You are on page 1of 7

Toxicology 410 (2018) 125–131

Contents lists available at ScienceDirect

Toxicology
journal homepage: www.elsevier.com/locate/toxicol

Neurotoxic effects of organophosphorus pesticides and possible association T


with neurodegenerative diseases in man: A review
Milan Jokanović
Experta Consulting, Belgrade, Serbia

A R T I C LE I N FO A B S T R A C T

Keywords: In this article the neurotoxic disorders appearing in patients exposed to organophosphorus pesticides and known
Organophosphorus compounds mechanisms involved are reviewed. Organophosphorus compounds cause four main neurotoxic effects in hu-
Neurotoxicity mans: the cholinergic syndrome, the intermediate syndrome, organophosphate-induced delayed polyneuropathy
Acetylcholinesterase and chronic organophosphate-induced neuropsychiatric disorder. Compared to the cholinergic syndrome, that
Neurodegenerative diseases
causes millions of cases of poisoning with fatality of more than 15% each year, other disorders involve much
Toxicity
smaller number of patients. Possible link of exposure to organophosphorus pesticides with neurodegenerative
diseases, dementia, attention deficit hyperactivity disorder and Parkinson's disease in man is also approached.
This article is focused on neurotoxic disorders appearing after acute and chronic exposure to organophosphates
with emphasis on molecular mechanisms, clinical presentation, pathogenesis, and possibilities for prevention/
medical treatment.

1. Introduction 2. The cholinergic syndrome

Organophosphorus compounds (OP) have been used as pesticides Signs and symptoms of cholinergic syndrome occurring in acute
and developed as warfare nerve agents such as soman, sarin, tabun, VX poisoning with OP pesticides are predictable from their biochemical
and others. Pesticide poisoning results from occupational, accidental, mechanism of action and are directly related to the levels of acet-
and intentional exposure (Clark, 2002). The epidemiological pattern of ylcholinesterase (AChE) activity. In cases of human poisoning, general
poisoning shows significant variation in number of deaths and form of acute symptoms of peripheral nicotinic and muscarinic intoxication are
poisoning between developing and industrial countries (Jeyaratnam, clearly apparent (World Health Organization, 1986). These symptoms
1990; Van der Hoek et al., 1998; Eddleston, 2000; Eddleston and include miosis; sweating, rhinorrhea, lacrimation, and salivation; ab-
Phillips, 2004). According to the World Health Organization, about 1 dominal cramps and other gastrointestinal symptoms; respiratory dif-
million accidental and 2 million suicidal poisonings with organopho- ficulties and cough; dyspnea, constriction sensation in the chest,
sphorus insecticides are reported per year, with more than 300,000 wheezing; twitching of facial muscles and tongue, tremors, and fasci-
fatalities (Jeyaratnam, 1990). Medical management is difficult, with culations; bradycardia and electrocardiogram (ECG) changes, pallor,
case fatality generally more than 15% (Eddleston et al., 2008; and cyanosis; anorexia, nausea, vomiting, diarrhea, and involuntary
Jokanović et al., 2010). urination and defecation. These signs and symptoms are accompanied
OP cause four main neurotoxic disorders in humans: the cholinergic by central effects such as dizziness, tremulousness, and confusion;
syndrome, the intermediate syndrome, organophosphate-induced de- ataxia; headache, fatigue, and paresthesia. Finally, seizures, convul-
layed polyneuropathy (OPIDP) and chronic organophosphate-induced sions, twitching, coma, and respiratory failure may occur. If the poi-
neuropsychiatric disorder (COPIND). Most of the cases of poisoning can soned patient survives the first day of poisoning, there are personality
be prevented by better administrative control, restricted access to OP changes, mood swings, aggressive events and psychotic episodes in-
pesticides, effective measures of personal protection and education of cluding schizoid reactions, paranoid delusions, and exacerbations of
OP pesticide applicators and medical personnel. preexisting psychiatric problems. Sleep is poor due to nightmares and
hallucinations; disturbances or deficits in memory and attention, and
additional delayed effects also occur. Death usually occurs due to re-
spiratory failure resulting from a combination of central and peripheral

E-mail address: milan.jokanovic@gmail.com.

https://doi.org/10.1016/j.tox.2018.09.009
Received 23 March 2018; Received in revised form 4 September 2018; Accepted 6 September 2018
Available online 25 September 2018
0300-483X/ © 2018 Elsevier B.V. All rights reserved.

Descargado para LUIS HERRERA GUTIERREZ (l.herrera1406@gmail.com) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en mayo 17, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Jokanović Toxicology 410 (2018) 125–131

