine and Tetraethylammonium in the Treatment of 6-hydroxydopamine-induced Parkinson's Disease". Behavioural Brain Research. 223 (1): 70–74. doi:10.1016/j.bbr.2011.04.021. PMID 21540059. S2CID 7100177.
  • Harry, G. J.; Billingsley, Melvin; Bruinink, Arendd; Campbell, Iain L.; Classen, Werner; Dorman, David C.; Galli, Corrado; Ray, David; Smith, Robert A.; Tilson, Hugh A. (1998). "In Vitro Techniques for the Assessment of Neurotoxicity". Environmental Health Perspectives. 106 (Suppl 1): 131–58. doi:10.2307/3433917. JSTOR 3433917. PMC 1533280. PMID 9539010.
  • Häussinger, Dieter (2006). "Low Grade Cerebral Edema and the Pathogenesis of Hepatic Encephalopathy in Cirrhosis". Hepatology. 43 (6): 1187–190. doi:10.1002/hep.21235. PMID 16729329.
  • Hensley, K (1994). "A Model for β-Amyloid Aggregation and Neurotoxicity Based on Free Radical Generation by the Peptide: Relevance to Alzheimer Disease". Proceedings of the National Academy of Sciences. 91 (8): 3270–274. Bibcode:1994PNAS...91.3270H. doi:10.1073/pnas.91.8.3270. PMC 43558. PMID 8159737.
  • Herbert, M. R. (2006) "Autism and Environmental Genomics." NeuroToxicology, pp. 671–84. Web.
  • Hodge, A. Trevor (2002) Roman Aqueducts and Water Supply. London: Duckworth.
  • How, C (2003). "Tetrodotoxin Poisoning". The American Journal of Emergency Medicine. 21 (1): 51–54. doi:10.1053/ajem.2003.50008. PMID 12563582.
  • Hue, Bernard; Buckingham, Steven D.; Buckingham, David; Sattelle, David B. (2007). "Actions of Snake Neurotoxins on an Insect Nicotinic Cholinergic Synapse". Invertebrate Neuroscience. 7 (3): 173–78. doi:10.1007/s10158-007-0053-3. PMID 17710455. S2CID 31599093.
  • Iadecola Constantino (1997). "Bright and Dark Sides of Nitric Oxide in Ischemic Brain Injury". Trends in Neurosciences. 20 (3): 132–39. doi:10.1016/s0166-2236(96)10074-6. PMID 9061868. S2CID 45011034.
  • Jacob, Reed B.; McDougal, Owen M. (2010). "The M-superfamily of Conotoxins: a Review". Cellular and Molecular Life Sciences. 67 (1): 17–27. doi:10.1007/s00018-009-0125-0. PMC 3741454. PMID 19705062.
  • Jevtovic-Todorovic, Vesna; Hartman, Richard E.; Izumi, Yukitoshi; Benshoff, Nicholas D.; Dikranian, Krikor; Zorumski, Charles F.; Olney, John W.; Wozniak, David F. (2003). "Early Exposure to Common Anesthetic Agents Causes Widespread Neurodegeneration in the Developing Rat Brain and Persistent Learning Deficits". The Journal of Neuroscience. 23 (3): 876–82. doi:10.1523/JNEUROSCI.23-03-00876.2003. PMC 6741934. PMID 12574416.
  • Jones, K (1973). "Pattern Of Malformation In Offspring Of Chronic Alcoholic Mothers". The Lancet. 301 (7815): 1267–271. doi:10.1016/s0140-6736(73)91291-9. PMID 4126070.
  • Kiernan, Matthew C.; Isbister, Geoffrey K.; Cindy; Lin, S.-Y.; Burke, David; Bostock, Hugh (2005). "Acute Tetrodotoxin-induced Neurotoxicity after Ingestion of Puffer Fish". Annals of Neurology. 57 (3): 339–48. doi:10.1002/ana.20395. PMID 15732107. S2CID 40455149.
  • King, Steven W.; Savory, John; Wills, Michael R.; Gitelman, H. J. (1981). "The Clinical Biochemistry of Aluminum". Critical Reviews in Clinical Laboratory Sciences. 14 (1): 1–20. doi:10.3109/10408368109105861. PMID 7016437.
  • Konopacka, Agnieszka; Konopacki, Filip A.; Albrecht, Jan (2009). "Protein Kinase G Is Involved in Ammonia-induced Swelling of Astrocytes". Journal of Neurochemistry. 109: 246–51. doi:10.1111/j.1471-4159.2009.05802.x. PMID 19393034. S2CID 42719321.
  • Lafon-Cazal, Mireille; Pietri, Sylvia; Culcasi, Marcel; Bockaert, Joel (1993). "NMDA-dependent Superoxide Production and Neurotoxicity". Nature. 364 (6437): 535–37. Bibcode:1993Natur.364..535L. doi:10.1038/364535a0. PMID 7687749. S2CID 4246917.
  • Lasley, SM; Green, MC; Gilbert, ME (1999). "Influence of exposure period on in vivo hippocampal glutamate and GABA release in rats chronically exposed to lead". Neurotoxicology. 20 (4): 619–29. PMID 10499360.
  • Lau, F. L.; Wong, C. K.; Yip, S. H. (1995). "Puffer Fish Poisoning". Emergency Medicine Journal. 12 (3): 214–15. doi:10.1136/emj.12.3.214. PMC 1342486. PMID 8581253.
  • Lewendon, G.; Kinra, S.; Nelder, R.; Cronin, T. (2001). "Should Children with Developmental and Behavioural Problems Be Routinely Screened for Lead?". Archives of Disease in Childhood. 85 (4): 286–88. doi:10.1136/adc.85.4.286. PMC 1718950. PMID 11567935.
  • Lidsky Theodore I (2003). "Lead Neurotoxicity in Children: Basic Mechanisms and Clinical Correlates". Brain. 126 (1): 5–19. doi:10.1093/brain/awg014. PMID 12477693.
  • Liu, Kuang-Kai; Chen, Mei-Fang; Chen, Po-Yi; Lee, Tony J F.; Cheng, Chia-Liang; Chang, Chia-Ching; Ho, Yen-Peng; Jui-I, Chao (2008). "Alpha-bungarotoxin Binding to Target Cell in a Developing Visual System by Carboxylated Nanodiamond". Nanotechnology. 19 (20): 205102. Bibcode:2008Nanot..19t5102L. doi:10.1088/0957-4484/19/20/205102. PMID 21825732. S2CID 1260143.
  • Liu, Yuan; McDermott, Suzanne; Lawson, Andrew; Aelion, C. Marjorie (2010). "The Relationship between Mental Retardation and Developmental Delays in Children and the Levels of Arsenic, Mercury and Lead in Soil Samples Taken near Their Mother's Residence during Pregnancy". International Journal of Hygiene and Environmental Health. 213 (2): 116–23. doi:10.1016/j.ijheh.2009.12.004. PMC 2836425. PMID 20045663.
  • Lorenzo, A (1994). "β-Amyloid Neurotoxicity Requires Fibril Formation and Is Inhibited by Congo Red". Proceedings of the National Academy of Sciences. 91 (25): 12243–2247. Bibcode:1994PNAS...9112243L. doi:10.1073/pnas.91.25.12243. PMC 45413. PMID 7991613.
  • Lotti, Marcello, and Angelo Moretto (1989) "Organophosphate-Induced Delayed Polyneuropathy." Toxicological Reviews, 24 (1) (2005): 37–49.
  • Martinez-Finley, Ebany J.; Goggin, Samantha L.; Labrecque, Matthew T.; Allan, Andrea M. (2011). "Reduced Expression of MAPK/ERK Genes in Perinatal Arsenic-exposed Offspring Induced by Glucocorticoid Receptor Deficits". Neurotoxicology and Teratology. 33 (5): 530–37. doi:10.1016/j.ntt.2011.07.003. PMC 3183307. PMID 21784148.
  • Martini, Frederic, Michael J. Timmons, and Robert B. Tallitsch (2009) Human Anatomy. San Francisco: Pearson/Benjamin Cummings.
  • Matsuoka, Masato; Igisu, Hideki; Kohriyama, Kazuaki; Inoue, Naohide (1991). "Suppression of Neurotoxicity of Ammonia by L-carnitine". Brain Research. 567 (2): 328–31. doi:10.1016/0006-8993(91)90814-c. PMID 1817738. S2CID 10250413.
  • McCleskey, E. W. (1987). "Omega-conotoxin: Direct and Persistent Blockade of Specific Types of Calcium Channels in Neurons but Not Muscle". Proceedings of the National Academy of Sciences. 84 (12): 4327–331. Bibcode:1987PNAS...84.4327M. doi:10.1073/pnas.84.12.4327. PMC 305078. PMID 2438698.
  • Meldrum, B.; Garthwaite, J. (1990). "Excitatory Amino Acid Neurotoxicity and Neurodegenerative Disease". Trends in Pharmacological Sciences. 11 (9): 379–87. doi:10.1016/0165-6147(90)90184-a. PMID 2238094.
  • Metcalf, James S.; Codd, Geoffrey A. (2009). "Cyanobacteria, neurotoxins and water resources: Are there implications for human neurodegenerative disease?". Amyotrophic Lateral Sclerosis. 10: 74–78. doi:10.3109/17482960903272942. PMID 19929737. S2CID 41880444.
  • Mitchell, J.Jean; Paiva, Michael; Barrow Heaton, Marieta (1999). "The Antioxidants Vitamin E and β-Carotene Protect Against Ethanol-Induced Neurotoxicity in Embryonic Rat Hippocampal Cultures". Alcohol. 17 (2): 163–68. doi:10.1016/s0741-8329(98)00051-2. PMID 10064385.
  • Moore RE (1977). "Toxins from blue-green algae". BioScience. 27 (12): 797–802. doi:10.2307/1297756. JSTOR 1297756.
  • Montecucco C (1986). "How Do Tetanus and Botulinum Toxins Bind to Neuronal Membranes?". Trends in Biochemical Sciences. 11 (8): 314–17. doi:10.1016/0968-0004(86)90282-3.
  • Morris, Stephanie A., David W. Eaves, Aleksander R. Smith, and Kimberly Nixon (2009) "Alcohol Inhibition of Neurogenesis: A Mechanism of Hippocampal Neurodegeneration in an Adolescent Alcohol Abuse Model." Hippocampus: NA.
  • Nadler; Victor, J.; Perry, Bruce W.; Cotman, Carl W. (1978). "Intraventricular Kainic Acid Preferentially Destroys Hippocampal Pyramidal Cells". Nature. 271 (5646): 676–77. Bibcode:1978Natur.271..676N. doi:10.1038/271676a0. PMID 625338. S2CID 4267990.
  • National Center for Environmental Assessment (2006) "Toxicological Reviews of Cyanobacterial Toxins: Anatoxin-a" NCEA-C-1743
  • Norenberg, M. D.; Rao, K. V. Rama; Jayakumar, A. R. (2004). "Ammonia Neurotoxicity and the Mitochondrial Permeability Transition". Journal of Bioenergetics and Biomembranes. 36 (4): 303–07. doi:10.1023/b:jobb.0000041758.20071.19. PMID 15377862. S2CID 39221153.
