Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jul 1.
Published in final edited form as: Food Chem Toxicol. 2017 May 6;105:456–474. doi: 10.1016/j.fct.2017.05.003

Potential contribution of insecticide exposure and development of obesity and type 2 diabetes

Xiao Xiao 1, John M Clark 2, Yeonhwa Park 1,*
PMCID: PMC5518693  NIHMSID: NIHMS877455  PMID: 28487232

Abstract

The introduction of insecticides has greatly improved agricultural productivity and human nutrition; however, the wide use of insecticides has also sparked growing concern over their health impacts. Increased rate of cancers, neurodegenerative disorders, reproductive dysfunction, birth defects, respiratory diseases, cardiovascular diseases and aging have been linked with insecticide exposure. Meanwhile, a growing body of evidence is suggesting that exposure to insecticides can also potentiate the risk of obesity and type 2 diabetes. This review summarizes the relationship between insecticide exposure and development of obesity and type 2 diabetes using epidemiological and rodent animal studies, including potential mechanisms. The evidence as a whole suggests that exposure to insecticides is linked to increased risk of obesity and type 2 diabetes.

Keywords: Insecticide, obesity, glucose metabolism, type 2 diabetes

1. Introduction

The prevalence of obesity among adults in the United States more than doubled since the 1960s, increasing from 13.4 in 1962 to 35.7 percent in 2010 (NCHS, 2012). As obesity is a well known risk factor for other diseases, especially type 2 diabetes, the incidences of type 2 diabetes are also rising (ADA, 2013; CDC, 2014; WHO, 2013). The current epidemic of obesity and type 2 diabetes cannot be fully explained by changes in societal, genetic, behavioral, or dietary habits of individuals, suggesting unknown factors contribute to these disease outbreaks.

The challenge of growing more food to feed the world’s expanding population has driven need to control pest insects, which are harmful to crop yield. Application of insecticides are considered to be a major contributing factor for increased agricultural productivity in the 20th century (van Emden and Peakall, 1996). In addition to its role in agriculture, insecticides are widely used in industry, households, and military to control insect pests that are disease vectors, suggesting its essential role in human life (Sparks, 2013). However, insecticides are one of the major environmental contaminations and the extensive use of insecticides has caused wide public concern over their potential risk of inducing human chronic diseases, including obesity and diabetes (Hectors et al., 2011; Karami-Mohajeri and Abdollahi, 2011; Kuo et al., 2013; Lee et al., 2014a; Rezg et al., 2010b). Among insecticides, organochlorines and organophosphorus are the most investigated insecticides linked with obesity and/or type 2 diabetes in humans and rodents (Karami-Mohajeri and Abdollahi, 2011). Although limited, other types of insecticides, such as carbamates, pyrethroids, and neonicotinoids, are associated with development of obesity and/or type 2 diabetes as well (Montgomery et al., 2008; Narendra et al., 2008; Saldana et al., 2007; Sun et al., 2016b; Wang et al., 2011b). More recently, in vitro and in vivo studies reported that pyrethroids, a neonicotinoid, and a phenylpyrazole (fipronil), all were involved in potentiated adipogenesis and/or altered glucose responsiveness, as representative of obesity and type 2 diabetes, respectively (Kim et al., 2013, 2014a; Park et al., 2013; Shen et al., 2017; Sun et al., 2016a; Sun et al., 2016b). Moreover, reports of interaction between dietary fat and insecticides on these markers, further begs the question of the role of insecticides and health. Thus, this review focuses on summarizing the current reports on insecticide exposure and development of obesity and type 2 diabetes, including suggested mechanisms, to further expand our current understanding.

2. Methods

Bibliographic databases including PubMed and Web of Science were searched for the keywords ‘insecticide’ and ‘obesity’; ‘insecticide’ and ‘diabetes’; ‘insecticide’ and ‘body weight’; ‘insecticide’ and ‘glucose metabolism’; and ‘insecticide’ and ‘lipid metabolism’. Data were collected from 1966 to December 2016. From the initial search of PubMed and Web of Science, each article was reviewed to evaluate title and abstract content, and to eliminate duplicates and those were not related to our purposes. A total of 111 articles were selected and their full texts were reviewed including 52 human studies and 59 animal studies. Our exclusion criteria were (1) publications containing no original research (reviews, editorials, or non-research letters); (2) studies not carried out in humans, mice, or rats; or (3) human studies without providing information on markers of obesity or diabetes as described below. We collected the following data for human studies: authors, journal, year of publication, country, insecticide, study design, study population, changes in body weight and/or body mass index, waist circumference, diabetes risk (increased blood glucose levels, insulin resistance, or gestational diabetes), lipid markers (triglycerides and/or cholesterols), and other critical comments. Variables extracted from animal studies include: authors, journal, year of publication, species, treatment methods (dose, route, and duration), sex, body weight change, major markers of glucose homeostasis (glucose, insulin, or insulin resistance) and lipid metabolism (triglycerides and/or cholesterol), and other metabolic markers involved in glucose and lipid metabolisms (e.g., blood leptin, other lipids, glycogen content, markers of adipogenesis, gluconeogenesis, glycogenolysis, glycolysis, and/or inflammation). The findings are presented in Tables.

3. Insecticide classification and action

Insecticides can be classified based primarily on their chemical structures and mode of actions. Major classes of insecticides include organochlorines, organophosphorus, carbamates, pyrethroids, and neonicotinoids. A few examples are shown in Table 1.

Table 1.