effects, paralysis of the respiratory muscles, and depression of the brain no longer apparent (Senanayake and Karalliedde, 1987) and before the
respiratory center (Karchmar, 2007; World Health Organization, 1986; onset of organophosphate-induced delayed polyneuropathy (OPIDP,
IPCS, 1998; Clark, 2002; Eyer, 2003; Marrs and Vale, 2006; Jokanović discussed in the following section). Following exposure to various OP
et al., 2011). pesticides, clinical manifestations of IMS typically occur within 24 to
The first four to six hours are the most critical in acute poisoning 96 h, and affect patients without fasciculation or other cholinergic
with OP pesticides. If there is improvement in symptoms after initial signs. The incidence of IMS is between 7.7% and 84% cases with OP
treatment then the patient is very likely to survive if adequate treat- poisoning (John et al., 2003). Jayawardane et al. (2009) defined IMS as
ment is continued (IPCS, 1998). a spectrum disorder affecting the neuromuscular junction with two
The mechanism of cholinergic syndrome involves inhibition of main forms: a forme fruste variety associated with mild weakness and
AChE at synapses and neuromuscular junctions in cholinergic pathways the classical IMS with weakness of 3/5 or less than 3/5 on the Medical
leading to accumulation of acetylcholine and overstimulation of post- Research Council grading where the patients from the latter category
synaptic muscarinic and nicotinic receptors. Inhibition of AChE occurs are at risk of developing late onset respiratory failure. Although IMS is
after phosphorylation of hydroxyl group at serine at the active site of well recognized as a disorder of neuromuscular junctions, its exact
the enzyme. Following inhibition, AChE can be spontaneously re- etiology, incidence, and risk factors are not clearly understood. IMS
activated at the rate that depends on chemical structure of OP. For OP generally occurred among patients with prolonged and severe inhibi-
having dimethyl radicals the AChE reactivation is relatively rapid with tion of AChE and consequent persistent excess of ACh in the neuro-
a half-time of about 1–2 h, while that for OP having diethyl functional muscular junctions, but not in every patient (Yang and Deng, 2007).
groups is 31–57 h. For OP belonging to the group of warfare nerve Other risk factors of IMS involve delayed metabolism of OP pesticides,
agents (soman, sarin, tabun, VX) having branched radicals spontaneous impaired organ function, severity of poisoning, elevated muscle en-
AChE reactivation does not occur at all. For certain OP, an additional zymes, and inadequate or delayed therapy with atropine and pyr-
step can occur on phosphorylated AChE known as aging reaction that idinium oximes. IMS has been linked with exposure to specific OP
represents non-enzymatic time-dependent loss of one alkyl group (R) pesticides having dimethyl phosphate structure (e.g. fenthion, di-
bound to the phosphorus. The aging reaction depends on the chemical methoate, monocrotophos, dichlorvos, methylparathion), but also de-
structure of the inhibitor and leads to a stable non-reactivatable form of veloped after exposure to parathion (ethyl phosphate) and methami-
phosphorylated AChE (Reiner and Pleština, 1979; Worek et al., 1999; dophos (phosphoramidate) (De Bleecker et al., 1993; Yang and Deng,
Jokanović and Stojiljković, 2006; Jokanović and Prostran, 2009; 2007; Balali-Mood and Balali-Mood, 2008; Bird et al., 2016). Two ty-
Jokanović, 2012a). pical cases of IMS caused by fenthion and diazinon were discussed by
The duration of effects is determined mainly by the properties of the Jokanović et al. (2010). So far, the IMS has not been reported after
compound: its liposolubility, the stability of the OP-AChE complex and poisoning with warfare nerve agents (Balali-Mood and Saber, 2012).
whether it is reactivatable after the use of cholinesterase reactivators A case of carbofuran-induced IMS, possibly indicating the same
(such as oximes). It is important to note that only OP containing P]O mechanisms of OP- and carbamate-induced IMS, was reported by Paul
bond (known as direct inhibitors) are potent AChE inhibitors; those and Mannathukkaran (2005).
having a P]S group (indirect inhibitors) must be metabolically acti- Marked weakness of neck flexion and varying degree of proximal
vated to P]O group (Jokanović, 2001). The signs and symptoms of limb muscle weakness, manifesting as weakness of shoulder abduction
poisoning with direct inhibitors appear quickly during or after ex- and hip flexion, are the regular clinical features. Respiratory in-
posure, while those with indirect inhibitors appear slowly and last sufficiency is also common and frequently draws medical attention to
longer, even up to several days after cessation of exposure. the onset of the syndrome. Other possible manifestations are involve-
Clinical diagnosis of acute poisoning with OP compounds is rela- ment of muscles innervated by motor cranial nerves and decreased deep
tively simple and is based on medical history, circumstances of ex- tendon reflexes. Studies conducted in nineties have shown that IMS
posure, clinical presentation, and laboratory tests. Confirmation of di- goes along with excretion of cholinesterase inhibitor metabolites in the
agnosis can be made by measurement of erythrocyte AChE or plasma urine and by severe depression in cholinesterase levels. It was suggested
cholinesterase (ChE). Activities of these enzymes have been accepted as that the condition might reflect the recirculation of lipid soluble cho-
biomarkers of exposure and/or toxicity of OP. Erythrocyte AChE is linesterase inhibitors from body fat compartments or gastric fluids (De
identical to the enzyme present in the target synapses and its levels are Bleecker, 2006).
assumed to reflect the effects in target organs. For that reason, ery- John et al. (2003) demonstrated an association between the devel-
throcyte AChE is regarded as biomarker of toxicity of these compounds. opment of IMS and increased activity of enzymes creatine phosphoki-
On the other hand, ChE is also an esterase that can react with OP in the nase and lactate dehydrogenase, the markers of muscle function, in 25
same was as AChE. While the effects of inhibition of AChE can be fatal patients with acute organophosphate poisoning. Muscle injury was seen
(depending on the dose taken), inhibition of ChE does not have any in all patients beginning at admission, peaking over the first 5 days and
known consequences on the normal body functions (Jokanović and then declining over the next 5 days. Khan et al. (2001) and Dhandapani
Maksimović, 1997). ChE is a circulating plasma glycoprotein synthe- et al. (2003) demonstrated that oxidative cellular damage of muscle
sised in the liver, which does not serve any known physiological membranes could be associated with muscle necrosis. In a prospective
function. Since ChE (EC 3.1.1.8) is classified as different enzyme from study conducted by De Bleecker (1995) muscle biopsy performed in 19
AChE (EC 3.1.1.7), not being involved in cholinergic transmission in the patients with OP poisoning showed only minimal number of necrotic
nervous system, it does not provide accurate information regarding the fibers. IMS might involve the reduction in number of functioning cho-
severity of the OP poisoning (Jokanović, 2009, 2012b). Many OP in- linergic receptors at the postjunctional membrane, or a failure of
secticides (e.g. chlorpyrifos, demethon, malathion) appear to be more acetylcholine release. All these abnormal findings on electromyography
potent inhibitors of ChE than they are of erythrocyte AChE and, as the suggested a combined presynaptic and postsynaptic defect, without
consequence, ChE inhibition might occur to a greater extent than AChE sensory impairment (Baker and Sedgwick, 1996). Abdollahi and
inhibition (Jokanović, 2009, 2012b). Karami-Mohajeri (2012) suggested additional mechanisms of IMS in-
cluding prolonged AChE inhibition, muscle necrosis, down regulation
3. The intermediate syndrome or desensitization of postsynaptic ACh receptors and oxidative stress-
related myopathy.
Clinical manifestations of the intermediate syndrome (IMS) occur Management resembles that of rapidly developing respiratory dis-
within 24 to 96 h after exposure to OP when the symptoms of the tress and failure. Delays in instituting respiratory support may result in
cholinergic syndrome (e.g. muscle fasciculations, muscarinic signs) are death. The respiratory support is required during 7–21 days. In patients

126

Descargado para LUIS HERRERA GUTIERREZ (l.herrera1406@gmail.com) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en mayo 17, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Jokanović Toxicology 410 (2018) 125–131