  • Olivera, Baldomero M.; Cruz, Lourdes J.; De Santos, Victoria; LeCheminant, Garth; Griffin, David; Zeikus, Regina; McIntosh, J. Michael; Galyean, Robert; Varga, Janos (1987). "Neuronal Calcium Channel Antagonists. Discrimination between Calcium Channel Subtypes Using .omega.-conotoxin from Conus Magus Venom". Biochemistry. 26 (8): 2086–090. doi:10.1021/bi00382a004. PMID 2441741.
  • Olney, John W (2002). "New Insights and New Issues in Developmental Neurotoxicology". NeuroToxicology. 23 (6): 659–68. doi:10.1016/s0161-813x(01)00092-4. PMID 12520755.
  • Pirazzini, Marco, Ornella Rossetto, Paolo Bolognese, Clifford C. Shone, and Cesare Montecucco (2011) "Double Anchorage to the Membrane and Intact Inter-chain Disulfide Bond Are Required for the Low PH Induced Entry of Tetanus and Botulinum Neurotoxins into Neurons." Cellular Microbiology: No. Print.
  • Rabe, Ausma; He Lee, Moon; Shek, Judy; Wisniewski, Henryk M. (1982). "Learning Deficit in Immature Rabbits with Aluminum-induced Neurofibrillary Changes". Experimental Neurology. 76 (2): 441–46. doi:10.1016/0014-4886(82)90220-5. PMID 6896495. S2CID 7495252.
  • Rocha, R. A.; Gimeno-Alcaniz, J. V.; Martín-Ibanez, Raymond; Canals, J. M.; Vélez, D.; Devesa, V. (2011). "Arsenic and Fluoride Induce Neural Progenitor Cell Apoptosis". Toxicology Letters. 203 (3): 237–44. doi:10.1016/j.toxlet.2011.03.023. PMID 21439358.
  • Roed, A (1989). "The Effects of Tetraethylammonium during Twitch and Tetanic Stimulation of the Phrenic Nerve Diaphragm Preparation in the Rat". Neuropharmacology. 28 (6): 585–92. doi:10.1016/0028-3908(89)90137-8. PMID 2547180. S2CID 1916154.
  • Rothman, S.; Thurston, J.; Hauhart, R. (1987). "Delayed Neurotoxicity of Excitatory Amino Acids In Vitro". Neuroscience. 22 (2): 471–80. doi:10.1016/0306-4522(87)90347-2. PMID 3670595. S2CID 6038419.
  • Schlesinger, Edward B (1946). "Curare A Review of Its Therapeutic Effects and Their Physiological Basis". The American Journal of Medicine. 1 (5): 518–30. doi:10.1016/0002-9343(46)90073-3. PMID 21001466.
  • Siebler, M.; Koller, H.; Schmalenbach, C.; Muller, H. (1988). "GABA Activated Chloride Currents in Cultured Rat Hippocampal and Septal Region Neurons Can Be Inhibited by Curare and Atropine". Neuroscience Letters. 93 (2–3): 220–24. doi:10.1016/0304-3940(88)90085-7. PMID 2468110. S2CID 10645685.
  • Simpson, L. L. (1986). "Molecular Pharmacology of Botulinum Toxin and Tetanus Toxin". Annual Review of Pharmacology and Toxicology. 26 (1): 427–53. doi:10.1146/annurev.pharmtox.26.1.427. PMID 3521461.
  • Soroceanu, Liliana; Gillespie, Yancey; Khazaeli, M. B.; Sontheimer, Harold (1998). "Use of Chlorotoxin for Targeting of Primary Brain Tumors". Cancer Research. 58 (21): 4871–879. PMID 9809993.
  • Spencer PS, Schaumburg HH, Ludolph AC (Eds) (2000) Experimental and Clinical Neurotoxicology. Oxford University Press, Oxford, pp. 1310.
  • Stanfield, Peter R (1983). "Tetraethylammonium Ions and the Potassium Permeability of Excitable Cells". Reviews of Physiology, Biochemistry and Pharmacology. 97: 1–49. doi:10.1007/bfb0035345. ISBN 978-3-540-11701-8. PMID 6306751.
  • Stewart I, Seawright AA, Shaw GR (2008). "Cyanobacterial poisoning in livestock, wild mammals and birds – an overview" (PDF). Cyanobacterial Harmful Algal Blooms: State of the Science and Research Needs. Advances in Experimental Medicine and Biology. 619: 613–637. doi:10.1007/978-0-387-75865-7_28. ISBN 978-0-387-75864-0. PMID 18461786.
  • Taffe, M. A.; Kotzebue, R. W.; Crean, R. D.; Crawford, E. F.; Edwards, S.; Mandyam, C. D. (2010). "From the Cover: Long-lasting Reduction in Hippocampal Neurogenesis by Alcohol Consumption in Adolescent Nonhuman Primates". Proceedings of the National Academy of Sciences. 107 (24): 11104–1109. Bibcode:2010PNAS..10711104T. doi:10.1073/pnas.0912810107. PMC 2890755. PMID 20534463.
  • Takadera, Tsuneo; Suzuki, Risa; Mohri, Tetsuro (1990). "Protection by Ethanol of Cortical Neurons from N-methyl-d-aspartate-induced Neurotoxicity Is Associated with Blocking Calcium Influx". Brain Research. 537 (1–2): 109–14. doi:10.1016/0006-8993(90)90346-d. PMID 1982237. S2CID 22160700.
  • Thyagarajan, B.; Krivitskaya, N.; Potian, J. G.; Hognason, K.; Garcia, C. C.; McArdle, J. J. (2009). "Capsaicin Protects Mouse Neuromuscular Junctions from the Neuroparalytic Effects of Botulinum Neurotoxin A." Journal of Pharmacology and Experimental Therapeutics. 331 (2): 361–71. doi:10.1124/jpet.109.156901. PMC 2775269. PMID 19654265.
  • Tymianski, Michael; Charlton, Milton P.; Carlen, Peter L.; Tator, Charles H. (2003). "Source Specificity of Early Calcium Neurotoxicity in Cultured Embryonic Spinal Neurons". The Journal of Neuroscience. 13 (5): 2095–104. doi:10.1523/JNEUROSCI.13-05-02085.1993. PMC 6576557. PMID 8097530.
  • Tsetlin, V.I; Hucho, F. (2004). "Snake and Snail Toxins Acting on Nicotinic Acetylcholine Receptors: Fundamental Aspects and Medical Applications". FEBS Letters. 557 (1–3): 9–13. doi:10.1016/s0014-5793(03)01454-6. PMID 14741333.
  • USEPA (United States Environmental Protection Agency) (1998) Health Effects Test Guidelines. OPPTS 870.6200. Neurotoxicity screening battery. Washington DC, USEPA.
  • Vahidnia, A., G.B. Van Der Voet, and F.A. De Wolff (2007) "Arsenic Neurotoxicity A Review." Human & Experimental Toxicology, 26 (10) : 823–32.
  • Walton, J (2006). "Aluminum in Hippocampal Neurons from Humans with Alzheimer's Disease". NeuroToxicology. 27 (3): 385–94. doi:10.1016/j.neuro.2005.11.007. PMID 16458972.
  • Widmaier, Eric P., Hershel Raff, Kevin T. Strang, and Arthur J. Vander (2008) Vander's Human Physiology: the Mechanisms of Body Function. Boston: McGraw-Hill Higher Education.
  • Williamson, Lura C.; Halpern, Jane L.; Montecucco, Cesare; Brown, J. E.; Neale, Elaine A. (1996). "Clostridial Neurotoxins and Substrate Proteolysis in Intact Neurons". The Journal of Biological Chemistry. 271 (13): 7694–699. doi:10.1074/jbc.271.13.7694. PMID 8631808.
  • Wood S. A.; Rasmussen J. P.; Holland P. T.; Campbell R.; Crowe A. L. M. (2007). "First Report of the Cyanotoxin Anatoxin-A from Aphanizomenon issatschenkoi (cyanobacteria)". Journal of Phycology. 43 (2): 356–365. doi:10.1111/j.1529-8817.2007.00318.x. S2CID 84284928.
  • Yamada, Kiyofumi; Tanaka, Tomoko; Han, Daiken; Senzaki, Kouji; Kameyama, Tsutomu; Nabeshima, Toshitaka (1999). "Protective Effects of Idebenone and α-tocopherol on β-amyloid-(1–42)-induced Learning and Memory Deficits in Rats: Implication of Oxidative Stress in β-amyloid-induced Neurotoxicity In vivo". European Journal of Neuroscience. 11 (1): 83–90. doi:10.1046/j.1460-9568.1999.00408.x. PMID 9987013. S2CID 84408095.
  • Yan; Du, Shi; Chen, Xi; Fu, Jin; Chen, Ming; Zhu, Huaijie; Roher, Alex; Slattery, Timothy; Zhao, Lei; Nagashima, Mariko; Morser, John; Migheli, Antonio; Nawroth, Peter; Stern, David; Marie Schmidt, Ann (1996). "RAGE and Amyloid-β Peptide Neurotoxicity in Alzheimer's Disease". Nature. 382 (6593): 685–91. Bibcode:1996Natur.382..685Y. doi:10.1038/382685a0. PMID 8751438. S2CID 4372023.
  • Yang, X (2007) Occurrence of the cyanobacterial neurotoxin, anatoxin-a, in New York State waters ProQuest. ISBN 978-0-549-35451-2.
  • Zhang, J.; Dawson, V.; Dawson, T.; Snyder, S. (1994). "Nitric Oxide Activation of Poly(ADP-ribose) Synthetase in Neurotoxicity". Science. 263 (5147): 687–89. Bibcode:1994Sci...263..687Z. doi:10.1126/science.8080500. PMID 8080500.