Major classes of insecticides with examples and their structures

Organochlorines Organophosphorus Carbamates Pyrethroids Neonicotinoids
Aldrin Chlorpyrifos Aldicarb Allethrin Acetamiprid
DDT/DDE Diazinon Bendiocard Bifenthrin Clothanidin
Dieldrin Dichlorvos Carbaryl Cismethrin Dinotefuran
Heptachlor Malathion Dioxacarb Cyhalothrin Imidacloprid
Lindane Parathion Fenobucarb Deltamethrin Niternpyram
Methoxychlor Profenofos Isoprocarb Permethrin Thiacloprid
Methomyl Tefluthrin Thiamethoxam

Organochlorine insecticides can be divided into dichlorodiphenyltrichloroethane (DDT)-type and chlorinated alicyclic-type (cyclodienes) based on their distinctive mechanisms of action. DDT-type insecticides (such as DDT, DDE, and DDD) are known to inhibit the closing of voltage-sensitive sodium channel (VSSC) in neurons, resulting in repetitive firing of action potentials, while chlorinated alicyclic-type insecticides (e.g. aldrin, dieldrin, heptachlor, and endosulfan) bind to γ-aminobutyric acid (GABA) chloride ionophore complex, which inhibits chloride influx into the nerve (Coats, 1990; Karami-Mohajeri and Abdollahi, 2011). Even though most countries have not used organochlorines for the last several decades, due to extremely stable chemical characteristics, DDE (as a major metabolite of DDT) can currently be found in human serum, adipose tissue, and many foods (Karami-Mohajeri and Abdollahi, 2011; USDA, 2014).

Organophosphorus insecticides are irreversible inhibitors of cholinesterases, including acetyl cholinesterase, resulting in hyper-stimulation of cholinergic nerves (e.g. muscarinic and nicotinic acetylcholine receptor) (Abou-Donia, 2003). As the largest insecticide class in the world in 1980s, organophosphorus insecticides occupied 71% of world insecticides market in 1987; however, the use of organophosphorus insecticides dropped to around 52% in 1999 and to 13% in 2013 due to its environmental persistence and mammalian toxicity (Casida and Quistad, 2004; Nauen and Bretschneider, 2002; Sparks, 2013).

Carbamates, which account for 6% of global insecticides (Sparks, 2013), have the similar mechanism of action with organophosphorus insecticides, but their neurotoxic effects are relatively more moderate than organophosphorus because the inhibition of acetyl cholinesterase is reversible and carbamates are known to be rapidly metabolized by human and animals (Karami-Mohajeri and Abdollahi, 2011; Risher et al., 1987).

Pyrethroids are structural analogs to naturally occurring insecticide, pyrethrin, found in Chrysanthemum flower heads. Pyrethroids can cause over excitation of the neuron by delaying the closing of VSSC, producing an effect similar to, but more pronounced than, DDT due to its better sodium channel binding capacity (Davies et al., 2007; Soderlund et al., 2002). By 2013, pyrethroids accounted for approximately 17% of the global insecticide market (Sparks, 2013).

Neonicotinoids are a relatively new family of insecticides with structural resemblance to nicotine (Casida and Durkin, 2013). Acting on nicotinic acetylcholine receptors, neonicotinoids can stimulate these receptors at low doses, while blocking these receptors at high doses, leading to paralysis and death (Gervais et al., 2012). Neonicotinoids have become the fastest growing class of insecticides, representing ~27% of the global insecticide market in 2013, which makes them the largest single insecticide class on the market (Sparks, 2013; Sparks and Nauen, 2015). Due to the potential link between use of neonicotinoids and the reduction of bee population, the European Commission has banned use of three neonicotinoids (imidaclorprid, thiamethoxam, and clothianidin) in 2013 (The European Commision).

4. Effects of insecticide exposure on glucose and lipid metabolisms

4.1. Effects of insecticides on the risk of diabetes in human

Numerous epidemiological studies have found a potential association between insecticide exposure with increased risks of diabetes (summarized in Table 2). For this table, we have included elevated fasting blood glucose levels and insulin resistance as evidence of diabetes risk. Organochlorine and organophosphorus insecticides are the most studied, but few others reported on the role of pyrethroids, carbamates and neonicotinoids on diabetes.

Table 2.