with IMS continuous and careful monitoring of respiratory function with bilateral support and could perform housework; and 3 patients
(partial pressure of O2 and CO2 in arterial blood, arterial O2 saturation) could not self-care. Neurophysiological investigations showed normal
and blood pH are necessary (Karalliedde et al., 2006). The recovery electroencephalogram and visual, brainstem auditory and somatosen-
among patients who survived IMS starts with the improvement of sory evoked potentials. Motor evoked potential obtained from the upper
muscular power in cranial nerve-innervated muscles, followed by re- limbs had normal central motor conduction time, but it was delayed or
spiratory muscles, proximal muscles, and neck flexors. As IMS generally absent in bilateral lower limbs. Motor and sensory nerve conduction
takes place at the same time with severe OP toxicity and persistent velocity and electromyography studies were normal except for two
inhibition of AChE, early gastrointestinal decontamination, followed by severely affected patients. The prognosis for functional recovery de-
appropriate treatment with atropine and oximes, and prompt initiation pends on the degree of pyramidal involvement with ataxia and paralysis
of respiratory support, should be helpful in ameliorating the severity of representing a permanent outcome of severe OPIDP (Jokanović, 2017).
IMS. Yilmaz et al. (2013) reported clinical improvement in 13 out of 17 Several mechanisms of OPIDP have been proposed. OPIDP is in-
patients with IMS treated with therapeutic plasma exchange performed itiated by phosphorylation and subsequent aging of > 70% the enzyme
with fresh frozen plasma. The prognosis of IMS appears to be favorable originally known as neurotoxic esterase, but later renamed to neuro-
if respiratory failure can be promptly recognized and treated accord- pathy target esterase (NTE) in peripheral nerves (Johnson, 1982; Lotti,
ingly (De Bleecker et al., 1993; De Bleecker, 2006; Karalliedde et al., 1992). NTE was discovered by Martin Johnson, who described its most
2006; Yang and Deng, 2007). important toxicological and biochemical features (Johnson, 1982). NTE
is an integral membrane protein in vertebrate neurons present in all
4. Organophosphate induced delayed polyneuropathy neurons, but not in glia (Glynn, 1999; Li et al., 2003). It is codified by
gene PNPLA6 (Sogorb et al., 2016). The active site of NTE contains
OPIDP is a unique toxicological phenomenon caused by a single Ser966 and two aspartates Asp960 and Asp1086 that appear essential for
exposure to certain OP with effects usually appearing after 10 to 20 enzymatic activity. The physiological substrate for NTE is considered to
days or later. OPIDP is toxicologically different from the cholinergic be lysolecithin (Quistad et al., 2003). NTE regulates phospholipid me-
syndrome in that it is based on different mechanisms not involving tabolism and is known to be a phospholipase B (Read et al., 2009;
AChE and appearing a few weeks after cholinergic syndrome has been Chang and Wu, 2010. Inactivation of NTE may reduce the degradation
medically solved with standard therapeutic measures and patient of phosphatidylcholine to glycerophosphocholine. The deficiency of the
usually dismissed from hospital. OPIDP is also a different syndrome NTE activity may lead to abnormal accumulation of phosphatidylcho-
from IMS. line-containing membranes in cells, which may interfere with normal
There were two major outbreaks of OP poisoning in which OPIDP membrane lipid homeostasis and fluidity affecting the initiation of
appeared in thousands cases of poisoning with triorthocresyl phosphate neurites. Since NTE hydrolyzes lysolecithin (Vose et al., 2008; Greiner
(TOCP) that occurred mainly due to beverage and food contamination et al., 2010), the delay in initiation of neurites may also be caused by an
in USA in 1930 and Morocco in 1959 (Johnson, 1982; Morgan, 1982; abnormal accumulation of lysolecithin in NTE-deficient cells (Li et al.,
Lotti, 1992; Jokanović et al., 2004). Many cases of OPIDP due to TOCP 2005).
poisoning were reported in Romania, Sri Lanka, former Yugoslavia and Sogorb et al. (2016) suggested that gene PNPLA6 and/or the NTE
China. In addition to TOCP, several other OP pesticides have been re- protein might play a role in early neurodifferentiation stages. NTE is
ported to cause OPIDP in man (Jokanović et al., 2002; Lotti and involved in intracellular membrane trafficking and cell-signaling
Moretto, 2005; Jokanović and Kosanović, 2010; Jokanović et al., 2011; pathway between neurons and glial cells (Glynn, 2006). There is a re-
Satoh and Jokanović, 2014; Jokanović, 2017). Cases of OPIDP caused lationship between NTE activity and the axonal maintenance since it
by pesticides were discussed in more details by Lotti and Moretto facilitates the transport of macromolecules from the neuron to the distal
(2005). ends of long axons (Read et al., 2009).
OPIDP is a rare neurodegenerative disorder in humans character- It was also suggested that NTE may be involved in the regulation of
ized by loss of function and ataxia of distal parts of sensory and motor calcium entrance into cells being responsible for the maintenance of
axons in peripheral nerves and ascending and descending tracts of normal function of calcium channels, and that increasing calcium ac-
spinal cord. The early neurological symptoms usually are sharp, cramp- tivated neutral protease activity is responsible for triggering OPIDP
like pains in the calves, tingling in the feet and hands followed by distal (Emerick et al., 2010). Song and Xie (2012) proposed that the activa-
numbness and parenthesis. Pain and weakness in muscles spread and tion of calpains (calcium-dependent cysteine proteases) plays a critical
patients rapidly become unable to keep their balance. Progressive leg role in OPIDN. The molecular mechanisms involved is still unclear.
weakness occurs with depression of tendon reflexes. Symptoms also Medical treatment of OPIDP in humans is symptomatic. Standard
appear in the arms and forearms. Sensory loss may be mild. Muscle treatment of OP poisoned patients comprising atropine, oxime and
tonus of the limbs gradually increase and spasticity appears in the lower diazepam was not effective in treatment of OPIDP. However, there were
limbs. Physical examination reveals distal symmetrical mostly motor several reports in the literature describing attempts of treatment of
polyneuropathy, with flaccid weakness of distal limb muscles in the OPIDP in animals and these studies were reviewed by Lotti (1992) and
lower limbs. In severe OPIDP quadriplegia with foot and wrist drop Jokanović et al. (2004), but none of these treatments have been tested
were observed as well as mild pyramidal signs (Lotti, 1992). Some in patients so far.
functional recovery may be seen in less severe cases with more distal
involvement and sparing of spinal cord axons, but pyramidal and other 5. Chronic organophosphate-induced neuropsychiatric disorder
signs of central neurological involvement appear with time. It appears
that clinical signs of OPIDP in children are considerably milder than in Chronic exposure to OP has been associated with impaired neuro-
adults. The recovery was observed only sensory nerves, while motor behavioral performance in some, but not all, epidemiological studies
neurons may permanently lose its function as confirmed by Morgan (Ray and Richards, 2001). Chronic organophosphate-induced neu-
(1982) who described the lack of improvement during 47 years in 11 ropsychiatric disorders (COPIND) occur without cholinergic symptoms
patients poisoned with TOCP. Similar findings were also reported by and apparently are not dependent on AChE inhibition (Ray and
Tosi et al. (1994) who found still persistent symptoms of OPIDP in 7 Richards, 2001; Singh and Sharma, 2000). COPIND usually appears
patients poisoned with TOCP 50 years ago. However, the study pre- with a delay and persists for a long period possibly suggesting the
sented by Wang et al. [12] conducted 13 years after TOCP poisoning of permanent damage of the central nervous system (Savage et al., 1988;
74 patients revealed that out of 61 survivors, 35 patients almost re- De Silva et al., 2006; Tan et al., 2009). The most common symptoms of
gained normal function of limbs and work outside; 23 patients walked COPIND include cognitive deficit (impairment in memory,

127

Descargado para LUIS HERRERA GUTIERREZ (l.herrera1406@gmail.com) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en mayo 17, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Jokanović Toxicology 410 (2018) 125–131