Further reading

  • at The Society for Neuroscience
  • at University of Texas Medical School
  • In Vitro Neurotoxicology: An Introduction at Springerlink
  • Biology of the NMDA Receptor at NCBI
  • Advances in the Neuroscience of Addiction, 2nd edition at NCBI

External links

  • Environmental Protection Agency at United States Environmental Protection Agency
  • at Oxford Medical Journals
  • Neurotoxicology at Elsevier Journals
  • Neurotoxin Institute at Neurotoxin Institute
  • Neurotoxins] at Toxipedia

neurotoxin, neurotoxicology, redirects, here, scientific, journal, neurotoxicology, journal, confused, with, nerve, agent, toxins, that, destructive, nerve, tissue, causing, neurotoxicity, extensive, class, exogenous, chemical, neurological, insults, that, adv. Neurotoxicology redirects here For the scientific journal see Neurotoxicology journal Not to be confused with nerve agent Neurotoxins are toxins that are destructive to nerve tissue causing neurotoxicity 3 Neurotoxins are an extensive class of exogenous chemical neurological insults 4 that can adversely affect function in both developing and mature nervous tissue 5 The term can also be used to classify endogenous compounds which when abnormally contacted can prove neurologically toxic 4 Though neurotoxins are often neurologically destructive their ability to specifically target neural components is important in the study of nervous systems 6 Common examples of neurotoxins include lead 7 ethanol drinking alcohol 8 glutamate 9 nitric oxide 10 botulinum toxin e g Botox 11 tetanus toxin 12 and tetrodotoxin 6 Some substances such as nitric oxide and glutamate are in fact essential for proper function of the body and only exert neurotoxic effects at excessive concentrations Neurotoxins can be found in a number of organisms including some strains of cyanobacteria 1 that can be found in algal blooms or washed up on shore in a green scum 2 Neurotoxins inhibit neuron control over ion concentrations across the cell membrane 6 or communication between neurons across a synapse 13 Local pathology of neurotoxin exposure often includes neuron excitotoxicity or apoptosis 14 but can also include glial cell damage 15 Macroscopic manifestations of neurotoxin exposure can include widespread central nervous system damage such as intellectual disability 5 persistent memory impairments 16 epilepsy and dementia 17 Additionally neurotoxin mediated peripheral nervous system damage such as neuropathy or myopathy is common Support has been shown for a number of treatments aimed at attenuating neurotoxin mediated injury such as antioxidant 8 and antitoxin 18 administration Contents 1 Background 2 Applications in neuroscience 3 Mechanisms of activity 3 1 Inhibitors 3 1 1 Sodium channel 3 1 1 1 Tetrodotoxin 3 1 2 Potassium channel 3 1 2 1 Tetraethylammonium 3 1 3 Chloride channel 3 1 3 1 Chlorotoxin 3 1 4 Calcium channel 3 1 4 1 Conotoxin 3 1 5 Synaptic vesicle release 3 1 5 1 Botulinum toxin 3 1 5 2 Tetanus toxin 3 1 6 Blood brain barrier 3 1 6 1 Aluminium 3 1 6 2 Mercury 3 2 Receptor agonists and antagonists 3 2 1 Anatoxin a 3 2 2 Bungarotoxin 3 2 2 1 Caramboxin 3 2 2 2 Curare 3 3 Cytoskeleton interference 3 3 1 Ammonia 3 3 2 Arsenic 3 4 Calcium mediated cytotoxicity 3 4 1 Lead 3 5 Neurotoxins with multiple effects 3 5 1 Ethanol 3 5 2 n Hexane 3 6 Receptor selective neurotoxins 3 6 1 MPP 3 7 Endogenous neurotoxin sources 3 7 1 Nitric oxide 3 7 2 Glutamate 4 See also 5 Notes 6 References 7 Further reading 8 External linksBackground Edit Illustration of typical multipolar neuron Exposure to neurotoxins in society is not new 19 as civilizations have been exposed to neurologically destructive compounds for thousands of years One notable example is the possible significant lead exposure during the Roman Empire resulting from the development of extensive plumbing networks and the habit of boiling vinegared wine in lead pans to sweeten it the process generating lead acetate known as sugar of lead 20 In part neurotoxins have been part of human history because of the fragile and susceptible nature of the nervous system making it highly prone to disruption The nervous tissue found in the brain spinal cord and periphery comprises an extraordinarily complex biological system that largely defines many of the unique traits of individuals As with any highly complex system however even small perturbations to its environment can lead to significant functional disruptions Properties leading to the susceptibility of nervous tissue include a high surface area of neurons a high lipid content which retains lipophilic toxins high blood flow to the brain inducing increased effective toxin exposure and the persistence of neurons through an individual s lifetime leading to compounding of damages 21 As a result the nervous system has a number of mechanisms designed to protect it from internal and external assaults including the blood brain barrier The blood brain barrier BBB is one critical example of protection which prevents toxins and other adverse compounds from reaching the brain 22 As the brain requires nutrient entry and waste removal it is perfused by blood flow Blood can carry a number of ingested toxins however which would induce significant neuron death if they reach nervous tissue Thus protective cells termed astrocytes surround the capillaries in the brain and absorb nutrients from the blood and subsequently transport them to the neurons effectively isolating the brain from a number of potential chemical insults 22 Astrocytes surrounding capillaries in the brain to form the blood brain barrier This barrier creates a tight hydrophobic layer around the capillaries in the brain inhibiting the transport of large or hydrophilic compounds In addition to the BBB the choroid plexus provides a layer of protection against toxin absorption in the brain The choroid plexuses are vascularized layers of tissue found in the third fourth and lateral ventricles of the brain which through the function of their ependymal cells are responsible for the synthesis of cerebrospinal fluid CSF 23 Importantly through selective passage of ions and nutrients and trapping heavy metals such as lead the choroid plexuses maintain a strictly regulated environment which contains the brain and spinal cord 22 23 Choroid plexus By being hydrophobic and small or inhibiting astrocyte function some compounds including certain neurotoxins are able to penetrate into the brain and induce significant damage In modern times scientists and physicians have been presented with the challenge of identifying and treating neurotoxins which has resulted in a growing interest in both neurotoxicology research and clinical studies 24 Though clinical neurotoxicology is largely a burgeoning field extensive inroads have been made in the identification of many environmental neurotoxins leading to the classification of 750 to 1000 known potentially neurotoxic compounds 21 Due to the critical importance of finding neurotoxins in common environments specific protocols have been developed by the United States Environmental Protection Agency EPA for testing and determining neurotoxic effects of compounds USEPA 1998 Additionally in vitro systems have increased in use as they provide significant improvements over the more common in vivo systems of the past Examples of improvements include tractable uniform environments and the elimination of contaminating effects of systemic metabolism 24 In vitro systems however have presented problems as it has been difficult to properly replicate the complexities of the nervous system such as the interactions between supporting astrocytes and neurons in creating the BBB 25 To even further complicate the process of determining neurotoxins when testing in vitro neurotoxicity and cytotoxicity may be difficult to distinguish as exposing neurons directly to compounds may not be possible in vivo as it is in vitro Additionally the response of cells to chemicals may not accurately convey a distinction between neurotoxins and cytotoxins as symptoms like oxidative stress or skeletal modifications may occur in response to either 26 In an effort to address this complication neurite outgrowths either axonal or dendritic in response to applied compounds have recently been proposed as a more accurate distinction between true neurotoxins and cytotoxins in an in vitro testing environment Due to the significant inaccuracies associated with this process however it has been slow in gaining widespread support 27 Additionally biochemical mechanisms have become more widely used in neurotoxin testing such that compounds can be screened for sufficiency to induce cell mechanism interference like the inhibition of acetylcholinesterase capacity of organophosphates includes DDT and sarin gas 28 Though methods of determining neurotoxicity still require significant development the identification of deleterious compounds and toxin exposure symptoms has undergone significant improvement Applications in neuroscience EditThough diverse in chemical properties and functions neurotoxins share the common property that they act by some mechanism leading to either the disruption or destruction of necessary components within the nervous system Neurotoxins however by their very design can be very useful in the field of neuroscience As the nervous system in most organisms is both highly complex and necessary for survival it has naturally become a target for attack by both predators and prey As venomous organisms often use their neurotoxins to subdue a predator or prey very rapidly toxins have evolved to become highly specific to their target channels such that the toxin does not readily bind other targets 29 see Ion Channel toxins As such neurotoxins provide an effective means by which certain elements of the nervous system may be accurately and efficiently targeted An early example of neurotoxin based targeting used radiolabeled tetrodotoxin to assay sodium channels and obtain precise measurements about their concentration along nerve membranes 29 Likewise through isolation of certain channel activities neurotoxins have provided the ability to improve the original Hodgkin Huxley model of the neuron in which it was theorized that single generic sodium and potassium channels could account for most nervous tissue function 29 From this basic understanding the use of common compounds such as tetrodotoxin tetraethylammonium and bungarotoxins have led to a much deeper understanding of the distinct ways in which individual neurons may behave Mechanisms of activity EditAs neurotoxins are compounds which adversely affect the nervous system a number of mechanisms through which they function are through the inhibition of neuron cellular processes These inhibited processes can range from membrane depolarization mechanisms to inter neuron communication By inhibiting the ability for neurons to perform their expected intracellular functions or pass a signal to a neighboring