Effects of insecticide exposure on risk of diabetes in human

Insecticidea Country Subject Risk of
diabetesc
Other
comments
Reference
Organochlorine
Aldrin USA Pesticide applicators (Montgomery et al., 2008)
Chlordane USA General population (Lee et al., 2006)
USA Pesticides applicators (Montgomery et al., 2008)
p,p′-DDD South Korea Age ≥ 40 × (Son et al., 2010)
p,p′-DDE Sweden Aged 50–74 years old × (Glynn et al., 2003)
Sweden Fishermen and theirwives (Rylander L, 2005)
USA General population (Lee et al., 2006)
USA Native Americans (Codru et al., 2007)
USA Mexican Americans (Cox et al., 2007)
Sweden Fishermen’s wives (Rignell-Hydbom et al., 2007)
USA Non-diabetic c (Lee et al., 2007b)
Canada First Nation Community members (Philibert et al., 2009)
Sweden 50–59 years old (Rignell-Hydbom et al., 2009)
USA Great Lakes sport fish consumer (Turyk et al., 2009)
USA General population (Everett and Matheson, 2010)
USA African and White Americans × (Lee et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
Slovakia Polluted area (Ukropec et al., 2010)
Finland 57–70 years old (Airaksinen et al., 2011)
Belgium Hospital patients, staff and volunteers × (Dirinck et al., 2011)
USA Non-diabetic d (Lee et al., 2011b)
Sweden Age > 70 × (Lee et al., 2011a)
Denmark General population (Faerch et al., 2012)
Spain General population × (Gasull et al., 2012)
Spain Hospital patients (Arrebola et al., 2013)
Canada First Nations community members × ↑ Blood DDE in diabetic individuals (Pal et al., 2013)
Benin 18–65 years old (Azandjeme et al., 2014)
Russia 8–9 years old boys ×d (Burns et al., 2014)
Belgium General population (Dirinck et al., 2014)
South Korea Hospital patients (Kim et al., 2014b)
Slovakia Polluted area (Langer et al., 2014)
Thailand General population (Teeyapant et al., 2014)
Saudi Arabia 30–50 years old (Al-Othman et al., 2015)
USA Great Lake Sport fish consumer (Turyk et al., 2015)
Belgium Aged 50–65 years old only in men (Van Larebeke et al., 2015)
France Newborns × ↓ Insulin & adiponectin level in female newborns (Debost-Legrand et al., 2016)
Canada Pregnant women ×e (Shapiro et al., 2016)
o,p'-DDE Denmark General population (Faerch et al., 2012)
p,p'-DDT USA Mexican Americans (Cox et al., 2007)
USA General population (Everett et al., 2007)
USA Non-diabetic ×d (Lee et al., 2007a)
USA General population (Everett and Matheson, 2010)
USA African and White Americans × (Lee et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
Slovakia Polluted area (Ukropec et al., 2010)
USA Non-diabetic ×d (Lee et al., 2011b)
Denmark General population (Faerch et al., 2012)
Spain General population × (Gasull et al., 2012)
Benin 18–65 years old (Azandjeme et al., 2014)
South Korea Hospital patients (Kim et al., 2014b)
Saudi Arabia 30–50 years old (Al-Othman et al., 2015)
o,p′-DDT South Korea Age ≥ 40 (Son et al., 2010)
Denmark General population (Faerch et al., 2012)
Dieldrin USA General population × (Everett and Matheson, 2010)
HCB Sweden 50–74 years old (Glynn et al., 2003)
USA Native Americans (Codru et al., 2007)
USA Mexican Americans × (Cox et al., 2007)
USA African and White Americans × (Lee et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
Slovakia Polluted area × ↑ only prediabetes (Ukropec et al., 2010)
USA Non-diabetic ×d (Lee et al., 2011b)
Sweden Age > 70 × (Lee et al., 2011a)
Denmark General population (Faerch et al., 2012)
Spain General population (Gasull et al., 2012)
Spain Hospital patients × (Arrebola et al., 2013)
Canada First Nations community members × ↑ Blood HCB (Pal et al., 2013)
Russia 8–9 years old boys ↑d (Burns et al., 2014)
Slovakia Polluted area (Langer et al., 2014)
South Korea > 40 years old (Lee et al., 2014b)
South Korea Hospital patients (Kim et al., 2014b)
Belgium 50–65 years old (Van Larebeke et al., 2015)
cis-nonachlor Canada First Nations community members (Pal et al., 2013)
South Korea Hospital patients (Kim et al., 2014b)
CB-153 Sweden Fishermen and their wives only in men (Rylander L, 2005)
USA General population (Lee et al., 2006)
β-HCH Sweden 50–74 years old × (Glynn et al., 2003)
USA Mexican Americans (Cox et al., 2007)
USA Non-diabetic ×d (Lee et al., 2007a)
USA Non-diabetic ×c (Lee et al., 2007b)
USA General population (Everett and Matheson, 2010)
USA African and White Americans × (Lee et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
Slovakia Polluted area × ↑ only prediabetes (Ukropec et al., 2010)
Belgium Hospital patients, staff and volunteers (Dirinck et al., 2011)
USA Non-diabetic ×d (Lee et al., 2011b)
Denmark General population × (Faerch et al., 2012)
Spain General population × (Gasull et al., 2012)
Spain Hospital patients (Arrebola et al., 2013)
Canada First Nations community members × ↑ Blood β-HCH (Pal et al., 2013)
Benin 18–65 years old (Azandjeme et al., 2014)
Saudi Arabia 18–65 years old (Al-Othman et al., 2014)
Russia 8–9 years old boys ×d (Burns et al., 2014)
South Korea Aged > 40 years (Lee et al., 2014b)
South Korea Hospital patients × (Kim et al., 2014b)
ɣ-HCH USA African and White Americans × (Lee et al., 2010)
USA Non-diabetic ×d (Lee et al., 2011b)
Saudi Arabia 18–65 years old (Al-Othman et al., 2014)
Heptachlor USA Pesticide applicators (Montgomery et al., 2008)
Heptachlor epoxide (A metabolite of heptachlor) USA General population (Everett and Matheson, 2010)
USA General population (Patel et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
South Korea Age > 40 (Lee et al., 2014b)
Mirex USA Native Americans (Codru et al., 2007)
USA General population × (Everett and Matheson, 2010)
USA African and White Americans (Lee et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
USA Non-diabetic ×d (Lee et al., 2011b)
Canada First Nations community members × ↑ Blood mirex in diabetic individuals (Pal et al., 2013)
Oxychlordane Sweden 50–74 years old × (Glynn et al., 2003)
USA General population (Lee et al., 2006)
USA Mexican Americans (Cox et al., 2007)
USA Non-diabetic d (Lee et al., 2007a)
USA Non-diabetic ×c (Lee et al., 2007b)
USA General population (Everett and Matheson, 2010)
USA African and White Americans × (Lee et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
Finland 57–70 years old (Airaksinen et al., 2011)
USA Non-diabetic ×d (Lee et al., 2011b)
Canada First Nations community members ↑ Blood oxychlordane in diabetic individuals (Pal et al., 2013)
South Korea Aged > 40 years (Lee et al., 2014b)
Canada Pregnant women ×e (Shapiro et al., 2016)
TNC Sweden 50–74 years old × (Glynn et al., 2003)
USA General population (Lee et al., 2006)
USA Mexican Americans (Cox et al., 2007)
USA Non-diabetic d (Lee et al., 2007a)
USA Non-diabetic ×c (Lee et al., 2007b)
USA General population (Everett and Matheson, 2010)
USA African and White Americans × (Lee et al., 2010)
South Korea Age ≥ 40 (Son et al., 2010)
Finland 57–70 years old (Airaksinen et al., 2011)
USA Non-diabetic ×d (Lee et al., 2011b)
Sweden age > 70 (Lee et al., 2011a)
Canada First Nations community members (Pal et al., 2013)
Benin 18–65 years old (Azandjeme et al., 2014)
Canada Pregnant women ×e (Shapiro et al., 2016)
Organophosphorus
Dialkylphosphate (Metabolites) France Newborns × ↑ Insulin level (Debost-Legrand et al., 2016)
Dimethylphosphate (Metabolites) Canada Pregnant women e (Shapiro et al., 2016)
Dimethylthiophosphate (Metabolites) Canada Pregnant women e (Shapiro et al., 2016)
Mixture Turkey Overdose patients c Case report (Amanvermez et al., 2010)
Iran Farmers (Malekirad et al., 2013)
India A 15-year-old girl c Case report (Swaminathan et al., 2013)
Diazinon Israel Children c (Weizman and Sofer, 1992)
USA Wives of pesticide applicators e (Saldana et al., 2007)
Dichlorvos USA Pesticides applicators (Montgomery et al., 2008)
Malathion Canada 81-year-old mother and her 39-year old son c Case report (Meller et al., 1981)
USA Wives of pesticide applicators ×e (Saldana et al., 2007)
Egypt Non-diabetic male famers (Raafat et al., 2012)
Phorate USA Wives of pesticide applicators e (Saldana et al., 2007)
Terbufos USA Wives of pesticide applicators × (Saldana et al., 2007)
Trichlorfon USA Pesticides applicators (Montgomery et al., 2008)
Carbamates
Carbaryl USA Wives of pesticide applicators ×e (Saldana et al., 2007)
Carbofuran USA Wives of pesticide applicators e (Saldana et al., 2007)
Pyrethroids
Allethrin India Mosquito repellent coils or mats users c (Narendra et al., 2008)
Permethrin USA Pesticides applicators × (Montgomery et al., 2008)
Prallethrin India Mosquito repellent coils or mats users c (Narendra et al., 2008)
Pyrethroid mixture China Pesticide factory workers (Wang et al., 2011a)
Pyrethroid mixture Bolivia Male pesticide sprayers (Hansen et al., 2014)
Other insecticide
Amitraz Turkey Overdose patients c Case report (Avsarogullari et al., 2006)
Mixture of chlopyrifos & cypermethrin Morocco A 30-year-old man c Case report (Badrane et al., 2014)
a