concentration and learning, problems with attention, information pro- of mortality and morbidity studies related to suicide among pesticide-
cessing, eye-hand coordination and reaction time), mood change (an- exposed populations, revealed high suicide rates in farming populations
xiety, depression, psychotic symptoms, emotional lability), chronic fa- (Parron et al., 1996).
tigue, autonomic dysfunction, peripheral neuropathy and The underlying mechanism of COPIND has not been established.
extrapyramidal symptoms such as dystonia, resting tremor, bradiky- Tan et al. (2009) hypothesized that COPIND could be derived from
nesia, postural instability and rigidity of face muscles (Jokanović et al., withdrawal of OP pesticide after chronic low-level exposure or acute
2011; Ray and Richards, 2001; Singh and Sharma, 2000; Ahmed and exposure. Other scientists have suggested that mechanisms other than
Davies, 1997; Davies et al., 2000a; Salvi et al., 2003; Kamel and inhibition of AChE might also be involved. These alternative mechan-
Hoppin, 2004; London et al., 2005; Roldan-Tapia et al., 2005; Ross isms may involve other protein targets (such as serine hydrolases, acyl
et al., 2013; Venkategowda et al., 2015; Jamal et al., 2016; Taghavian peptide hydrolase) present in the nervous system, interacting with some
et al., 2016). Suicidality and alcohol intolerance have also been re- OPs, that may be changed even after low level exposure leading to
ported (Davies et al., 2000a). cognitive damage (Ray, 1998; Ray and Richards, 2001; Pancetti et al.,
In children exposed to OPs impairment of memory, attention, verbal 2007). Several animal studies have shown that cognitive enhancing
learning, behavioral and motor coordination were observed (Ruckart action of low doses of certain OPs, such as dichlorvos, in rats were
et al., 2004). Some of the findings in these studies were apparently too unrelated to AChE inhibition. It was suggested that OPs may affect
subtle to be detected by a general neurological examination (Pope, neuropeptide metabolism through the release of endogenous opiates
2006). and/or through interactions with yet unidentified receptors (Kohen
Similar clinical features have also been reported by soldiers suf- et al., 1980; Kubek et al., 1997; Desi and Nagymajtenyi, 1999). It has
fering from the Gulf-War Syndrome, which led to the hypothesis that also been observed that administration of different OPs has different
the illness was caused by chronic exposure to chemical warfare agents behavioral presentation, suggesting that observed effects are not en-
with similar effects to OP pesticides (Gronseth, 2005). However, as tirely a result of AChE inhibition (Pope et al., 1992; van Dongen and
there was no correlation between the cause and the effect, it was con- Wolthuis, 1989v). London et al. (2005) reported that exposure to OPs
sidered unlikely that Gulf War veterans were suffering chronic effects may cause serotonin disturbances in the central nervous system, which
from illnesses caused by chemical warfare agent exposure (Riddle et al., are implicated in depression and suicidal behavior in humans. Rohlman
2003; Lotti, 2002). et al. (2011) suggested that oxidative stress and inflammation being
Kawana et al. (2001) investigated psychological and physiological mechanistically linked to chronic OP neurotoxicity correlate well with
symptoms in 582 sarin poisoned patients after the Tokyo subway neurobehavioral deficits observed consequent to neurodegenerative
system terrorist attack on March 20, 1995. The patients were given diseases (Rohlman et al., 2011).
emergency care at St. Luke’s International Hospital on the day of the
attack. It was demonstrated that victims of the Tokyo sarin attack 6. The link between exposure to OP pesticides and neurotoxic and
continued to suffer from physical and physiological symptoms 5 years neurodegenerative diseases in man
after. The most common physical symptoms reported were eye symp-
toms (tiredness of eyes, dim vision, difficulty with eye accomodation Most epidemiological studies of occupational exposure to pesticides
and distance vision, focus), tiredness, fatigue, muscle stiffness and are apparently of limited value due to the lack an adequate tox-
headache. Psychological symptoms did not decrease over the 5 years of icological evaluation. Most of them did not have a correlation between
the study except for depressed mood. Ten years after the Matsumoto the specific cause and the effect. In many cases the pesticides were not
and Tokyo accidents long-lasting psychological problems (such as for- even identified and the information about the exposure is obtained only
getfulness, chronic lack of concentration, depressive mood) were still through certain undefined questionnaires and interviews whose re-
evident (Yanagisawa et al., 2006). levance is unclear. The most important problems with these studies are:
Diagnostic criteria for COPIND include (Davies et al., 2000b): 1) not making any difference between chemical structure and me-
chanisms of toxicity of the pesticides involved; 2) not considering the
1 Repeated exposure to OP; presence and effects of chronic exposure to organic solvents and/or
2 At least four of the following: a) personality change and destabili- diluents that are present in much higher quantities in technical pesti-
zation of mood, b) impairment of concentration, c) impaired ex- cide formulation than active substance; 3) the possible presence of
ercise tolerance, d) reduced tolerance to alcohol, e) increased sen- toxicologically relevant impurities and/or isomers was not considered;
sitivity to OP; 4) exposure to pesticides was not clearly described in terms of dose
3 At least three of the following: a) exacerbation of “dippers flu”, b) level and duration of exposure, detection of pesticides and/or their
impulsive suicidal thinking, c) language disorder, d) improved sense metabolites in blood/urine or human tissues, whether safety equipment
of smell, e) deterioration of handwriting. was applied; 5) there was no indication whether different types of
pesticides were used during the spraying season in the same area and
In several epidemiological studies conducted among farm workers the possibility of exposure to different pesticides at the same time; 6) in
and pesticide applicators, neuropsychological damage accompanied case of OP insecticides the levels of AChE and ChE activity as well as
with damage of peripheral nervous system, anxiety and depression interaction with the enzymes involved in metabolism of OP compounds
were predominant among the exposed group (Steenland et al., 1994; (carboxylesterases, paraoxonases, CYP450's) were not considered.
London et al., 1998; Bessler and Stallones, 2008; Bayrami et al., 2012). Given the above limitations, in this study only the results of the
Agricultural workers tested about two years after a pesticide exposure studies where exposure to OP pesticides was confirmed will be sum-
episode showed significantly lower performance in verbal and visual marized.
attention, visual memory, sequencing and problem solving (Rosenstock
et al., 1991). Savage et al. (1988) showed abnormalities in psycho- 6.1. OP and Alzheimer’s dementia (AD)
metric testing and motor reflexes. Farm workers with symptoms of mild
OP pesticides intoxication requiring no hospitalization performed The analysis of exposures associated with incidence of AD revealed
worse on tests of cognitive and psychomotor function than non-exposed that in workers occupationally exposed to OP the risk is increased by
workers tested 2 years later. During the process of dipping sheep into 1.5 times (Hayden et al., 2010).
the basin with insecticide, workers exposed to OPs developed greater Rothlein et al. (2006) compared the neurobehavioral performance
vulnerability to psychiatric disorders such as significant anxiety and of 92 Hispanic immigrant farmworkers with 45 non-agricultural con-
depression (Jamal, 1997; Kamel and Hoppin, 2004). A literature review trols matched by age and educational level. They found that

128

Descargado para LUIS HERRERA GUTIERREZ (l.herrera1406@gmail.com) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en mayo 17, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Jokanović Toxicology 410 (2018) 125–131