cell neurotoxins can induce systemic nervous system arrest as in the case of botulinum toxin 13 or even nervous tissue death 30 The time required for the onset of symptoms upon neurotoxin exposure can vary between different toxins being on the order of hours for botulinum toxin 18 and years for lead 31 Neurotoxin classification NeurotoxinsNa channel inhibitors Tetrodotoxin 6 K channel inhibitors Tetraethylammonium 32 Cl channel inhibitors Chlorotoxin 33 Ca channel inhibitors Conotoxin 34 Inhibitors of synaptic vesicle release Botulinum toxin 35 Tetanus toxin 36 Blood brain barrier inhibitors Aluminium 37 Mercury 38 Receptor inhibitors antagonists Bungarotoxin 39 Curare 40 Receptor agonists Anatoxin a 41 42 Caramboxin 43 25I NBOMe 44 JWH 018 45 5 MEO DiPTCytoskeleton interference Ammonia 46 Arsenic 47 Ca mediated cytotoxicity Lead 48 Multiple effects Ethanol 49 50 N Hexane 51 Methanol 49 50 Receptor selective neurotoxins MPP Endogenous neurotoxin sources Nitric oxide 52 Glutamate 53 Dopamine 54 Inhibitors Edit Sodium channel Edit Tetrodotoxin Edit The puffer fish is known for carrying lethal amounts of tetrodotoxin Tetrodotoxin TTX is a poison produced by organisms belonging to the Tetraodontiformes order which includes the puffer fish ocean sunfish and porcupine fish 55 Within the puffer fish TTX is found in the liver gonads intestines and skin 6 56 TTX can be fatal if consumed and has become a common form of poisoning in many countries Common symptoms of TTX consumption include paraesthesia often restricted to the mouth and limbs muscle weakness nausea and vomiting 55 and often manifest within 30 minutes of ingestion 57 The primary mechanism by which TTX is toxic is through the inhibition of sodium channel function which reduces the functional capacity of neuron communication This inhibition largely affects a susceptible subset of sodium channels known as TTX sensitive TTX s which also happens to be largely responsible for the sodium current that drives the depolarization phase of neuron action potentials 6 Inhibited signaling response resulting from neuron exposure to tetrodotoxin TTX resistant TTX r is another form of sodium channel which has limited sensitivity to TTX and is largely found in small diameter axons such as those found in nociception neurons 6 When a significant level of TTX is ingested it will bind sodium channels on neurons and reduce their membrane permeability to sodium This results in an increased effective threshold of required excitatory signals in order to induce an action potential in a postsynaptic neuron 6 The effect of this increased signaling threshold is a reduced excitability of postsynaptic neurons and subsequent loss of motor and sensory function which can result in paralysis and death Though assisted ventilation may increase the chance of survival after TTX exposure there is currently no antitoxin The use of the acetylcholinesterase inhibitor Neostigmine or the muscarinic acetylcholine antagonist atropine which will inhibit parasympathetic activity however can increase sympathetic nerve activity enough to improve the chance of survival after TTX exposure 55 Potassium channel Edit Tetraethylammonium Edit Tetraethylammonium TEA is a compound that like a number of neurotoxins was first identified through its damaging effects to the nervous system and shown to have the capacity of inhibiting the function of motor nerves and thus the contraction of the musculature in a manner similar to that of curare 58 Additionally through chronic TEA administration muscular atrophy would be induced 58 It was later determined that TEA functions in vivo primarily through its ability to inhibit both the potassium channels responsible for the delayed rectifier seen in an action potential and some population of calcium dependent potassium channels 32 It is this capability to inhibit potassium flux in neurons that has made TEA one of the most important tools in neuroscience It has been hypothesized that the ability for TEA to inhibit potassium channels is derived from its similar space filling structure to potassium ions 58 What makes TEA very useful for neuroscientists is its specific ability to eliminate potassium channel activity thereby allowing the study of neuron response contributions of other ion channels such as voltage gated sodium channels 59 In addition to its many uses in neuroscience research TEA has been shown to perform as an effective treatment of Parkinson s disease through its ability to limit the progression of the disease 60 Chloride channel Edit Chlorotoxin Edit Chlorotoxin Cltx is the active compound found in scorpion venom and is primarily toxic because of its ability to inhibit the conductance of chloride channels 33 Ingestion of lethal volumes of Cltx results in paralysis through this ion channel disruption Similar to botulinum toxin Cltx has been shown to possess significant therapeutic value Evidence has shown that Cltx can inhibit the ability for gliomas to infiltrate healthy nervous tissue in the brain significantly reducing the potential invasive harm caused by tumors 61 62 Calcium channel Edit Conotoxin Edit Conotoxins represent a category of poisons produced by the marine cone snail and are capable of inhibiting the activity of a number of ion channels such as calcium sodium or potassium channels 63 64 In many cases the toxins released by the different types of cone snails include a range of different types of conotoxins which may be specific for different ion channels thus creating a venom capable of widespread nerve function interruption 63 One of the unique forms of conotoxins w conotoxin w CgTx is highly specific for Ca channels and has shown usefulness in isolating them from a system 65 As calcium flux is necessary for proper excitability of a cell any significant inhibition could prevent a large amount of functionality Significantly w CgTx is capable of long term binding to and inhibition of voltage dependent calcium channels located in the membranes of neurons but not those of muscle cells 66 Synaptic vesicle release Edit Botulinum toxin Edit Mechanism of Botulinum Toxin neurotoxicity Botulinum toxin BTX is a group of neurotoxins consisting of eight distinct compounds referred to as BTX A B C D E F G H which are produced by the bacterium Clostridium botulinum and lead to muscular paralysis 67 A notably unique feature of BTX is its relatively common therapeutic use in treating dystonia and spasticity disorders 67 as well as in inducing muscular atrophy 11 despite being the most poisonous substance known 18 BTX functions peripherally to inhibit acetylcholine ACh release at the neuromuscular junction through degradation of the SNARE proteins required for ACh vesicle membrane fusion 35 As the toxin is highly biologically active an estimated dose of 1mg kg body weight is sufficient to induce an insufficient tidal volume and resultant death by asphyxiation 13 Due to its high toxicity BTX antitoxins have been an active area of research It has been shown that capsaicin active compound responsible for heat in chili peppers can bind the TRPV1 receptor expressed on cholinergic neurons and inhibit the toxic effects of BTX 18 Tetanus toxin Edit Tetanus neurotoxin TeNT is a compound that functionally reduces inhibitory transmissions in the nervous system resulting in muscular tetany TeNT is similar to BTX and is in fact highly similar in structure and origin both belonging to the same category of clostridial neurotoxins 12 Like BTX TeNT inhibits inter neuron communication by means of vesicular neurotransmitter NT release 36 One notable difference between the two compounds is that while BTX inhibits muscular contractions TeNT induces them Though both toxins inhibit vesicle release at neuron synapses the reason for this different manifestation is that BTX functions mainly in the peripheral nervous system PNS while TeNT is largely active in the central nervous system CNS 68 This is a result of TeNT migration through motor neurons to the inhibitory neurons of the spinal cord after entering through endocytosis 69 This results in a loss of function in inhibitory neurons within the CNS resulting in systemic muscular contractions Similar to the prognosis of a lethal dose of BTX TeNT leads to paralysis and subsequent suffocation 69 Blood brain barrier Edit Aluminium Edit Neurotoxic behavior of Aluminium is known to occur upon entry into the circulatory system where it can migrate to the brain and inhibit some of the crucial functions of the blood brain barrier BBB 37 A loss of function in the BBB can produce significant damage to the neurons in the CNS as the barrier protecting the brain from other toxins found in the blood will no longer be capable of such action Though the metal is known to be neurotoxic effects are usually restricted to patients incapable of removing excess ions from the blood such as those experiencing renal failure 70 Patients experiencing aluminium toxicity can exhibit symptoms such as impaired learning and reduced motor coordination 71 Additionally systemic aluminium levels are known to increase with age and have been shown to correlate with Alzheimer s Disease implicating it as a neurotoxic causative compound of the disease 72 Despite its known toxicity in its ionic form studies are divided on the potential toxicity of using aluminium in packaging and cooking appliances Mercury Edit Mercury is capable of inducing CNS damage by migrating into the brain by crossing the BBB 38 Mercury exists in a number of different compounds though methylmercury MeHg dimethylmercury and diethylmercury are the only significantly neurotoxic forms Diethylmercury and dimethylmercury are considered some of the most potent neurotoxins ever discovered 38 MeHg is usually acquired through consumption of seafood as it tends to concentrate in organisms high on the food chain 73 It is known that the mercuric ion inhibits amino acid AA and glutamate Glu transport potentially leading to excitotoxic effects 74 Receptor agonists and antagonists Edit Anatoxin a Edit External video Very Fast Death FactorUniversity of Nottingham Anatoxin a Investigations into anatoxin a also known as Very Fast Death Factor began in 1961 following the deaths of cows that drank from a lake containing an algal bloom in Saskatchewan Canada 41 42 It is a cyanotoxin produced by at least four different genera of cyanobacteria and has been reported in North America Europe Africa Asia and New Zealand 75 Toxic effects from anatoxin a progress very rapidly because it acts directly on the nerve cells neurons The progressive symptoms of anatoxin a exposure are loss of coordination twitching convulsions and rapid death by respiratory paralysis The nerve tissues which communicate with muscles contain a receptor called the nicotinic acetylcholine receptor Stimulation of these receptors causes a muscular contraction The anatoxin a molecule is shaped so it fits this receptor and in this way it mimics the natural neurotransmitter