Abbreviation for insecticides: 2,2′-bis (4-chlorophenyl)-1,1-dichlorodiethylene (p,p'-DDD), 2,2′-bis (4-chlorophenyl)-1,1-dichloroethylene (p,p'- DDE); 2,2′-bis (4-chlorophenyl)-1,1,1-trichloro-ethane (p,p'- DDT); Hexachlorobenzene (HCB); 2,2’,4,4’,5,5’-Hexachlorobiphenyl (CB-153); β-Hexachlorocyclohexane (β-HCH); ɣ-Hexachlorocyclohexane (ɣ-HCH); trans-nonachlor (TNC).

b

Abbreviation: ↑, increase; ×, no association with; Gestational diabetes mellitus (GDM).

Risk of diabetes including elevated blood glucose levelc, insulin resistanced, and gestational diabetes mellituse.

Most studies of organochlorines linked the blood levels of organochlorines or their metabolites with increased risk of diabetes in humans, as these are known to be persistent in human tissues, particularly fatty tissues (Table 2). Overall there is strong indication that exposure to these insecticides leads to increased risk of diabetes. In addition, Boada et al. (Boada et al., 2007) reported a negative correlation between blood levels of aldrin and p,p’-DDD and insulin-like growth factor-1, which are also known to have implications for development of insulin resistance (Aguirre et al., 2016).

Organophosphorus and carbamate insecticides have also been generally linked with increased risk of diabetes in humans, including pesticide applicators who were exposed to dichlorvos and trichlorfon (Montgomery et al., 2008). Interestingly, Saldana et al. (Saldana et al., 2007) reported that wives of pesticide applicators of organophosphorus insecticides (diazinon and phorate) along with carbamate insecticide (carbofuran) have increased risks of gestational diabetes mellitus, while no association was found for other organophosphorus (malathion and terbufos) or carbamate insecticide (carbaryl). In addition, a study reported that patients with acute organophosphorus poisoning due to suicide attempts had hyperglycemia (Amanvermez et al., 2010).

Reports on the effects of pyrethroids on diabetes from human studies are rather limited and no human study on neonicotinoids and diabetes is available currently. One study reported that pyrethroid mixture increases the risk of diabetes among pesticide factory workers in China (Wang et al., 2011a). Another study reported that exposure to pyrethroid insecticides (allethrin and prallethrin) increased plasma glucose levels in Indian men (Narendra et al., 2008). Since neonicotinoids have been used in the last few decades and they are designed to be relatively quickly degraded in biological systems, it is more challenging to investigate the exposure to neonicotinoids and human health perspectives (Casida, 2011). Overall, the majority of human studies have shown a positive association between exposure to certain type of insecticides and risk of diabetes.

4.2. Effects of insecticides on the risk of obesity in human

Compared to the large number of epidemiological studies on insecticide and diabetes, a relatively small number of studies reported a link between insecticide exposure and obesity (summarized in Table 3). Three reported positive association between organochlorine exposure (DDE and β-hexachlorocyclohexane) and body mass index (BMI) (Dirinck et al., 2011; Lee et al., 2011b; Mendez et al., 2011), while others found no association between organochlorine insecticide exposure (including DDE and β-HCH) and BMI (Dirinck et al., 2011; Glynn et al., 2003; Lee et al., 2011b; Mendez et al., 2011).

Table 3.