farmworkers performed worse than controls but more importantly there metabolite concentrations (indicators of exposure to dimethyl OPs) and
was a positive correlation between urinary OP metabolite levels and increased odds of ADHD in children 8–15 years. There was a 55–72%
poorer performance on some neurobehavioral tests, thus confirming increase in the odds of ADHD for a 10-fold increase in 3-dimethyl al-
that low level long term occupational exposure to organophosphates kylphosphate concentration. The authors concluded that the results
could adversely affect cognitive performance. support the hypothesis that current levels of OP pesticides exposure
It has been observed that individuals exposed to OP display memory may contribute to ADHD prevalence (London et al., 2012).
and learning impairment, especially after acute poisoning. Patients
seriously poisoned with OP can be subject to nonspecific brain injury 6.3. OP and Parkinson's disease (PD)
(such as anoxia) possibly indicating direct neurotoxic effects of OP.
However, current biomarkers of organophosphate exposure (such as Environmental exposure to chlorpyrifos has been compared be-
blood cholinesterase activity and urinary levels of OP metabolites) do tween cases of PD and the matched controls by an analysis of ground-
not correlate consistently with neurobehavioral performance (Rohlman water. It was found that elevated risk of PD after using well water
et al., 2011). contaminated with chlorpyrifos was 1.87-fold (Gatto et al., 2009). In
Another association of OP to AD comes from a number of studies another similar study it was reported that residential exposure to
reporting that certain polymorphisms on the PON1 gene for chlorpyrifos and diazinon increased the risk of PD by more than two
Paraoxonase 1, in accordance with similar results obtained in PD stu- times (Manthripragada et al., 2010). Increased risk of PD after exposure
dies (Manthripragada et al., 2010), correlated to increased risk for AD to OP insectides (mainly chlorpyrifos) was shown as odd ratio of 1.7
(Zaganas et al., 2013). (Mostafalou and Abdollahi, 2017; Narayan et al., 2013).
A more than 2-fold increase in the risk of PD incidence was asso-
6.2. OP and attention deficit hyperactivity disorder (ADHD) ciated with chlorpyrifos use in Texan agricultural workers (Dhillon
et al., 2008), and with potential exposure to high chlorpyrifos levels
Elevated odds ratio of 1.5 and 5.1 for ADHD have been linked with through well-water consumption in residents of rural California (Gatto
OPs by two studies examining the exposure via urine and blood samples et al., 2009). When PON1 was taken into consideration it was found
(Bouchard et al., 2010; Suarez-Lopez et al., 2013). Comparing the cases that the association between potential exposure to chlorpyrifos and PD
of ADHD with control regarding exposure to OPs indicated that the risk was strengthened (by around 3-fold) in carriers of the PON1 55 MM
biomarkers of OPs were 2–3 times more detectable in the urine sample variant, which encodes for possible functional variants of xenobiotic
of the cases than that of controls (Yu et al., 2016). metabolizing enzymes (Manthripragada et al., 2010; Freire and
A cohort study measuring the OP biomarkers in the urine samples of Koifman, 2012).
the mothers revealed an odds ratio of 1.3 for the risk of ADHD in as- In conclusion, in this article the neurotoxic disorders appearing in
sociation with maternal exposure to OPs (Marks et al., 2010). Another patients poisoned with OP pesticides and known mechanisms involved
study measuring the urine concentration of OPs indicated that maternal are reviewed. OP cause four main neurotoxic effects in humans: the
exposure to chlorpyrifos increased the risk of ADHD in boys and at- cholinergic syndrome, the intermediate syndrome, organophosphate-
tention deficit in girls with respective odds ratio 5.5 and 5.8 induced delayed polyneuropathy and chronic organophosphate-in-
(Fortenberry et al., 2014). The link between prenatal exposure to duced neuropsychiatric disorder. Compared to the cholinergic syn-
chlorpyrifos and higher incidence of ADHD with a risk estimate of 6.5 drome, that causes millions of cases of poisoning with fatality of more
was resulted from a cohort study examining the exposure via blood than 15% each year, other disorders involve much smaller number of
level measurement of the pesticides (Rauh et al., 2011; Mostafalou and patients. Possible link of exposure to organophosphorus pesticides with
Abdollahi, 2017). neurodegenerative diseases, dementia, attention deficit hyperactivity
Epidemiological studies connecting exposure to OP pesticides and disorder and Parkinson's disease in man is also approached. This article
neurodevelopment have focused on populations with high exposure is focused of neurotoxic disorders appearing after acute and chronic
relative to the general population. Prenatal exposure to OP pesticides exposure to OP with emphasis on molecular mechanisms, clinical pre-
was associated with increased risk of developmental disorders, delays in sentation, pathogenesis, and possibilities for prevention/medical
cognitive development, and attentional deficits (Eskenazi et al., 2007; treatment.
Bouchard et al., 2011; Marks et al., 2010; Engel et al., 2007; Rauh et al.,
2011). Postnatal OP exposure has been associated with behavioral Conflict of interest
problems, poorer short-term memory and motor skills, and longer re-
action time in children (London et al., 2012). None.
The later study examined the cross-sectional association between
urinary dialkyl phosphate metabolite concentrations and ADHD pre- Acknowledgement
valence in children of ages 8–15 years. The urinary dialkyl phosphate
metabolites measured in this study were 3-dimethyl alkylphosphate This study was supported in part by the Ministry of Education,
molecules and 3-diethyl alkylphosphate molecules. The study was Science and Technological Development of the Republic of Serbia
conducted in 1139 children aged 8–15 years. The metabolite di- (Project 175045).
methylthiophosphate was the most commonly detected 3-dimethyl al-
kylphosphate (in 64.3% of children). Children with creatinine adjusted References
dimethylthiophosphate concentrations above the median of detectable
values had double the odds of ADHD compared to those with con- Abdollahi, M., Karami-Mohajeri, S., 2012. A comprehensive review on experimental and
centrations below the detection (London et al., 2012). clinical findings in intermediate syndrome caused by organophosphate poisoning.
Toxicol. Appl. Pharmacol. 258, 309–314.
The odds of meeting the diagnostic criteria for hyperactive im- Ahmed, G.M., Davies, D.R., 1997. Chronic organophosphate exposure: toward the defi-
pulsive subtype increased significantly with higher 3-diethyl alkyl- nition of a neuropsychiatric syndrome. J. Nutr. Environ. Med. 7, 169–176.
phosphate, 3-dimethyl alkylphosphate, and total dialkyl phosphate Baker, D.J., Sedgwick, F.M., 1996. Single fiber electromyographic changes in man after
organophosphate exposure. Hum. Exp. Toxicol. 15, 369–375.
(adjusted odds ratio for a 10-fold increase in concentration 2.2 and 1.9 Balali-Mood, M., Balali-Mood, K., 2008. Neurotoxic disorders of organophosphorous
respectively). The odds of inattentive subtype increased with higher compounds and their managements. Arch. Ir. Med. 11, 65–89.
concentrations of 3-dimethyl alkylphosphate metabolites although this Balali-Mood, M., Saber, H., 2012. Recent advances in the treatment of organopho-
sphorous poisonings. Iran. J. Med. Sci. 37, 74–91.
did not reach significance level (adjusted odds ratio 1.5). The study Bayrami, M., Hashemi, T., Malekirad, A.A., Ashayeri, H., Faraji, F., Abdollahi, M., 2012.
indicated an association between urinary 3-dimethyl alkylphosphate

129

Descargado para LUIS HERRERA GUTIERREZ (l.herrera1406@gmail.com) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en mayo 17, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Jokanović Toxicology 410 (2018) 125–131