normally used by the receptor acetylcholine Once it has triggered a contraction anatoxin a does not allow the neurons to return to their resting state because it is not degraded by cholinesterase which normally performs this function As a result the muscle cells contract permanently the communication between the brain and the muscles is disrupted and breathing stops 76 77 When it was first discovered the toxin was called the Very Fast Death Factor VFDF because when it was injected into the body cavity of mice it induced tremors paralysis and death within a few minutes In 1977 the structure of VFDF was determined as a secondary bicyclic amine alkaloid and it was renamed anatoxin a 78 79 Structurally it is similar to cocaine 80 There is continued interest in anatoxin a because of the dangers it presents to recreational and drinking waters and because it is a particularly useful molecule for investigating acetylcholine receptors in the nervous system 81 The deadliness of the toxin means that it has a high military potential as a toxin weapon 82 Bungarotoxin Edit Bungarotoxin is a compound with known interaction with nicotinic acetylcholine receptors nAChRs which constitute a family of ion channels whose activity is triggered by neurotransmitter binding 83 Bungarotoxin is produced in a number of different forms though one of the commonly used forms is the long chain alpha form a bungarotoxin which is isolated from the banded krait snake 39 Though extremely toxic if ingested a bungarotoxin has shown extensive usefulness in neuroscience as it is particularly adept at isolating nAChRs due to its high affinity to the receptors 39 As there are multiple forms of bungarotoxin there are different forms of nAChRs to which they will bind and a bungarotoxin is particularly specific for a7 nAChR 84 This a7 nAChR functions to allow calcium ion influx into cells and thus when blocked by ingested bungarotoxin will produce damaging effects as ACh signaling will be inhibited 84 Likewise the use of a bungarotoxin can be very useful in neuroscience if it is desirable to block calcium flux in order to isolate effects of other channels Additionally different forms of bungarotoxin may be useful for studying inhibited nAChRs and their resultant calcium ion flow in different systems of the body For example a bungarotoxin is specific for nAChRs found in the musculature and k bungarotoxin is specific for nAChRs found in neurons 85 Caramboxin Edit Caramboxin Caramboxin CBX is a toxin found in star fruit Averrhoa carambola Individuals with some types of kidney disease are susceptible to adverse neurological effects including intoxication seizures and even death after eating star fruit or drinking juice made of this fruit Caramboxin is a new nonpeptide amino acid toxin that stimulate the glutamate receptors in neurons Caramboxin is an agonist of both NMDA and AMPA glutamatergic ionotropic receptors with potent excitatory convulsant and neurodegenerative properties 43 Curare Edit The term curare is ambiguous because it has been used to describe a number of poisons which at the time of naming were understood differently from present day understandings In the past the characterization has meant poisons used by South American tribes on arrows or darts though it has matured to specify a specific categorization of poisons which act on the neuromuscular junction to inhibit signaling and thus induce muscle relaxation 86 The neurotoxin category contains a number of distinct poisons though all were originally purified from plants originating in South America 86 The effect with which injected curare poison is usually associated is muscle paralysis and resultant death 87 Curare notably functions to inhibit nicotinic acetylcholine receptors at the neuromuscular junction Normally these receptor channels allow sodium ions into muscle cells to initiate an action potential that leads to muscle contraction By blocking the receptors the neurotoxin is capable of significantly reducing neuromuscular junction signaling an effect which has resulted in its use by anesthesiologists to produce muscular relaxation 88 Cytoskeleton interference Edit Ammonia Edit An Astrocyte a cell notable for maintaining the blood brain barrier Ammonia toxicity is often seen through two routes of administration either through consumption or through endogenous ailments such as liver failure 89 90 One notable case in which ammonia toxicity is common is in response to cirrhosis of the liver which results in hepatic encephalopathy and can result in cerebral edema Haussinger 2006 This cerebral edema can be the result of nervous cell remodeling As a consequence of increased concentrations ammonia activity in vivo has been shown to induce swelling of astrocytes in the brain through increased production of cGMP Cyclic Guanosine Monophosphate within the cells which leads to Protein Kinase G mediated PKG cytoskeletal modifications 46 The resultant effect of this toxicity can be reduced brain energy metabolism and function Importantly the toxic effects of ammonia on astrocyte remodeling can be reduced through administration of L carnitine 89 This astrocyte remodeling appears to be mediated through ammonia induced mitochondrial permeability transition This mitochondrial transition is a direct result of glutamine activity a compound which forms from ammonia in vivo 91 Administration of antioxidants or glutaminase inhibitor can reduce this mitochondrial transition and potentially also astrocyte remodeling 91 Arsenic Edit Arsenic is a neurotoxin commonly found concentrated in areas exposed to agricultural runoff mining and smelting sites Martinez Finley 2011 One of the effects of arsenic ingestion during the development of the nervous system is the inhibition of neurite growth 92 which can occur both in PNS and the CNS 93 This neurite growth inhibition can often lead to defects in neural migration and significant morphological changes of neurons during development 94 often leading to neural tube defects in neonates 95 As a metabolite of arsenic arsenite is formed after ingestion of arsenic and has shown significant toxicity to neurons within about 24 hours of exposure The mechanism of this cytotoxicity functions through arsenite induced increases in intracellular calcium ion levels within neurons which may subsequently reduce mitochondrial transmembrane potential which activates caspases triggering cell death 94 Another known function of arsenite is its destructive nature towards the cytoskeleton through inhibition of neurofilament transport 47 This is particularly destructive as neurofilaments are used in basic cell structure and support Lithium administration has shown promise however in restoring some of the lost neurofilament motility 96 Additionally similar to other neurotoxin treatments the administration of certain antioxidants has shown some promise in reducing neurotoxicity of ingested arsenic 94 Calcium mediated cytotoxicity Edit Lead Edit Lead pipes and solder are common sources of ingested lead Lead is a potent neurotoxin whose toxicity has been recognized for at least thousands of years 97 Though neurotoxic effects for lead are found in both adults and young children the developing brain is particularly susceptible to lead induced harm effects which can include apoptosis and excitotoxicity 97 An underlying mechanism by which lead is able to cause harm is its ability to be transported by calcium ATPase pumps across the BBB allowing for direct contact with the fragile cells within the central nervous system 98 Neurotoxicity results from lead s ability to act in a similar manner to calcium ions as concentrated lead will lead to cellular uptake of calcium which disrupts cellular homeostasis and induces apoptosis 48 It is this intracellular calcium increase that activates protein kinase C PKC which manifests as learning deficits in children as a result of early lead exposure 48 In addition to inducing apoptosis lead inhibits interneuron signaling through the disruption of calcium mediated neurotransmitter release 99 Neurotoxins with multiple effects Edit Ethanol Edit Male baby exhibiting Fetal Alcohol Syndrome FAS As a neurotoxin ethanol has been shown to induce nervous system damage and affect the body in a variety of ways Among the known effects of ethanol exposure are both transient and lasting consequences Some of the lasting effects include long term reduced neurogenesis in the hippocampus 100 101 widespread brain atrophy 102 and induced inflammation in the brain 103 Of note chronic ethanol ingestion has additionally been shown to induce reorganization of cellular membrane constituents leading to a lipid bilayer marked by increased membrane concentrations of cholesterol and saturated fat 50 This is important as neurotransmitter transport can be impaired through vesicular transport inhibition resulting in diminished neural network function One significant example of reduced inter neuron communication is the ability for ethanol to inhibit NMDA receptors in the hippocampus resulting in reduced long term potentiation LTP and memory acquisition 49 NMDA has been shown to play an important role in LTP and consequently memory formation 104 With chronic ethanol intake however the susceptibility of these NMDA receptors to induce LTP increases in the mesolimbic dopamine neurons in an inositol 1 4 5 triphosphate IP3 dependent manner 105 This reorganization may lead to neuronal cytotoxicity both through hyperactivation of postsynaptic neurons and through induced addiction to continuous ethanol consumption It has additionally been shown that ethanol directly reduces intracellular calcium ion accumulation through inhibited NMDA receptor activity and thus reduces the capacity for the occurrence of LTP 106 In addition to the neurotoxic effects of ethanol in mature organisms chronic ingestion is capable of inducing severe developmental defects Evidence was first shown in 1973 of a connection between chronic ethanol intake by mothers and defects in their offspring 107 This work was responsible for creating the classification of fetal alcohol syndrome a disease characterized by common morphogenesis aberrations such as defects in craniofacial formation limb development and cardiovascular formation The magnitude of ethanol neurotoxicity in fetuses leading to fetal alcohol syndrome has been shown to be dependent on antioxidant levels in the brain such as vitamin E 108 As the fetal brain is relatively fragile and susceptible to induced stresses severe deleterious effects of alcohol exposure can be seen in important areas such as the hippocampus and cerebellum The severity of these effects is directly dependent upon the amount and frequency of ethanol consumption by the mother and the stage in development of the fetus 109 It is known that ethanol exposure results in reduced antioxidant levels mitochondrial dysfunction Chu 2007 and subsequent neuronal death seemingly as a result of increased generation