Effects of insecticide exposure on body weight change and other lipid markers in humans

Insecticidea Study information Resultsb Reference

Country Description BMI Others
Organochlorine

p,p’-DDE Sweden Women = (Glynn et al., 2003)

USA Non-diabetic N/A = TG (Lee et al., 2007b)

Belgium Obese and lean men & women = (Dirinck et al., 2011)

USA African and White Americans ↑ TG (Lee et al., 2011b)
↓ HDL-C

Spain Women in early pregnancy & their newborn children ↑ Weight in the first 6 months; (Mendez et al., 2011)
↑ BMI at 14 months in infancy

Denmark General Population N/A ↑ Lipid oxidation; ↑FFA (Faerch et al., 2012)

Slovakia Polluted area ↑ TG & cholesterol; ↓ testosterone in males (Langer et al., 2014)

Belgium Aged 50–65 years old Only in men (Van Larebeke et al., 2015)

p,p’-DDT USA African and White Americans = = TG (Lee et al., 2011b)
= HDL-C

HCB Sweden Women = (Glynn et al., 2003)

USA African and White Americans = = TG (Lee et al., 2011b)
= HDL-C

Spain Women in early pregnancy & their newborn children = (Mendez et al., 2011)

Denmark General Population N/A ↑ Lipid oxidation (Faerch et al., 2012)

Saudi Adults 30–50 years old N/A ↑ TG; ↓ HDL-cholesterol (Al-Othman et al., 2014)

Slovakia Polluted area ↑ TG & cholesterol; ↓ testosterone in males (Langer et al., 2014)

Belgium Aged 50–65 years old Only in women (Van Larebeke et al., 2015)

β-HCH Sweden Women = (Glynn et al., 2003)

USA Non-diabetic N/A = TG (Lee et al., 2007b)

Belgium Obese and lean men & women ↑ Waist & subcutaneous abdominal fat mass (Dirinck et al., 2011)

USA African and White Americans = = TG (Lee et al., 2011b)
= HDL-C

Spain Women in early pregnancy & their newborn children = (Mendez et al., 2011)

ɣ-HCH USA African and White Americans = = TG (Lee et al., 2011b)
= HDL-C

Oxychlordane Sweden Women = (Glynn et al., 2003)

USA Non-diabetic N/A ↑ TG (Lee et al., 2007b)

USA African and White Americans = ↑ TG (Lee et al., 2011b)
= HDL-C

TNC Sweden Women = (Glynn et al., 2003)

USA Non-diabetic N/A = TG (Lee et al., 2007b)

USA African and White Americans = = TG (Lee et al., 2011b)
= HDL-C

Mirex USA African and White Americans = = TG (Lee et al., 2011b)
= HDL-C

Organophosphorus

Malathion Egypt Non-diabetic male famers ↑ Waist circumference (Raafat et al., 2012)

Pyrethroids

Allethrin India Men N/A ↑ TG, phospholipids, lipid peroxides, & VLDL-C; (Narendra et al., 2008)
↓ Cholesterol &glycolipids;
= HDL-C & LDL-C

Prallethrin India Men N/A ↑ TG, phospholipids, lipid peroxides, & VLDL-C; (Narendra et al., 2008)
↓ Cholesterol & glycolipids;
= HDL-C & LDL-C
a

Abbreviation for insecticides: 2,2′-bis (4-chlorophenyl)-1,1-dichloroethylene (p,p'- DDE); 2,2′-bis (4-chlorophenyl)-1,1,1-trichloro-ethane (p,p'- DDT); Hexachlorobenzene (HCB); β-Hexachlorocyclohexane (β-HCH); ɣ-Hexachlorocyclohexane (ɣ-HCH); trans-nonachlor (TNC).

b

Abbreviation for results: ↑, increase; ↓, decrease; =, no change; BMI, body mass index; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; N/A, not available; TG, triglyceride; VLDL-C, very low-density lipoprotein cholesterol.

For other markers of lipid metabolism, there were no effects of organochlorines on high-density lipoprotein cholesterol (HDL-cholesterol), except one study reported negative correlation between DDE and HDL (Lee et al., 2007b). Others reported that pyrethroid insecticides, allethrin and prallethrin, were linked with disturbed lipid metabolism by increasing triglycerides, phospholipids, very low-density lipoprotein cholesterol (VLDL-C), but no effects on HDL (Narendra et al., 2008).

4.3. Effects of insecticides on glucose and lipid metabolisms in animals

Although different classes of insecticides have slightly different mechanism of insecticidal actions, many share common characteristics by acting on the nerve system. Common symptoms after exposure to insecticides include spasm, muscular tremors, and convulsions (Casida and Durkin, 2013; Nauen and Bretschneider, 2002). Overstimulation of the nervous system increases the energy demands and disturbs the functions of several organs, resulting in the disorder of energy homeostasis that can lead to altered glucose and lipid metabolisms (Matin et al., 1990a; Matin et al., 1990b; Pournourmohammadi et al., 2007; Rezg et al., 2008).

4.3.1. Effects of insecticides on blood glucose level in rodents

Studies have shown that exposure to all major types of insecticides induced hyperglycemia in experimental mice and/or rats (summarized in Table 4). Among them, organochlorine and organophosphorus compounds are the most documented with relatively less reports on carbamates, pyrethroids, and neonicotinoids. Studies have demonstrated that exposure to certain organochlorines, organophosphorus, pyrethroids, and neonicotinoids elevated blood glucose levels, although inconsistent results have also been reported (Table 4). This inconsistency might derive from the difference in dose and route of exposure, animal species, exposure duration, as well as methods to determine insulin resistance (Rezg et al., 2006). In fact, others suggested that insecticide-induced hyperglycemia is only temporary: blood glucose concentration increased initially and then decreased with the possibility of reaching hypoglycemia (Gupta, 1974; Rezg et al., 2006; Rezg et al., 2007). More importantly, our group recently reported that low doses of orally administrated permethrin or imidacloprid (levels lower than NOAEL) potentiate insulin resistance, only when high-fat diet was provided in mice (Sun et al., 2016b; Xiao et al., 2015; Xiao et al., 2016). These results suggest that low dose insecticide exposure should be evaluated along with other factors contributing to development of diabetes.

Table 4.