Electroencephalogram, cognitive state, psychological disorders, clinical symptom, Updated 1998. Available online at:. http://www.inchem.org/documents/pims/
and oxidative stress in horticulture farmers exposed to organophosphate pesticides. chemical/pimg001.htm.
Toxicol. Ind. Health 28, 90–96. Jamal, G.A., 1997. Neurological syndromes of organophosphorus compounds. Adverse
Bessler, C.L., Stallones, L., 2008. A cohort study of pesticide poisoning and depression in drug reaction. Toxicol. Rev. 16, 133–170.
Colorado farm residents. Ann. Epidemiol. 18, 768–774. Jamal, F., Quazi, S., Haque, Q.S., Singh, S., 2016. Interrelation of glycemic status and
Bird, S.B., Krajačić, P., Sawamoto, K., Bunya, N., Loro, E., Khurana, T.S., 2016. neuropsychiatric disturbances in farmers with organophosphorus pesticide toxicity.
Pharmacotherapy to protect the neuromuscular junction after acute organopho- Open Biochem. J. 10, 27–34.
sphorus pesticide poisoning. Ann. N.Y. Acad. Sci. 1374 (1), 86–93. Jayawardane, P., Senanayake, N., Dawson, A., 2009. Electrophysiological correlates of
Bouchard, M.F., Bellinger, D.C., Wright, R.O., Weisskopf, M.G., 2010. Attention-deficit/ intermediate syndrome following acute organophosphate poisoning. Clin. Toxicol.
hyperactivity disorder and urinary metabolites of organophosphate pesticides. 47, 193–205.
Pediatrics 125, e1270–e1277. Jeyaratnam, J., 1990. Pesticide poisoning: a major global health problem. World Health
Bouchard, M.F., Chevrier, J., Harley, K.G., Kogut, K., Vedar, M., Calderon, N., Trujillo, C., Stat. Q. 43, 139–144.
Johnson, C., Bradman, A., Barr, D.B., Eskenazi, B., 2011. Prenatal exposure to or- John, M., Oommen, A., Zachariah, A., 2003. Muscle injury in organophosphorous poi-
ganophosphate pesticides and IQ in 7-year-old children. Environ. Health Perspect. soning and its role in the development of intermediate syndrome. Neuro Toxicol. 24,
119, 1189–1195. 43–53.
Chang, P.A., Wu, Y.J., 2010. Neuropathy target esterase: an essential enzyme for neural Johnson, M.K., 1982. The target for initiation of delayed neurotoxicity by organopho-
development and axonal maintenance. Int. J. Biochem. Cell Biol. 42, 573–575. sphorus esters: biochemical studies and toxicological applications. In: In: Hodgson,
Clark, R.F., 2002. Insecticides: organic phosphorus compounds and carbamates. E., Bend, J.R., Philpot, R.M. (Eds.), Revews in Biochemical Toxicology, vol. 4.
Goldfrank’s Toxicological Emergencies, 7 ed. McGraw-Hill Professional, New York, Elsevier, New York, pp. 141–212.
pp. 1346–1360. Jokanović, M., 2001. Biotransformation of organophosphorus compounds. Toxicology
Davies, R., Ghouse, A., Tegwedd, F., 2000a. Chronic exposure to organophosphates: 166, 139–160.
background and clinical picture. Adv. Psychiatr. Treat. 6, 187–192. Jokanović, M., 2009. Medical treatment of acute poisoning with organophosphorus and
Davies, R., Ghouse, A., Tegwedd, F., 2000b. Psychiatric aspects of chronic exposure to carbamate pesticides. Toxicol. Lett. 190, 107–115.
organophosphates: diagnosis and management. Adv. Psychiatr. Treat. 6, 356–361. Jokanović, M., 2012a. Neurotoxic disorders and medical management of patients poi-
De Bleecker, J., 1995. The intermediate syndrome in organophosphate poisoning: an soned with organophosphorus and carbamate pesticides. In: Jokanović, M. (Ed.), The
overview of experimental and clinical observations. J. Toxicol. Clin. Toxicol. 33, Impact of Pesticides. Academic Publishers, pp. 39–62.
683–686. Jokanović, M., 2012b. Structure-activity relationship and efficacy of pyridinium oximes
De Bleecker, J.L., 2006. Intermediate syndrome in organophosphate poisoning. In: Gupta, in the treatment of poisoning with organophosphorus compounds: a review of recent
R.C. (Ed.), Toxicology of Organophosphate and Carbamate Compounds. Elsevier data. Curr. Top. Med. Chem. 12, 1775–1789.
Academic Press, Amsterdam, pp. 371–380 Chapter 26. Jokanović, M., 2017. Ataxia in patients poisoned with organophosphorus compounds. J.
De Bleecker, J., Van Den Neucker, K., Colardyn, F., 1993. Intermediate syndrome in or- Neurol. Transl. Neurosci. 5, 1081–1083.
ganophosphorus poisoning: a prospective study. Crit. Care Med. 21, 1706–1711. Jokanović, M., Kosanović, M., 2010. Neurotoxic effects in patients poisoned with orga-
De Silva, H.J., Samarawickrema, N.A., Wickremasinghe, A.R., 2006. Toxicity due to or- nophosphorus pesticides. Environ. Toxicol. Pharmacol. 29, 195–201.
ganophosphorus compounds: what about chronic exposure? Trans. Roy. Soc. Trop. Jokanović, M., Maksimović, M., 1997. Abnormal cholinesterase activity: understanding
Med. Hyg. 100, 803–806. and interpretation. Eur. J. Clin. Chem. Clin. Biochem. 35, 11–16.
Desi, I., Nagymajtenyi, L., 1999. Electrophysiological biomarkers of an organopho- Jokanović, M., Prostran, M., 2009. Pyridinium oximes as cholinesterase reactivators.
sphorous pesticide, dichlorvos. Toxicol. Lett. 107, 55–64. Structure-activity relationship and efficacy in the treatment of poisoning with orga-
Dhandapani, M., Zachariah, A., Kavitha, M.R., Jeyaseelan, L., Oommen, A., 2003. nophosphorus compounds. Curr. Med. Chem. 16, 2177–2188.
Oxidative damage in intermediate syndrome of acute organophosphorus poisoning. Jokanović, M., Stojiljković, M.P., 2006. Current understanding of the application of
Ind. J. Med. Res. 117, 253–259. pyridinium oximes as cholinesterase reactivators in treatment of organophosphate
Dhillon, A.S., Tarbutton, G.L., Levin, J.L., Plotkin, G.M., Lowry, L.K., Nalbone, J.T., poisoning. Eur. J. Pharmacol. 553, 10–17.
Shepherd, S., 2008. Pesticide/environmental exposures and parkinson’s disease in Jokanović, M., Stukalov, P.V., Kosanović, M., 2002. Organophosphate induced delayed
east texas. J. Agromed. 13, 37–48. polyneuropathy. Curr. Drug Targets Cns Neurol. Disord. 1, 591–600.