of reactive oxidative species ROS 30 This is a plausible mechanism as there is a reduced presence in the fetal brain of antioxidant enzymes such as catalase and peroxidase 110 In support of this mechanism administration of high levels of dietary vitamin E results in reduced or eliminated ethanol induced neurotoxic effects in fetuses 8 n Hexane Edit n Hexane is a neurotoxin which has been responsible for the poisoning of several workers in Chinese electronics factories in recent years 111 112 113 51 Receptor selective neurotoxins Edit MPP Edit MPP the toxic metabolite of MPTP is a selective neurotoxin which interferes with oxidative phosphorylation in mitochondria by inhibiting complex I leading to the depletion of ATP and subsequent cell death This occurs almost exclusively in dopaminergic neurons of the substantia nigra resulting in the presentation of permanent parkinsonism in exposed subjects 2 3 days after administration Endogenous neurotoxin sources Edit Unlike most common sources of neurotoxins which are acquired by the body through ingestion endogenous neurotoxins both originate from and exert their effects in vivo Additionally though most venoms and exogenous neurotoxins will rarely possess useful in vivo capabilities endogenous neurotoxins are commonly used by the body in useful and healthy ways such as nitric oxide which is used in cell communication 114 It is often only when these endogenous compounds become highly concentrated that they lead to dangerous effects 9 Nitric oxide Edit Though nitric oxide NO is commonly used by the nervous system in inter neuron communication and signaling it can be active in mechanisms leading to ischemia in the cerebrum Iadecola 1998 The neurotoxicity of NO is based on its importance in glutamate excitotoxicity as NO is generated in a calcium dependent manner in response to glutamate mediated NMDA activation which occurs at an elevated rate in glutamate excitotoxicity 52 Though NO facilitates increased blood flow to potentially ischemic regions of the brain it is also capable of increasing oxidative stress 115 inducing DNA damage and apoptosis 116 Thus an increased presence of NO in an ischemic area of the CNS can produce significantly toxic effects Glutamate Edit Glutamate like nitric oxide is an endogenously produced compound used by neurons to perform normally being present in small concentrations throughout the gray matter of the CNS 9 One of the most notable uses of endogenous glutamate is its functionality as an excitatory neurotransmitter 53 When concentrated however glutamate becomes toxic to surrounding neurons This toxicity can be both a result of direct lethality of glutamate on neurons and a result of induced calcium flux into neurons leading to swelling and necrosis 53 Support has been shown for these mechanisms playing significant roles in diseases and complications such as Huntington s disease epilepsy and stroke 9 See also EditBabycurus toxin 1 CangitoxinNotes Edit Sivonen K 1999 Toxins produced by cyanobacteria Vesitalous 5 11 18 Scottish Government Blue Green Algae Cyanobacteria in Inland Waters Assessment and Control of Risks to Public Health Retrieved 15 December 2011 Dorland s Medical Dictionary for Health Consumers a b Spencer 2000 a b Olney 2002 a b c d e f g h Kiernan 2005 Lidsky 2003 a b c Heaton Barrow Marieta Mitchell J Jean Paiva Michael 2000 Amelioration of Ethanol Induced Neurotoxicity in the Neonatal Rat Central Nervous System by Antioxidant Therapy Alcoholism Clinical and Experimental Research 24 4 512 18 doi 10 1111 j 1530 0277 2000 tb02019 x PMID 10798588 a b c d Choi 1987 Zhang 1994 a b Rosales Raymond L Arimura Kimiyoshi Takenaga Satoshi Osame Mitsuhiro 1996 Extrafusal and Intrafusal Muscle Effects in Experimental Botulinum Toxin A Injection Muscle amp Nerve 19 4 488 96 doi 10 1002 sici 1097 4598 199604 19 4 lt 488 aid mus9 gt 3 0 co 2 8 PMID 8622728 S2CID 20849034 a b Simpson 1986 a b c Arnon 2001 Dikranian 2001 Deng 2003 Jevtovic Todorovic 2003 Nadler 1978 a b c d Thyagarajan 2009 Neurotoxins Definition Epidemiology Etiology Hodge 2002 a b Dobbs 2009 a b c Widmaier Eric P Hershel Raff Kevin T Strang and Arthur J Vander 2008 Vander s Human Physiology the Mechanisms of Body Function Boston McGraw Hill Higher Education a b Martini 2009 a b Costa 2011 Harry 1998 Gartlon 2006 Radio Nicholas M Mundy William R 2008 Developmental Neurotoxicity Testing in Vitro Models for Assessing Chemical Effects on Neurite Out growth NeuroToxicology 29 3 361 376 doi 10 1016 j neuro 2008 02 011 PMID 18403021 Lotti 2005 a b c Adams 2003 a b Brocardo 2011 Lewendon 2001 a b Haghdoost Yazdi 2011 a b DeBin 1993 McClesky 1987 a b Garcia Rodriguez 2011 a b Williamson 1996 a b Banks 1988 a b c Aschner 1990 a b c Dutertre 2006 Koller 1988 a b Carmichael 1978 a b Carmichael 1975 a b Garcia Cairasco N Moyses Neto M Del Vecchio F Oliveira J A C Dos Santos F L Castro O W Arisi G M Dantas M R Carolino R O G Coutinho Netto J Dagostin A L A Rodrigues M C A Leao R M Quintiliano S A P Silva L F Gobbo Neto L Lopes N P 2013 Elucidating the Neurotoxicity of the Star Fruit Angewandte Chemie International Edition 52 49 13067 13070 doi 10 1002 anie 201305382 PMID 24281890 Rutgrere 2012 Roller 1994 a b Konopacka 2009 a b DeFuria 2006 a b c Bressler 1999 a b c Lovinger D White G Weight F 1989 Ethanol Inhibits NMDA activated Ion Current in Hippocampal Neurons Science 243 4899 1721 724 Bibcode 1989Sci 243 1721L doi 10 1126 science 2467382 PMID 2467382 a b c Leonard B E 1986 Is Ethanol a Neurotoxin the Effects of Ethanol on Neuronal Structure and Function Alcohol and Alcoholism 21 4 325 38 doi 10 1093 oxfordjournals alcalc a044638 PMID 2434114 a b Occupational Safety and Health Guideline for n Hexane Archived 2011 12 18 at the Wayback Machine OSHA gov a b Garthwaite 1988 a b c Choi 1990 Ben Shachar D Zuk R Glinka Y 1995 Dopamine neurotoxicity inhibition of mitochondrial respiration J Neurochem 64 2 718 23 doi 10 1046 j 1471 4159 1995 64020718 x PMID 7830065 S2CID 9060404 a b c Chowdhury F R Ahasan H A M Nazmul Rashid A K M Mamunur Mamun A Al Khaliduzzaman S M 2007 Tetrodotoxin Poisoning a Clinical Analysis Role of Neostigmine and Short term Outcome of 53 Cases Singapore Medical Journal 48 9 830 33 PMID 17728964 Ahasan 2004 Lau 1995 a b c Standfield 1983 Roed 1989 Haghdoost Yasdi 2011 Deshane 2003 Soroceanu 1998 a b Jacob 2010 Olivera 1987 Cruz 1986 McCleskey 1987 a b Brin Mitchell F 1997 Botulinum Toxin Chemistry Pharmacology Toxicity and Immunology Muscle amp Nerve 20 S6 146 68 Montecucco 1986 a b Pirazzini 2011 King 1981 Rabe 1982 Walton 2006 Chan 2011 Brookes 1988 Yang 2007 Wood 2007 National Center for Environmental Assessment Devlin 1977 Moore 1977 Metcalf 2009 Stewart 2008 Dixit 2005 Tsetlin 2003 a b Liu 2008 Hue 2007 a b Bisset 1992 Schlesinger 1946 Griffith Harold R Johnson G Enid 1942 The Use Of Curare In General Anesthesia Anesthesiology 3 4 418 420 doi 10 1097 00000542 194207000 00006 S2CID 71400545 a b Matsuoka 1991 Buzanska 2000 a b Norenberg 2004 Liu 2009 full citation needed Vahidnia 2007 a b c Rocha 2011 Brender 2005 DeFuria 2007 a b Lidskey 2003 Bradbury 1993 Lasley 1999 Taffe 2010 Morris 2009 Bleich 2003 Blanco 2005 Davis 1992 Bernier 2011 Takadera 1990 Jones 1973 Mitchell 1999 Gil Mohapel 2010 Bergamini 2004 Workers poisoned while making iPhones ABC News October 25 2010 Dirty Secrets Archived 2017 05 25 at the Wayback Machine ABC Foreign Correspondent 2010 Oct 26 Mr Daisey and the Apple Factory This American Life January 6 2012 Iadecola 1998 Beckman 1990 Bonfoco 1995References EditAdams Michael E Olivera Baldomero M 1994 Neurotoxins Overview of an Emerging Research Technology Trends in Neurosciences 17 4 151 55 doi 10 1016 0166 2236 94 90092 2 PMID 7517594 S2CID 3984106 Arnon Stephen S Schechter Robert Inglesby Thomas V Henderson Donald A Bartlett John G Ascher Michael S Eitzen Edward Fine Anne D Hauer Jerome Layton Marcelle Lillibridge Scott Osterholm Michael T O Toole Tara Parker Gerald Perl Trish M Russell Philip K Swerdlow David L Tonat Kevin 2001 Botulinum Toxin as a Biological Weapon The Journal of the American Medical Association 285 8 1059 069 doi 10 1001 jama 285 8 1059 PMID 11209178 Aschner M Aschner J 1990 Mercury Neurotoxicity Mechanisms of Blood brain barrier Transport Neuroscience amp Biobehavioral Reviews 14 2 169 76 doi 10 1016 s0149 7634 05 80217 9 PMID 2190116 S2CID 8777931 Banks William A Kastin Abba J 1989 Aluminum Induced Neurotoxicity Alterations in Membrane Function at the Blood brain barrier Neuroscience amp Biobehavioral Reviews 13 1 47 53 doi 10 1016 s0149 7634 89 80051 x PMID 2671833 S2CID 46507895 Baum Baicker Cynthia 1985 The Health Benefits of Moderate Alcohol Consumption A Review of the Literature Drug and Alcohol Dependence 15 3 207 27 doi 10 1016 0376 8716 85 90001 8 PMID 4028954 Beckman J S 1990 Apparent Hydroxyl Radical Production by Peroxynitrite Implications for Endothelial Injury from Nitric Oxide and Superoxide Proceedings of the National Academy of Sciences 87 4 1620 624 Bibcode 1990PNAS 87 1620B doi 10 1073 pnas 87 4 1620 PMC 53527 PMID 2154753 Bergamini Carlo M Gambetti Stefani Dondi Alessia Cervellati Carlo 2004 Oxygen Reactive Oxygen Species and Tissue Damage Current Pharmaceutical Design 10 14 1611 626 doi 10 2174 1381612043384664 PMID 15134560 Bernier Brian E Whitaker Leslie R Morikawa Hitoshi 2011 Previous Ethanol Experience Enhances Synaptic Plasticity of NMDA Receptors in the Ventral Tegmental Area The Journal of Neuroscience 31 14 5305 212 doi 10 1523 JNEUROSCI 5282 10 2011 PMC 3086894 PMID 21471355 a href Template Cite journal html title Template Cite journal cite journal a CS1 maint multiple names authors list link Bisset Norman G 1992 War and Hunting Poisons of the New World Part 1 Notes on the Early History of Curare Journal of Ethnopharmacology 36 1 1 26 doi 10 1016 0378 8741 92 90056 w PMID 1501489 Blanco Ana M Valles Soraya L Pascual Maria Guerri Consuelo 2005 Involvement of TLR4 Type I IL 1 Receptor Signaling in the Induction of Inflammatory Mediators and Cell Death Induced by Ethanol in Cultured Astrocytes The Journal of Immunology 175 10 6893 899 doi 10 4049 jimmunol 175 10 6893 PMID 16272348 Bleich S 2003 Hyperhomocysteinemia as a New Risk Factor for Brain Shrinkage in Patients with Alcoholism Neuroscience Letters 335 3 179 82 doi 10 1016 s0304 3940 02 01194 1 PMID 12531462 S2CID 33032529 Bonfoco E 1995 Apoptosis and Necrosis Two Distinct Events Induced Respectively by Mild and Intense Insults with N Methyl D Aspartate or Nitric Oxide Superoxide in Cortical Cell Cultures Proceedings of the National Academy of Sciences 92 16 7162 166 Bibcode 1995PNAS 92 7162B doi 10 1073 pnas 92 16 7162 PMC 41299 PMID 7638161 Bradbury MW Deane R 1993 Permeability of the blood brain barrier to lead Review Neurotoxicology 14 2 3 131 6 PMID 8247388 Brender J Suarez L Felkner M Gilani Z Stinchcomb D Moody K Henry J Hendricks K 2006 Maternal Exposure to Arsenic Cadmium Lead and Mercury and Neural Tube Defects in Offspring