Effects of insecticide-induced alteration of glucose and lipid metabolisms in mice and rats

Insecticide Treatment Sexd Resultse Reference


Dose
(mg/kg a)
Route
b
Durationc BW Glucose Insulin TG Cholesterol Other comments
Mice
Organochlorine

DDE 0.4 & 2 Oral Daily for 5 d, tested after 7, 14, 21 & 28d M ↑ (2) ↑ (2; 7 & 21 d) = N/A N/A = Glucagon; (Howell et al., 2014)
= Insulin resistance;
= pAkt Ser473/Akt (liver, muscle & fat);
= Leptin & resistin

2 Oral Daily for 5d, 1 wk rest, +13 wk HFD or LFD w/o DDE M = ↑ in HFD (wk 4 & 8) ↓ in HFD = = = Leptin & resistin; (Howell et al., 2015)
Muscle:
↑ Glut4 in HFD;
Fat:
= Glut4;
Liver:
↓ Lipogenesis in HFD;
↑ FA oxidation in HFD;
↑ FA uptake in HFD;
↓ Gluconeogenesis in HFD

DDT 50 Oral Single, tested after 0–18h & 7 d M N/A ↓ 5–7h, ↑ Baseline glucose in GTT after 7 d = (18h) N/A N/A ↓ Glucose tolerance at 18h; (Yau and Mennear, 1977)
↓ Insulin secretion from isolated islet of pancreas at 18h, but not at 7d

1.7 Oral Daily for 16 d (GND11.5-PND5) F & pups ↓ in ♂ (PND5); ↓ (PND5); ↑ in ♀ w/ HFD; ↑ in ♀ w/ HFD ↑ in ♀ w/ HFD; ↑ Insulin resistance in ♀ w/ HFD & ♂ w/ LFD; (La Merrill et al., 2014)
↑ Fat mass in adult ♀;
↑ in adult ♀ = (6 m post-wean) = (6 m post-wean) = Lipid level (during 6 m of post-weaning);
= Food intake;
= FFAs

Organophosphorus

Chlorpyrifos 2 Oral (Diet) 8 wk Mf = ↑ Insulin resistance; (Peris-Sampedro et al., 2015)
↑ Food intake;
↑ Leptin in ApoE3f

Diazinon 6.5 (1/10 LD50) i.p. 5 times weekly for 7 wk M N/A N/A Tea and olive leaves extract prevented diazinon-disturbed glucose and lipid metabolism (Al-Attar and Abu Zeid, 2013)

Carbamates

Furadan 0.125, 0.25, & 0.5 i.p. Weekly for 2,4, & 6 wk M N/A N/A N/A ↑ Lipid content in serum, liver & kidney; (Gupta et al., 1986)
↑ Phospholipid in serum, liver, & kidney;
↓ Phospholipid in brain;
= FFAs;
↓ Lipase activity in liver and serum

Pyrethroids

Cypermethrin 10 Oral Daily for 28 d M N/A ↑; (Ince et al., 2012)
↑ VLDL, ↓ HDL

Deltamethrin 1 & 3 (1/4 of NOAEL) Oral (Diet) Every 3 d in gestation & lactation in dams F & ♂ pups = in ♂ pups N/A N/A N/A N/A Gene expression in fat of ♂ pups: (Armstrong et al., 2013)
↓ Glucose transport: Glut4 (1) & Glut2;
↓ Lipogenic gene (1) : SREBP1, ACC-1, FABP4, CD36, LPL & SCD-1;
↓ Adipogenesis (1): PPARγ & CEBPα

Neonicotinoids

Imidacloprid 5,10, &15 Oral Daily for 15 d M ↓ (15) N/A N/A N/A N/A ↑ Liver & kidney weight (15) (Arfat; et al., 2014)

0.06, 0.6 & 6 Oral Daily for 12 wk M = ↑ Leptin; = FFA; (Sun et al., 2016b)
Fat:
↑ cell size; ↑ CD36, SREBP1, TNFα;
↓ CaMKKβ, pAMPKα/AMPKα, pACC/ACC;
Liver:
↑ PEPCK; ↓ CaMKKβ, PPARα, pAMPKα/AMPKα, pACC/ACC, Sirt1, PGC-1α;
Muscle:
↓ GLUT4; ↑ PDK4; = CPT1

Rats
Organochlorine

DDT 1 Oral (Diet) 180 d M N/A N/A; ↓ in sedentary rats; N/A ↓ in sedentary rats; ↑ in exercised rats; = Food intake, carcass & liver lipids; (Berdanier and de Dennis, 1977)
Significant interaction DDT × Exercise Significant interaction DDT × Exercise Significant interaction DDT × Exercise (glucose tolerance)

20 Oral Daily for 189 d F = N/A N/A N/A N/A (Chadwick et al., 1988)

25 & 50 i.p. Daily for 6 d (GND8-14) F/M ↑ in F3 N/A N/A N/A N/A Gestational rats (F0) were treated and the third generation (F3) were observed (Skinner et al., 2013)

Dieldrin 30 Oral Single, test after 24h M = N/A N/A N/A N/A ↑ Liver glycogen & TG; (Bhatia and Venkitasub ramanian, 1972)
= Liver phospholipid & cholesterol

2 Oral 6 m M N/A N/A = (Shakoori et al., 1984)

HCB *20 & 100; Oral (Diet) 4 wk F/M ↑ ♂ (40) w/ food deprivation, ↑ in ♂ (20 & 100) w/o food deprivation N/A N/A N/A N/A = Food intake; (Villeneuve et al., 1977)
40 & 200 w/ food deprivation; ↑ Liver weight w/ food restriction (20);
↑ Liver hypertrophy w/ food deprivation (200)

ɣ-HCH (lindane) 5,10,20, & 40 Oral Daily for 189 d; F ↑ (20 & 40) N/A N/A N/A N/A ↑ Food intake & food efficiency (20 & 40); (Chadwick et al., 1988)
daily for 15 wk ↑ Liver weight;
40 caused death