Eddleston, M., 2000. Patterns and problems of deliberate self-poisoning in the developing Jokanović, M., Kosanović, M., Stukalov, P.V., 2004. Organophosphate induced delayed
world. QJM 93, 715–731. polyneuropathy. Med. Chem. Rev. Online 1, 123–131.
Eddleston, M., Phillips, M.R., 2004. Self-poisoning with pesticides. BMJ 328, 42–44. Jokanović, M., Antonijević, B., Vučinić, S., 2010. Epidemiological studies of antic-
Eddleston, M., Juszczak, E., Buckley, N.A., Senarathna, L., Mohamed, F., Dissanayake, W., holinesterase pesticide poisoning in Serbia. In: Satoh, T., Gupta, R.C. (Eds.),
Hittarage, A., Azher, S., Jeganathan, K., Jayamanne, S., Sheriff, M.H.R., Warrell, Anticholinesterase Pesticides: Metabolism, Neurotoxicity and Epidemiology. John
D.A., 2008. Multiple-dose activated charcoal in acute self-poisoning: a randomised Wiley & Sons, pp. 481–494 Chapter 35.
controlled trial. Lancet 371, 579–587. Jokanović, M., Kosanović, M., Brkić, D., Vukomanović, P., 2011. Organophosphate in-
Emerick, G.L., Peccinini, R.G., de Oliveira, G.H., 2010. Organophosphorus -induced de- duced delayed polyneuropathy in man: an overview. Clin. Neurol. Neurosurg. 113,
layed neuropathy: a simple and efficient therapeutic strategy. Toxicol. Lett. 192, 7–10.
238–244. Kamel, F., Hoppin, J.A., 2004. Association of pesticide exposure with neurologic dys-
Engel, S.M., Berkowitz, G.S., Barr, D.B., Teitelbaum, S.L., Siskind, J., Meisel, S.J., function and disease. Environ. Health Perspect. 112, 950–958.
Wetmur, J.G., Wolff, M.S., 2007. Prenatal organophosphate metabolite and organo- Karalliedde, L., Baker, D., Marrs, T., 2006. Organophosphate-induced intermediate syn-
chlorine levels and performance on the Brazelton Neonatal Behavioral Assessment drome: aetiology and relationships with myopathy. Toxicol. Rev. 25, 1–14.
Scale in a multiethnic pregnancy cohort. Am. J. Epidemiol. 165, 1397–1404. Karchmar, A.G., 2007. Anticholinesterases and war gases. In: Karchmar, A.G. (Ed.),
Eskenazi, B., Marks, A.R., Bradman, A., Harley, K., Barr, D.B., Johnson, C., Morga, N., Exploring the Vertebrate Central Cholinergic Nervous System. Springer, pp. 237–310
Jewell, N.P., 2007. Organophosphate pesticide exposure and neurodevelopment Chapter 7.
health in young Mexican-American children. Environ. Health Perspect. 115, Kawana, N., Ishimatsu, S., Kanda, K., 2001. Psycho-physiological effects of the terrorist
792–798. sarin attack on the Tokyo subway system. Mil. Med. 166 (Suppl. 2), 23.
Eyer, P., 2003. The role of oximes in the management of organophosphorus pesticide Khan, S., Hemalatha, R., Jeyaseelan, L., Oommen, A., Zachariah, A., 2001. Neuroparalysis
poisoning. Toxicol. Rev. 22, 165–190. and oxime efficacy in organophosphate poisoning; a study of butyrylcholinesterase.
Fortenberry, G.Z., Meeker, J.D., Sanchez, B.N., Barr, D.B., Panuwet, P., Bellinger, D., Hum. Exp. Toxicol. 20, 169–174.
Schnaas, L., Solano-Gonzalez, M., Ettinger, A.S., Hernandez-Avila, M., Hu, H., Tellez- Kohen, G.L., Henderson, G., Karczmar, A.G., 1980. Di-isopropyl phosphofluoridate in-
Rojo, M.M., 2014. Urinary 3,5,6-trichloro-2-pyridinol (TCPY) in pregnant women duced antinociception: possible role of endogenous opioids. Eur. J. Pharmacol. 61,
from Mexico city: distribution, temporal variability, and relationship with child at- 167–173.
tention and hyperactivity. Int. J. Hyg. Environ. Health 217, 405–412. Kubek, M.J., Shih, T.M., Meyerhoff, J.L., 1997. Thyrotropin-releasing hormone (TRH) is
Freire, C., Koifman, S., 2012. Pesticide exposure and Parkinson’s disease: epidemiological markedly increased in the rat brain following soman-induced convulsions. Brain Res.
evidence of association. NeuroToxicol. 33, 947–971. 747, 328–331.
Gatto, N.M., Cockburn, M., Bronstein, J., Manthripragada, A.D., Ritz, B., 2009. Well- Li, Y., Dinsdale, D., Glynn, P., 2003. Protein domains, catalytic activity and subcellular
water consumption and Parkinson’s disease in rural California. Environ. Health distribution of neuropathy target esterase in mammalian cells. J. Biol. Chem. 278,
Perspect. 117, 1912–1918. 8820–8825.
Glynn, P., 1999. Neuropathy target esterase. Biochem. J. 344, 625–631. Li, Z., Szurek, P.F., Jiang, C., Pao, A., Bundy, B., Le, W.D., Bradley, A., Yu, Y.E., 2005.
Glynn, P., 2006. A mechanism for organophosphate-induced delayed neuropathy. Neuronal differentiation of NTE-deficient embryonic stem cells. Biochem. Biophys.
Toxicol. Lett. 162, 94–97. Res. Commun. 330, 1103–1109.
Greiner, A.J., Richardson, R.J., Worden, R.M., Ofoli, R.Y., 2010. Influence of lysopho- London, L., Nell, V., Thompson, M.L., Myers, J.E., 1998. Effects of long term organo-
spholipid hydrolysis by the catalytic domain of neuropathy target esterase on the phosphate exposures on neurological symptoms, vibration sense and tremor among
fluidity of bilayer lipid membranes. Biochim. Biophys. Acta 1798, 1533–1539. South African farm workers. Scand. J. Work Environ. Health 24, 18–29.
Gronseth, G.S., 2005. Gulf war syndrome: a toxic exposure? A systematic review. Neurol. London, L., Flisher, A.J., Wesseling, C., Mergler, D., Kromhout, H., 2005. Suicide and
Clin. 23, 523–540. exposure to organophosphate pesticides: cause or effect? Am. J. Ind. Med. 47,
Hayden, K.M., Norton, M.C., Darcey, D., Ostbye, T., Zandi, P.P., Breitner, J.C., Welsh- 308–321.
Bohmer, K.A., 2010. Occupational exposure to pesticides increases the risk of in- London, L., Beseler, C., Bouchard, M.F., Bellinger, D.C., Colosio, C., Grandjean, P., Harari,
cident AD: the Cache County study. Neurol 74, 1524–1530. R., Kootbodien, T., Kromhout, H., Little, F., Meijster, T., Moretto, A., Rohlman, D.S.,
IPCS, 1998. Organophosphorus Pesticides. International Programme on Chemical Safety Stallones, L., 2012. Neurobehavioral and neurodevelopmental effects of pesticide
Poisons Information Monograph G001, World Health Organization, Geneva, 1989; exposures. Neuro Toxicol. 33, 887–896.