Environmental Research 101 1 132 39 Bibcode 2006ER 101 132B doi 10 1016 j envres 2005 08 003 PMID 16171797 Bressler J Kim KA Chakraborti T Goldstein G 1999 Molecular mechanisms of lead neurotoxicity Review Neurochem Res 24 4 595 600 doi 10 1023 A 1022596115897 PMID 10227691 S2CID 3017703 Brocardo Patricia S Gil Mohapel Joana Christie Brian R 2011 The Role of Oxidative Stress in Fetal Alcohol Spectrum Disorders Brain Research Reviews 67 1 2 209 25 doi 10 1016 j brainresrev 2011 02 001 PMID 21315761 S2CID 1213492 Brookes N 1988 Specificity and Reversibility of the Inhibition by HgCl of Glutamate Transport in Astrocyte Cultures Journal of Neurochemistry 50 4 1117 122 doi 10 1111 j 1471 4159 1988 tb10581 x PMID 2894409 S2CID 27522735 Buzanska L Zablocka B Dybel A Domanska Janik K Albrecht J 2000 Delayed Induction of Apoptosis by Ammonia in C6 Glioma Cells Neurochemistry International 37 2 3 287 97 doi 10 1016 s0197 0186 00 00030 9 PMID 10812214 S2CID 33351533 Carmichael WW Biggs DF Gorham PR 1975 Toxicology and pharmacological action of Anabaena flos aquae toxin Science 187 4176 542 544 Bibcode 1975Sci 187 542C doi 10 1126 science 803708 PMID 803708 Carmichael WW Gorham PR 1978 Anatoxins from clones of Anabaena flos aquae isolated from lakes of western Canada Mitt Infernal Verein Limnol 21 285 295 Chan H M 2011 Mercury in Fish Human Health Risks Encyclopedia of Environmental Health 697 704 Choi D 1988 Calcium mediated Neurotoxicity Relationship to Specific Channel Types and Role in Ischemic Damage Trends in Neurosciences 11 10 465 69 doi 10 1016 0166 2236 88 90200 7 PMID 2469166 S2CID 36909560 Choi D W Rothman S M 1990 The Role of Glutamate Neurotoxicity in Hypoxic Ischemic Neuronal Death Annual Review of Neuroscience 13 1 171 82 doi 10 1146 annurev neuro 13 1 171 PMID 1970230 Choi Dennis W 1987 Ionic Dependence of Glutamate Neurotoxicity The Journal of Neuroscience 7 2 369 79 doi 10 1523 JNEUROSCI 07 02 00369 1987 PMC 6568907 PMID 2880938 Choi Dennis W Maulucci Gedde Margaret Kriegstein Arnold R 1987 Glutamate Neurotoxicity in Cortical Cell Culture The Journal of Neuroscience 7 2 357 68 doi 10 1523 JNEUROSCI 07 02 00357 1987 PMC 6568898 PMID 2880937 Chu Jennifer Tong Ming Monte Suzanne M 2007 Chronic Ethanol Exposure Causes Mitochondrial Dysfunction and Oxidative Stress in Immature Central Nervous System Neurons Acta Neuropathologica 113 6 659 73 doi 10 1007 s00401 007 0199 4 PMID 17431646 S2CID 25418731 Clancy Barbara Finlay Barbara L Darlington Richard B Anand K J S 2007 Extrapolating Brain Development from Experimental Species to Humans NeuroToxicology 28 5 931 37 doi 10 1016 j neuro 2007 01 014 PMC 2077812 PMID 17368774 Costa Lucio G Gennaro Giordano and Marina Guizzetti 2011 In Vitro Neurotoxicology Methods and Protocols New York Humana Coyle Joseph T Schwarcz Robert 1976 Lesion of Striatal Neurons with Kainic Acid Provides a Model for Huntington s Chorea Nature 246 5574 244 46 Bibcode 1976Natur 263 244C doi 10 1038 263244a0 PMID 8731 S2CID 4271762 Cruz Lourdes J Olivera Baldomero M 1987 Calcium Channel Antagonists w Conotoxin Defines a New High Affinity Site The Journal of Biological Chemistry 14 261 6230 233 Davis S Butcher S P Morris R 1992 The NMDA Receptor Antagonist D 2 amino 5phosphonopentanoate D AP5 Impairs Spatial Learning and LTP in Vivo at Intracerebral Concentrations Comparable to Those That Block LTP in Vitro The Journal of Neuroscience 12 1 21 34 doi 10 1523 JNEUROSCI 12 01 00021 1992 PMC 6575679 PMID 1345945 Dawson V L 1991 Nitric Oxide Mediates Glutamate Neurotoxicity in Primary Cortical Cultures Proceedings of the National Academy of Sciences 88 14 6368 371 Bibcode 1991PNAS 88 6368D doi 10 1073 pnas 88 14 6368 PMC 52084 PMID 1648740 Debin John A John E Maggio and Gary R Strichartz 1993 Purification and Characterization of Chlorotoxin a Chloride Channel Ligand from the Venom of the Scorpion The American Physiological Society pp 361 69 DeFuria Jason Shea Thomas B 2007 Arsenic Inhibits Neurofilament Transport and Induces Perikaryal Accumulation of Phosphorylated Neurofilaments Roles of JNK and GSK 3b Brain Research 1181 74 82 doi 10 1016 j brainres 2007 04 019 PMID 17961518 S2CID 37068398 Defuria Jason 2006 The Environmental Neurotoxin Arsenic Impairs Neurofilament Dynamics by Overactivation of C JUN Terminal Kinase Potential Role for Amyotrophic Lateral Sclerosis UMI pp 1 16 Deng Wenbin Poretz Ronald D 2003 Oligodendroglia in Developmental Neurotoxicity NeuroToxicology 24 2 161 78 doi 10 1016 s0161 813x 02 00196 1 PMID 12606289 Deshane Jessy Garner Craig C Sontheimer Harald 2003 Chlorotoxin Inhibits Glioma Cell Invasion via Matrix Metalloproteinase 2 The Journal of Biological Chemistry 278 6 4135 144 doi 10 1074 jbc m205662200 PMID 12454020 Devlin JP Edwards OE Gorham PR Hunter NR Pike RK Stavric B 1977 Anatoxin a a toxic alkaloid from Anabaena flos aquae NRC 44h Can J Chem 55 8 1367 1371 doi 10 1139 v77 189 Archived from the original on 2012 07 09 Dikranian K 2001 Apoptosis in the in Vivo Mammalian Forebrain Neurobiology of Disease 8 3 359 79 doi 10 1006 nbdi 2001 0411 PMID 11447994 S2CID 21965033 Dixit A Dhaked RK Alam SI Singh L 2005 Military potential of biological neurotoxins Toxin Reviews 24 2 175 207 doi 10 1081 TXR 200057850 S2CID 85651107 Dobbs Michael R 2009 Clinical Neurotoxicology Philadelphia Saunders Elsevier Dutertre S Lewis R 2006 Toxin Insights into Nicotinic Acetylcholine Receptors Biochemical Pharmacology 72 6 661 70 doi 10 1016 j bcp 2006 03 027 PMID 16716265 Garcia Rodriguez C Geren I N Lou J Conrad F Forsyth C Wen W Chakraborti S Zao H Manzanarez G Smith T J Brown J Tepp W H Liu N Wijesuriya S Tomic M T Johnson E A Smith L A Marks J D 2011 Response Re Neutralizing Human Monoclonal Antibodies Binding Multiple Serotypes of Botulinum Neurotoxin by Garcia Rodriguez Et Al PEDS 2011 24 321 331 Protein Engineering Design and Selection 24 9 633 34 doi 10 1093 protein gzr012 Garthwaite John Charles Sarah L Chess Williams Russel 1988 Endothelim derived Relaxing Factor Release on Activation of NMDA Receptors Suggests Role as Intercellular Messenger in the Brain Nature 336 24 385 88 Bibcode 1988Natur 336 385G doi 10 1038 336385a0 PMID 2904125 S2CID 4361381 Gartlon J Kinsner A Balprice A Coecke S Clothier R 2006 Evaluation of a Proposed in Vitro Test Strategy Using Neuronal and Non neuronal Cell Systems for Detecting Neurotoxicity Toxicology in Vitro 20 8 1569 581 doi 10 1016 j tiv 2006 07 009 PMID 16959468 Gil Mohapel Joana Boehme Fanny Kainer Leah Christie Brian R 2010 Hippocampal Cell Loss and Neurogenesis after Fetal Alcohol Exposure Insights from Different Rodent Models Brain Research Reviews 64 2 283 303 doi 10 1016 j brainresrev 2010 04 011 PMID 20471420 S2CID 10159395 Haghdoost Yazdi Hashem Faraji Ayda Fraidouni Negin Movahedi Mohadeseh Hadibeygi Elham Vaezi Fatemeh 2011 Significant Effects of 4 aminopyridine and Tetraethylammonium in the Treatment of 6 hydroxydopamine induced Parkinson s Disease Behavioural Brain Research 223 1 70 74 doi 10 1016 j bbr 2011 04 021 PMID 21540059 S2CID 7100177 Harry G J Billingsley Melvin Bruinink Arendd Campbell Iain L Classen Werner Dorman David C Galli Corrado Ray David Smith Robert A Tilson Hugh A 1998 In Vitro Techniques for the Assessment of Neurotoxicity Environmental Health Perspectives 106 Suppl 1 131 58 doi 10 2307 3433917 JSTOR 3433917 PMC 1533280 PMID 9539010 Haussinger Dieter 2006 Low Grade Cerebral Edema and the Pathogenesis of Hepatic Encephalopathy in Cirrhosis Hepatology 43 6 1187 190 doi 10 1002 hep 21235 PMID 16729329 Hensley K 1994 A Model for b Amyloid Aggregation and Neurotoxicity Based on Free Radical Generation by the Peptide Relevance to Alzheimer Disease Proceedings of the National Academy of Sciences 91 8 3270 274 Bibcode 1994PNAS 91 3270H doi 10 1073 pnas 91 8 3270 PMC 43558 PMID 8159737 Herbert M R 2006 Autism and Environmental Genomics NeuroToxicology pp 671 84 Web Hodge A Trevor 2002 Roman Aqueducts and Water Supply London Duckworth How C 2003 Tetrodotoxin Poisoning The American Journal of Emergency Medicine 21 1 51 54 doi 10 1053 ajem 2003 50008 PMID 12563582 Hue Bernard Buckingham Steven D Buckingham David Sattelle David B 2007 Actions of Snake Neurotoxins on an Insect Nicotinic Cholinergic Synapse Invertebrate Neuroscience 7 3 173 78 doi 10 1007 s10158 007 0053 3 PMID 17710455 S2CID 31599093 Iadecola Constantino 1997 Bright and Dark Sides of Nitric Oxide in Ischemic Brain Injury Trends in Neurosciences 20 3 132 39 doi 10 1016 s0166 2236 96 10074 6 PMID 9061868 S2CID 45011034 Jacob Reed B McDougal Owen M 2010 The M superfamily of Conotoxins a Review Cellular and Molecular Life Sciences 67 1 17 27 doi 10 1007 s00018 009 0125 0 PMC 3741454 PMID 19705062 Jevtovic Todorovic Vesna Hartman Richard E Izumi Yukitoshi Benshoff Nicholas D Dikranian Krikor Zorumski Charles F Olney John W Wozniak David F 2003 Early Exposure to Common Anesthetic Agents Causes Widespread Neurodegeneration in the Developing Rat Brain and Persistent Learning Deficits The Journal of Neuroscience 23 3 876 82 doi 10 1523 JNEUROSCI 23 03 00876 2003 PMC 6741934 PMID 12574416 Jones K 1973 Pattern Of Malformation In Offspring Of Chronic Alcoholic Mothers The Lancet 301 7815 1267 271 doi 10 1016 s0140 6736 73 91291 9 PMID 4126070 Kiernan Matthew C Isbister Geoffrey K Cindy Lin S Y Burke David Bostock Hugh 2005 Acute Tetrodotoxin induced Neurotoxicity after Ingestion of Puffer Fish Annals of Neurology 57 3 339 48 doi 10 1002 ana 20395 PMID 15732107 S2CID 40455149 King Steven W Savory John Wills Michael R Gitelman H J 1981 The Clinical Biochemistry of Aluminum Critical Reviews in Clinical Laboratory Sciences 14 1 1 20 doi 10 3109 10408368109105861 PMID 7016437 Konopacka Agnieszka Konopacki Filip A Albrecht Jan 2009 Protein Kinase G Is Involved in Ammonia induced Swelling of Astrocytes Journal of Neurochemistry 109 246 51 doi 10 1111 j 1471 4159 2009 05802 x PMID 19393034 S2CID 42719321 Lafon Cazal Mireille Pietri Sylvia Culcasi Marcel Bockaert Joel 1993 NMDA dependent Superoxide Production and Neurotoxicity Nature 364 6437 535 37 Bibcode 1993Natur 364 535L doi 10 1038 364535a0 PMID 7687749 S2CID 4246917 Lasley SM Green MC Gilbert ME 1999 Influence of exposure period on in vivo hippocampal glutamate and GABA release in rats chronically exposed to lead Neurotoxicology 20 4 619 29 PMID 10499360 Lau F L Wong C K Yip S H 1995 Puffer Fish Poisoning Emergency Medicine Journal 12 3 214 15 doi 10 1136 emj 12 3 214 PMC 1342486 PMID 8581253 Lewendon G Kinra S Nelder R Cronin T 2001 Should Children with Developmental and Behavioural Problems Be Routinely Screened for Lead Archives of Disease in Childhood 85 4 286 88 doi 10 1136 adc 85 4 286 PMC 1718950 PMID 11567935 Lidsky Theodore I 2003 Lead Neurotoxicity in Children Basic Mechanisms and Clinical Correlates Brain 126 1 5 19 doi 10 1093 brain awg014 PMID 12477693 Liu Kuang Kai Chen Mei Fang Chen Po Yi Lee Tony J F Cheng Chia Liang Chang Chia Ching Ho Yen Peng Jui I Chao 2008 Alpha bungarotoxin Binding to