Organophosphorus

Acephate 2.5 s.c. Daily for 8 wk M N/A N/A N/A N/A ↓ Liver glycogen; (Deotare and Chakrabarti, 1981)
↑ Pyruvic acid and lactic acid in liver, heart, kidney, brain & blood

600 Oral Daily for 8 wk F/M N/A N/A N/A = ↓ Total lipids; (Choudhari and Chakrabarti, 1984)
Liver:
↑ Total lipids (↓ microsome, = mitochondria, ↑ cytosol);
↑ Phospholipids & total cholesterol (↓ microsome, ↓ mitochondria, & ↑ cytosol);
↑ FFA & TG;

140 (1/10 LD50) Oral Single, tested after 2–8h M N/A ↑ at 2h and return to normal N/A N/A ↓ in adrenal (2 & 6 h) ↑ Corticosterone (2 & 6 h); (Joshi and Rajini, 2009)
↑ Liver glycogen (6 h)
↓ Gluconeogenesis (G6P & TAT) at 6 h;
= Weights of adrenals & liver

Chlorpyrifos 1 s.c. Daily for 4 d (PND1-4), test in adulthood F/M = = ↑ in post-prandial ♂, = in fasting ♂; ↑ in ♂ ↑ in ♂ = FFAs (Slotkin et al., 2005)
↓ in ♀ (p < 0.08)

5 s.c. Daily for 4 m F ↑ (starting at 2 m) N/A N/A N/A N/A ↑ Perinephric fat weight; (Meggs and Brewer, 2007)
= Weights of heart, liver & gastrocnemius muscle

1, 2.5, & 4 Oral Daily for 35 d, GND7-PND21 to dams F& pups ↑ in ♂ pups starting at PND 51 (2.5); N/A N/A N/A N/A ↑ Body volume in ♂ pups at PND 100; (Lassiter and Brimijoin, 2008)
↓ Weight/volume ratio in ♂ pups at PND 100;
= in ♀ pups ↓ Leptin

Diazinon 40 (1/3 LD50) i.p. Single, tested after 2 h F N/A N/A N/A N/A ↓ Brain glycogen; ↑ GP & PGM; = G6P; (Husain and Ansari, 1988)
Atropine (cholinergic blocker), tolazoline (α-adrenergic blocker) and propranolol (β-adrenergic blocker) abolished or reduced diazinon-induced hyperglycemia & brain glycogenolysis

40 i.p. Single, tested after 2 h F N/A ↑; N/A N/A N/A ↑ Lactic acid; (Matin et al., 1990a)
= Pyruvic acid;
Liver & brain:
↓ Glycogen;
↑ Glycogenolysis (↑ GP & PGM, = G6P);
abolished by adrenalectomy ↑ Glycolysis (↑ HK & LDH, brain only; = G6PD);
↑ Gluconeogenesis (↑ F1,6D & PEPCK, liver only);
Adrenalectomy abolished above changes

128 (LD50) -1 d; Oral 1 (single), 2, 8 or 32 d M N/A N/A N/A ↑ at 128; 10, 15 d post-treatment: ↓ Phospholipids (Ibrahim and El-Gamal, 2003)
↓ (64);
↓ (16);
64 – 2d; = other doses = (4);
16 – 8d; ↓ HDL;
8 – 32 d ↑ LDL (except ↓ at 16)

15, 30, & 60 Oral Single, tested after 2 h M N/A N/A N/A N/A Liver: (Teimouri et al., 2006)
↑ GP & PEPCK (30 & 60)

15, 30, & 60 (1/20, 1/10, 1/5 LD50) Oral Single, tested after 2 h M N/A N/A N/A ↑ TNF-α; all effect abolished by cAMP & cGMP PDE inhibitor (Ghafour-Rashidi et al., 2007)

75 (1/4 LD50) Oral Single, tested after 28 d M N/A N/A N/A N/A ↑ Testosterone (Alahyary et al., 2008)

6.5 (1/10 LD50) i.p. Single, (tested after 2 wk) Mg = = = ↓ (Wistar); ↓ Glucose tolerance in GK; (Ueyama et al., 2008)
= HDL = Glut4 in fat

15, 30, & 60 i.p. Single, tested after 1 & 18 h M N/A N/A ↓ (1h); N/A N/A ↑ Langerhans islet GDH (1h, 30 & 60; 18h); (Jamshidi et al., 2009)
↑ Glutamate (1h; = 18 h)
= (18h) ↑ C-peptide;
↓ GDH gene expression (18h, 60)

Dichlorvos 4 (1/20 LD50), 40 (1/2 LD50) Oral Single; 3, 7, 14 d M N/A ↑ (40) N/A N/A N/A ↓ Glucose tolerance; (Teichert-Kuliszewska and Szymczyk, 1979)
Liver:
↑ GP & GS (40); ↓ uridine diphosphate glucose pyrophosphrylase

6 s.c. Daily for 8 wk M N/A N/A N/A Brain: (Sarin and Gill, 1999)
↑ GP;
↓ Glycogen, HK, PFK, LDH & glucose uptake

20 (1/4 LD50) N/A Single, tested after 1 or 3 d M N/A N/A N/A N/A N/A Liver: (Romero-Navarro et al., 2006)
↓ Glycogen & GK activity;
↑ GK mRNA level;
Pancreas:
= GK activity & mRNA;
= Insulin mRNA

Dimethoate 150 i.p. 15 & 30 d (every other days) M = (15 d); = (15 d); (Reena et al., 1989)
↑ (30 d) ↑ (30 d)

20 & 40 (1/20 & 1/10 LD50) Oral Daily for 30 d M ↓ (40) N/A N/A N/A ↑ Lipase & amylase; (Kamath and Rajini, 2007)
↓ Pancreas lipase & amylase;
↓ Glucose tolerance;
↑ Pancreas weight;
= Weights of liver, kidney & adrenals

Isofenphos 20 Oral Single, tested after 3–72h M N/A N/A N/A N/A N/A ↑ Muscle lipid; (Calore et al., 1999)
↓ Sarcoplasmic esterase;
↓ Muscle lipase (13–18h)