130

Descargado para LUIS HERRERA GUTIERREZ (l.herrera1406@gmail.com) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en mayo 17, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Jokanović Toxicology 410 (2018) 125–131

Lotti, M., 1992. The pathogenesis of organophosphate delayed polyneuropathy. Crit. Rev. following low-level exposure to organophosphate pesticides: a systematic and meta-
Toxicol. 21, 465–487. analytic review. Crit. Rev. Toxicol. 43, 21–44.
Lotti, M., 2002. Low-level exposures to organophosphorus esters and peripheral nerve Rothlein, J., Rohlman, D., Lasarev, M., Phillips, J., Muniz, J., McCaule, L., 2006.
function. Muscle Nerve 25, 492–504. Organophosphate pesticide exposure and neurobehavioral performance in agri-
Lotti, M., Moretto, A., 2005. Organophosphate-induced delayed polyneuropathy. Toxicol. cultural and non-agricultural Hispanic workers. Environ. Health Perspect. 114,
Rev. 24, 37–49. 691–696.
Manthripragada, A.D., Costello, S., Cockburn, M.G., Bronstein, J.M., Ritz, B., 2010. Salvi, R.M., Lara, D.R., Ghisolfi, E.S., Portela, L.V., Dias, R.D., Souza, D.O., 2003.
Paraoxonase 1, agricultural organophosphate exposure, and Parkinson disease. Neuropsychiatric evaluation in subjects chronically exposed to organophosphate
Epidemiol. 21, 87–94. pesticides. Toxicol. Sci. 72, 267–271.
Marks, A.R., Harley, K., Bradman, A., Kogut, K., Barr, D.B., Johnson, C., Calderon, N., Satoh, T., Jokanović, M., 2014. Toxicity and novel biomarkers of OP exposure. In: Balali-
Eskenazi, B., 2010. Organophosphate pesticide exposure and attention in young Mood, M., Abdollahi, M. (Eds.), Basic and Clinical Toxicology of Organophosphorous
Mexican-American children: the CHAMACOS study. Environ. Health Perspect. 118, Compounds. Springer, pp. 119–140.
1768–1774. Savage, E.P., Keefe, T.J., Mounce, L.M., Heaton, R.K., Lewis, J.A., Burcar, P.J., 1988.
Marrs, T.C., Vale, J.A., 2006. Management of organophosphorus pesticide poisoning. In: Chronic neurological sequelae of organophosphate pesticide poisoning. Arch.
Gupta, R.C. (Ed.), Toxicology of Organophosphorus and Carbamate Compounds. Environ. Health 43, 38–45.
Elsevier Academic Press, Amsterdam, pp. 715–733 Chapter 49. Senanayake, N., Karalliedde, L., 1987. Neurotoxic effects of organophosphorus in-
Morgan, J.P., 1982. The Jamaica ginger paralysis. J. Am. Med. Assoc. 248, 1864–1867. secticides: an intermediate syndrome. New Engl. J. Med. 316, 761–763.
Mostafalou, S., Abdollahi, M., 2017. Pesticides: an update of human exposure and toxi- Singh, S., Sharma, N., 2000. Neurological syndromes following organophosphate poi-
city. Arch. Toxicol. 91, 549–599. soning. Neurol. India 48, 308–313.
Narayan, S., Liew, Z., Paul, K., Lee, P.C., Sinsheimer, J.S., Bronstein, J.M., Ritz, B., 2013. Sogorb, M.A., Pamies, D., Estevan, C., Estevez, J., Vilanova, E., 2016. Roles of NTE
Household organophosphorus pesticide use and Parkinson’s disease. Int. J. protein and encoding gene in development and neurodevelopmental toxicity. Chem.
Epidemiol. 42, 1476–1485. Biol. Interact. 259, 352–357.
Pancetti, F., Olmos, C., Dagnino-Subiabre, A., Rozas, C., Morales, B., 2007. Song, F., Xie, K., 2012. Calcium-dependent neutral cysteine protease and organopho-
Noncholinesterase effects induced by organophosphate pesticides and their re- sphate-induced delayed neuropathy. Chem. Biol. Interact. 200, 114–118.
lationship to cognitive processes: implication for the action of acylpeptide hydrolase. Steenland, K., Jenkins, B., Ames, R.G., O’Maley, M., Chrislip, D., Russo, J., 1994. Chronic
J. Toxicol. Environ. Health B 10, 623–630. neurological sequelae to organophosphate poisoning. Am. J. Pub. Health 84,
Parron, T., Hernandez, A.F., Pla, A., Villanueva, E., 1996. Clinical and biochemical 731–736.
changes in greenhouse sprayers chronically exposed to pesticides. Hum. Exp. Toxicol. Suarez-Lopez, J.R., Himes, J.H., Jacobs Jr., D.R., Alexander, B.H., Gunnar, M.R., 2013.
12, 957–963. Acetylcholinesterase activity and neurodevelopment in boys and girls. Pediatrics 132,
Paul, N., Mannathukkaran, T.J., 2005. Intermediate syndrome following carbamate poi- e1649–e1658.
soning. Clin. Toxicol. 43, 867–868. Taghavian, F., Vaezi, G., Abdollahi, M., Malekirad, A.A., 2016. A comparative study of the
Pope, C.N., 2006. Central nervous system effects and neurotoxicity. In: Gupta, R.C. (Ed.), quality of life, depression, anxiety and stress in farmers exposed to organophosphate
Toxicology of Organophosphate and Carbamate Compounds. Elsevier Academic pesticides with those in a control group. J. Chem. Health Risks 6 (2), 143–151.
Press, Amsterdam, pp. 271–291 Chapter 20. Tan, D.H., Peng, S.Q., Wub, Y.L., Wang, Y.M., Lu, C.F., Yan, C.H., 2009. Chronic orga-
Pope, C.N., Chakraborti, T.K., Chapman, M.L., Farrar, J.D., 1992. Long-term neuro- nophosphate (OP)-induced neuropsychiatric disorder is a withdrawal syndrome.
chemical and behavioral effects induced by chlorpyrifos treatment. Pharmacol. Med. Hypotheses 72, 405–406.
Biochem. Behav. 42, 251–256. Tosi, L., Righetti, C., Adami, L., Zanette, G., 1994. October 1942: a strange epidemic
Quistad, G.B., Barlow, C., Winrow, C.J., Sparks, S.E., Casida, J.E., 2003. Evidence that paralysis in Saval, Verona, Italy. Revision and diagnosis 50 years later of tri-ortho-
mouse brain neuropathy target esterase is a lysophospholipase. PNAS 100, cresyl phosphate poisoning. J. Neurol. Neurosurg. Psychiatry 57, 810–813.
7983–7987. van der Hoek, W., Konradsen, F., Athukorala, K., Wanigadewa, T., 1998v. Pesticide
Rauh, V., Arunajadai, S., Horton, M., Perera, F., Hoepner, L., Barr, D.B., Whyattm, R., poisoning: a major health problem in Sri Lanka. Soc. Sci. Med. 46, 495–504.
2011. Seven-year neurodevelopmental scores and prenatal exposure to chlorpyrifos, a van Dongen, C.J., Wolthuis, O.L., 1989v. On the development of behavioral tolerance to
common agricultural pesticide. Environ. Health Perspect. 119, 1196–1201. organophosphates. I: behavioral and biochemical aspects. Pharmacol. Biochem.
Ray, D.E., 1998. Chronic effects of low level exposure to anticholinesterases - a me- Behav. 34, 473–481.
chanistic review. Toxicol. Lett. 102–103, 527–533. Venkategowda, P.M., Harde, Y.R., Rao, M., Mutkule, D.P., Yarramalle, S.P., Mithilesh, K.,
Ray, D.E., Richards, P.G., 2001. The potential for toxic effects of chronic, low-dose ex- Raut, M.K., 2015. Organophosphate poisoning and its neurological manifestations.
posure to organophosphates. Toxicol. Teratol. 120, 343–351. Int. J. Adv. Case Rep. 2 (21), 1303–1305.
Ruckart, P.Z., Kakolewski, K., Bove, F.J., Kaye, W.E., 2004. Long-term neurobehavioral Vose, S.C., Fujioka, K., Gulevich, A.G., Lin, A.Y., Holland, N.T., Casida, J.E., 2008.
health effects of methyl parathion exposure in children in Mississippi and Ohio. Cellular function of neuropathy target esterase in lysophosphatidylcholine action.
Environ. Health Perspect. 112, 46–51. Toxicol. Appl. Pharmacol. 232, 367–383.
Read, D.J., Li, Y., Chao, M.V., Cavanagh, J.B., Glynn, P., 2009. Neuropathy target esterase Worek, F., Diepold, C., Eyer, P., 1999. Dimethylphosphoryl-inhibited human cholines-
is required for adult vertebrate axon maintenance. J. Neurosci. 29, 11594–11600. terases: inhibition, reactivation, and aging kinetics. Arch. Toxicol. 73, 7–14.
Reiner, E., Pleština, R., 1979. Regeneration of cholinesterase activities in humans and rats World Health Organization, 1986. Organophosphorus Insecticides: A General
after inhibition by O,O-dimethyl-O-dimethyl-2,2-dichlorvinyl phosphate. Toxicol. Introduction, vol. 63 Environmental Health Criteria, Geneva.
Appl. Pharmacol. 49, 451–454. Yanagisawa, N., Morita, H., Nakajima, T., 2006. Sarin experiences in Japan: acute toxicity
Riddle, J.R., Brown, M., Smith, T., Ritchie, E.C., Brix, K.A., Romano, J., 2003. Chemical and long-term effects. J. Neurol. Sci. 249, 76–85.
warfare and the Gulf war: a review of the impact on Gulf veterans’ health. Mil. Med. Yang, C.C., Deng, J.F., 2007. Intermediate syndrome following organophosphate in-
168, 606–613. secticide poisoning. J. Chin. Med. Assoc. 70, 467–472.
Rohlman, D.S., Anger, W.K., Lein, P.J., 2011. Correlating neurobehavioral performance Yilmaz, M., Sebe, A., Ay, M.O., Gumusay, U., Topal, M., Atli, M., Icme, F., Satar, S., 2013.
with biomarkers of organophosphorous pesticide exposure. Neurotoxicology 32, Effectiveness of therapeutic plasma exchange in patients with intermediate syndrome
268–276. due to organophosphate intoxication. Am. J. Emerg. Med. 31, 953–957.
Roldan-Tapia, L., Parron, T., Sanchez-Santed, F., 2005. Neuropsychological effects of Yu, C.J., Du, J.C., Chiou, H.C., Chung, M.Y., Yang, W., Chen, Y.S., Fuh, M.R., Chien, L.C.,
long-term exposure to organophosphate pesticides. Neurotoxicol. Teratol. 27, Hwang, B., Chen, M.L., 2016. Increased risk of attention-deficit/hyperactivity dis-
259–266. order associated with exposure to organophosphate pesticide in Taiwanese children.
Rosenstock, L., Keifer, M., Daniell, W., McConnell, R., Claypoole, K., 1991. Chronic Androl. 4, 695–705.
central nervous system effects of acute organophosphate pesticide intoxication. Zaganas, I., Kapetanaki, S., Mastorodemos, V., Kanavouras, K., Colosio, C., Wilks, M.F.,
Lancet 338, 223–227. Tsatsakis, A.M., 2013. Linking pesticide exposure and dementia: What is the evi-
Ross, S.M., McManus, I.C., Harrison, V., Mason, O., 2013. Neurobehavioral problems dence? Toxicology 307, 3–11.

131

Descargado para LUIS HERRERA GUTIERREZ (l.herrera1406@gmail.com) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en mayo 17, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.

You might also like