Target Cell in a Developing Visual System by Carboxylated Nanodiamond Nanotechnology 19 20 205102 Bibcode 2008Nanot 19t5102L doi 10 1088 0957 4484 19 20 205102 PMID 21825732 S2CID 1260143 Liu Yuan McDermott Suzanne Lawson Andrew Aelion C Marjorie 2010 The Relationship between Mental Retardation and Developmental Delays in Children and the Levels of Arsenic Mercury and Lead in Soil Samples Taken near Their Mother s Residence during Pregnancy International Journal of Hygiene and Environmental Health 213 2 116 23 doi 10 1016 j ijheh 2009 12 004 PMC 2836425 PMID 20045663 Lorenzo A 1994 b Amyloid Neurotoxicity Requires Fibril Formation and Is Inhibited by Congo Red Proceedings of the National Academy of Sciences 91 25 12243 2247 Bibcode 1994PNAS 9112243L doi 10 1073 pnas 91 25 12243 PMC 45413 PMID 7991613 Lotti Marcello and Angelo Moretto 1989 Organophosphate Induced Delayed Polyneuropathy Toxicological Reviews 24 1 2005 37 49 Martinez Finley Ebany J Goggin Samantha L Labrecque Matthew T Allan Andrea M 2011 Reduced Expression of MAPK ERK Genes in Perinatal Arsenic exposed Offspring Induced by Glucocorticoid Receptor Deficits Neurotoxicology and Teratology 33 5 530 37 doi 10 1016 j ntt 2011 07 003 PMC 3183307 PMID 21784148 Martini Frederic Michael J Timmons and Robert B Tallitsch 2009 Human Anatomy San Francisco Pearson Benjamin Cummings Matsuoka Masato Igisu Hideki Kohriyama Kazuaki Inoue Naohide 1991 Suppression of Neurotoxicity of Ammonia by L carnitine Brain Research 567 2 328 31 doi 10 1016 0006 8993 91 90814 c PMID 1817738 S2CID 10250413 McCleskey E W 1987 Omega conotoxin Direct and Persistent Blockade of Specific Types of Calcium Channels in Neurons but Not Muscle Proceedings of the National Academy of Sciences 84 12 4327 331 Bibcode 1987PNAS 84 4327M doi 10 1073 pnas 84 12 4327 PMC 305078 PMID 2438698 Meldrum B Garthwaite J 1990 Excitatory Amino Acid Neurotoxicity and Neurodegenerative Disease Trends in Pharmacological Sciences 11 9 379 87 doi 10 1016 0165 6147 90 90184 a PMID 2238094 Metcalf James S Codd Geoffrey A 2009 Cyanobacteria neurotoxins and water resources Are there implications for human neurodegenerative disease Amyotrophic Lateral Sclerosis 10 74 78 doi 10 3109 17482960903272942 PMID 19929737 S2CID 41880444 Mitchell J Jean Paiva Michael Barrow Heaton Marieta 1999 The Antioxidants Vitamin E and b Carotene Protect Against Ethanol Induced Neurotoxicity in Embryonic Rat Hippocampal Cultures Alcohol 17 2 163 68 doi 10 1016 s0741 8329 98 00051 2 PMID 10064385 Moore RE 1977 Toxins from blue green algae BioScience 27 12 797 802 doi 10 2307 1297756 JSTOR 1297756 Montecucco C 1986 How Do Tetanus and Botulinum Toxins Bind to Neuronal Membranes Trends in Biochemical Sciences 11 8 314 17 doi 10 1016 0968 0004 86 90282 3 Morris Stephanie A David W Eaves Aleksander R Smith and Kimberly Nixon 2009 Alcohol Inhibition of Neurogenesis A Mechanism of Hippocampal Neurodegeneration in an Adolescent Alcohol Abuse Model Hippocampus NA Nadler Victor J Perry Bruce W Cotman Carl W 1978 Intraventricular Kainic Acid Preferentially Destroys Hippocampal Pyramidal Cells Nature 271 5646 676 77 Bibcode 1978Natur 271 676N doi 10 1038 271676a0 PMID 625338 S2CID 4267990 National Center for Environmental Assessment 2006 Toxicological Reviews of Cyanobacterial Toxins Anatoxin a NCEA C 1743 Norenberg M D Rao K V Rama Jayakumar A R 2004 Ammonia Neurotoxicity and the Mitochondrial Permeability Transition Journal of Bioenergetics and Biomembranes 36 4 303 07 doi 10 1023 b jobb 0000041758 20071 19 PMID 15377862 S2CID 39221153 Olivera Baldomero M Cruz Lourdes J De Santos Victoria LeCheminant Garth Griffin David Zeikus Regina McIntosh J Michael Galyean Robert Varga Janos 1987 Neuronal Calcium Channel Antagonists Discrimination between Calcium Channel Subtypes Using omega conotoxin from Conus Magus Venom Biochemistry 26 8 2086 090 doi 10 1021 bi00382a004 PMID 2441741 Olney John W 2002 New Insights and New Issues in Developmental Neurotoxicology NeuroToxicology 23 6 659 68 doi 10 1016 s0161 813x 01 00092 4 PMID 12520755 Pirazzini Marco Ornella Rossetto Paolo Bolognese Clifford C Shone and Cesare Montecucco 2011 Double Anchorage to the Membrane and Intact Inter chain Disulfide Bond Are Required for the Low PH Induced Entry of Tetanus and Botulinum Neurotoxins into Neurons Cellular Microbiology No Print Rabe Ausma He Lee Moon Shek Judy Wisniewski Henryk M 1982 Learning Deficit in Immature Rabbits with Aluminum induced Neurofibrillary Changes Experimental Neurology 76 2 441 46 doi 10 1016 0014 4886 82 90220 5 PMID 6896495 S2CID 7495252 Rocha R A Gimeno Alcaniz J V Martin Ibanez Raymond Canals J M Velez D Devesa V 2011 Arsenic and Fluoride Induce Neural Progenitor Cell Apoptosis Toxicology Letters 203 3 237 44 doi 10 1016 j toxlet 2011 03 023 PMID 21439358 Roed A 1989 The Effects of Tetraethylammonium during Twitch and Tetanic Stimulation of the Phrenic Nerve Diaphragm Preparation in the Rat Neuropharmacology 28 6 585 92 doi 10 1016 0028 3908 89 90137 8 PMID 2547180 S2CID 1916154 Rothman S Thurston J Hauhart R 1987 Delayed Neurotoxicity of Excitatory Amino Acids In Vitro Neuroscience 22 2 471 80 doi 10 1016 0306 4522 87 90347 2 PMID 3670595 S2CID 6038419 Schlesinger Edward B 1946 Curare A Review of Its Therapeutic Effects and Their Physiological Basis The American Journal of Medicine 1 5 518 30 doi 10 1016 0002 9343 46 90073 3 PMID 21001466 Siebler M Koller H Schmalenbach C Muller H 1988 GABA Activated Chloride Currents in Cultured Rat Hippocampal and Septal Region Neurons Can Be Inhibited by Curare and Atropine Neuroscience Letters 93 2 3 220 24 doi 10 1016 0304 3940 88 90085 7 PMID 2468110 S2CID 10645685 Simpson L L 1986 Molecular Pharmacology of Botulinum Toxin and Tetanus Toxin Annual Review of Pharmacology and Toxicology 26 1 427 53 doi 10 1146 annurev pharmtox 26 1 427 PMID 3521461 Soroceanu Liliana Gillespie Yancey Khazaeli M B Sontheimer Harold 1998 Use of Chlorotoxin for Targeting of Primary Brain Tumors Cancer Research 58 21 4871 879 PMID 9809993 Spencer PS Schaumburg HH Ludolph AC Eds 2000 Experimental and Clinical Neurotoxicology Oxford University Press Oxford pp 1310 Stanfield Peter R 1983 Tetraethylammonium Ions and the Potassium Permeability of Excitable Cells Reviews of Physiology Biochemistry and Pharmacology 97 1 49 doi 10 1007 bfb0035345 ISBN 978 3 540 11701 8 PMID 6306751 Stewart I Seawright AA Shaw GR 2008 Cyanobacterial poisoning in livestock wild mammals and birds an overview PDF Cyanobacterial Harmful Algal Blooms State of the Science and Research Needs Advances in Experimental Medicine and Biology 619 613 637 doi 10 1007 978 0 387 75865 7 28 ISBN 978 0 387 75864 0 PMID 18461786 Taffe M A Kotzebue R W Crean R D Crawford E F Edwards S Mandyam C D 2010 From the Cover Long lasting Reduction in Hippocampal Neurogenesis by Alcohol Consumption in Adolescent Nonhuman Primates Proceedings of the National Academy of Sciences 107 24 11104 1109 Bibcode 2010PNAS 10711104T doi 10 1073 pnas 0912810107 PMC 2890755 PMID 20534463 Takadera Tsuneo Suzuki Risa Mohri Tetsuro 1990 Protection by Ethanol of Cortical Neurons from N methyl d aspartate induced Neurotoxicity Is Associated with Blocking Calcium Influx Brain Research 537 1 2 109 14 doi 10 1016 0006 8993 90 90346 d PMID 1982237 S2CID 22160700 Thyagarajan B Krivitskaya N Potian J G Hognason K Garcia C C McArdle J J 2009 Capsaicin Protects Mouse Neuromuscular Junctions from the Neuroparalytic Effects of Botulinum Neurotoxin A Journal of Pharmacology and Experimental Therapeutics 331 2 361 71 doi 10 1124 jpet 109 156901 PMC 2775269 PMID 19654265 Tymianski Michael Charlton Milton P Carlen Peter L Tator Charles H 2003 Source Specificity of Early Calcium Neurotoxicity in Cultured Embryonic Spinal Neurons The Journal of Neuroscience 13 5 2095 104 doi 10 1523 JNEUROSCI 13 05 02085 1993 PMC 6576557 PMID 8097530 Tsetlin V I Hucho F 2004 Snake and Snail Toxins Acting on Nicotinic Acetylcholine Receptors Fundamental Aspects and Medical Applications FEBS Letters 557 1 3 9 13 doi 10 1016 s0014 5793 03 01454 6 PMID 14741333 USEPA United States Environmental Protection Agency 1998 Health Effects Test Guidelines OPPTS 870 6200 Neurotoxicity screening battery Washington DC USEPA Vahidnia A G B Van Der Voet and F A De Wolff 2007 Arsenic Neurotoxicity A Review Human amp Experimental Toxicology 26 10 823 32 Walton J 2006 Aluminum in Hippocampal Neurons from Humans with Alzheimer s Disease NeuroToxicology 27 3 385 94 doi 10 1016 j neuro 2005 11 007 PMID 16458972 Widmaier Eric P Hershel Raff Kevin T Strang and Arthur J Vander 2008 Vander s Human Physiology the Mechanisms of Body Function Boston McGraw Hill Higher Education Williamson Lura C Halpern Jane L Montecucco Cesare Brown J E Neale Elaine A 1996 Clostridial Neurotoxins and Substrate Proteolysis in Intact Neurons The Journal of Biological Chemistry 271 13 7694 699 doi 10 1074 jbc 271 13 7694 PMID 8631808 Wood S A Rasmussen J P Holland P T Campbell R Crowe A L M 2007 First Report of the Cyanotoxin Anatoxin A from Aphanizomenon issatschenkoi cyanobacteria Journal of Phycology 43 2 356 365 doi 10 1111 j 1529 8817 2007 00318 x S2CID 84284928 Yamada Kiyofumi Tanaka Tomoko Han Daiken Senzaki Kouji Kameyama Tsutomu Nabeshima Toshitaka 1999 Protective Effects of Idebenone and a tocopherol on b amyloid 1 42 induced Learning and Memory Deficits in Rats Implication of Oxidative Stress in b amyloid induced Neurotoxicity In vivo European Journal of Neuroscience 11 1 83 90 doi 10 1046 j 1460 9568 1999 00408 x PMID 9987013 S2CID 84408095 Yan Du Shi Chen Xi Fu Jin Chen Ming Zhu Huaijie Roher Alex Slattery Timothy Zhao Lei Nagashima Mariko Morser John Migheli Antonio Nawroth Peter Stern David Marie Schmidt Ann 1996 RAGE and Amyloid b Peptide Neurotoxicity in Alzheimer s Disease Nature 382 6593 685 91 Bibcode 1996Natur 382 685Y doi 10 1038 382685a0 PMID 8751438 S2CID 4372023 Yang X 2007 Occurrence of the cyanobacterial neurotoxin anatoxin a in New York State waters ProQuest ISBN 978 0 549 35451 2 Zhang J Dawson V Dawson T Snyder S 1994 Nitric Oxide Activation of Poly ADP ribose Synthetase in Neurotoxicity Science 263 5147 687 89 Bibcode 1994Sci 263 687Z doi 10 1126 science 8080500 PMID 8080500 Further reading EditBrain Facts Book at The Society for Neuroscience Neuroscience Texts at University of Texas Medical School In Vitro Neurotoxicology An Introduction at Springerlink Biology of the NMDA Receptor at NCBI Advances in the Neuroscience of Addiction 2nd edition at NCBIExternal links Edit Look up neurotoxin in Wiktionary the free dictionary Wikimedia Commons has media related to Neurotoxins Environmental Protection Agency at United States Environmental Protection Agency Alcohol and Alcoholism at Oxford Medical Journals Neurotoxicology at Elsevier Journals Neurotoxin Institute at Neurotoxin Institute 1 Neurotoxins at Toxipedia Retrieved from https en wikipedia org w index php title Neurotoxin amp oldid 1141654405, 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.