Malathion 2000 s.c. Single, tested after 0–6 h F N/A N/A N/A N/A (Ramu and Drexler, 1973)

500 i.p. Single, tested after 0–48 h F N/A ↑ (first 6 h) N/A N/A N/A ↑ Liver, kidney, heart, & spleen glycogen (all 6–12 h, except liver 6–24 h); (Gupta, 1974)
= Brain glycogen

46 (1/25 LD50) i.p. 15 d M N/A = N/A N/A N/A ↑ Liver glycogen, Adrenaline (8 d), noradrenaline (8 d) & dopamine (4 d). (Gowda et al., 1983)

650 i.p. Single, tested after 8 h M N/A N/A N/A N/A (Rodrigues et al., 1986)

500 i.p. Single, tested after 2 h F N/A N/A N/A N/A ↑ Lactate; = pyruvate; (Matin and Husain, 1987)
Brain:
↓ Glycogen;
↑ GP, PGM & HK;
= G6P & G6PD

125, 250, & 500 i.p. Single, tested after 0.5,1, & 2 h F N/A N/A N/A N/A N/A Brain (500 except glycogen): (Matin et al., 1990b)
↓ Glycogen (starting at 0.5 h);
↑ GP, PGM, HK & lactate;
=G6P, G6PD, LDH & pyruvate;
Adrenalectomy abolish these above changes;
↓ Succinic dehydrogenase;
Adrenal:
↓ Ascorbic acid;
↓ Cholesterol

5,10, & 20 Oral (Diet) 4 wk M N/A N/A N/A N/A ↑ Liver PEPCK & GP (Abdollahi et al., 2004a)

5,10, & 20 Oral (Diet) 4 wk M N/A ↑ (10 & 20) ↑ (10 & 20) N/A N/A Muscle: (Pournourmohammadi et al., 2005)
↑ PFK & GP (20);
= HK

100 Oral Daily for 32 d M = = N/A N/A N/A ↓ Food intake; (Rezg et al., 2006)
Liver:
↑ Weight, HK, & glycogen;
↓ GP

100 Oral Daily for 32 d M N/A = N/A N/A N/A ↓ Liver lipids; ↑ glycogen; (Rezg et al., 2007)
↓ Muscle glycogen

20 Oral Daily for 28 d M N/A N/A N/A N/A Liver: (Basiri et al., 2007)
↑ PEPCK;
↑ Mitochondrial GP;
Administration of Satureja khuzestanica essential oil (225 mg/kg/day) abolished the malathion induced effect

5,10, & 20 Oral (Diet) 28 d M N/A ↑ (10 & 20) ↑ (10 & 20) N/A N/A ↓ Pancreas insulin secretion (glucose stimulated); (Pournourmohammadi et al., 2007)
= (KCl stimulated)

100 (1/21 LD50) Oral Single, tested after 24 h M N/A N/A N/A N/A N/A ↑ Liver glycogen & HK; (Rezg et al., 2008)
↓ Liver GP;
Caffeic acid (100 mg/kg) abolished these effects;

100 (1/21 LD50) Oral Daily for 32 d M N/A N/A N/A =; ↓ Hypothalamic CRH mRNA; (Rezg et al., 2010a)
= HDL & LDL ↑ iNOS; = nNOS

Parathion 0.1 & 0.2 s.c. Daily for 4 d (PND1-4) F & M ♂: ↑ (0.1); ↑ (0.2) in only normal diet = ↓ only ♀ in the fasted state ↑ in ♂ w/ HFD; ↓ Food intake in ♂ (0.2); (Lassiter et al., 2008)
♀: ↓ (0.1 & 0.2); ↑ Food intake in ♀ (0.1);
↓ HbA1c;
↓ in ♀ w/ HFD (0.2); ↓ FFA in ♀ w/ normal diet;
↓ in ♀ ↓ β-hydroxybutyrate only in fasted ♂

Carbamates

Carbendazim 0.48, 0.96, 2.4, & 4.8 s.c. Single, tested after 12 & 24h M N/A ↓ after 12h (0.48 & 0.96); ↑ after 12h (4.8); N/A N/A = after 12h; (Veerappan et al., 2012)
↑ after 24h ↑ after 24h

Pyrethroids

Cismethrin 4 i.v. Single M N/A N/A N/A N/A ↑ Noradrenaline, adrenaline, & lactate (Cremer and Seville, 1982)

Cypermethrin 420 (1/10 LD50) Oral (Diet) 6 m M N/A =; N/A N/A ↓; N/A (Shakoori et al., 1988)
↑ in liver; = in liver

0.06, 0.12, 0.3, & 0.6 s.c. Single, tested after 12 & 24h M N/A ↑ after 12h (0.3 & 0.6); ↑ after 24h (0.06) N/A N/A ↑ after 12h (0.06 & 0.12); ↑ after 24 h (0.3 & 0.6) (Veerappan et al., 2012)

Decamethrin 40 i.p. Single F N/A N/A N/A N/A ↑ Lactate (Ray and Cremer, 1979)

Deltamethrin 1.28 (1/100 LD50) Oral Daily for 30 d M N/A N/A ↑; ↑ Total lipid; (Yousef et al., 2006)
↑ LDL & VLDL; Vitamin E attenuate adverse effect of deltamethrin
↓ HDL

1.5 & 2.6 i.v. Single M N/A N/A N/A N/A ↑ Noradrenaline, adrenaline, & lactate (Cremer and Seville, 1982)

Neonicotinoids

Imidacloprid 0, 5, 10, & 20 Oral Daily for 90 d F ↓ (20) ↑ (20) N/A = = ↓ Food intake (20); (Bhardwaj et al., 2010)
↑ Weight of liver, kidney, & adrenal (20)

10, 30, & 90 Oral Daily from GND 6-PND 21 to dams; F& pups = N/A N/A N/A N/A (Gawade et al., 2013)
Daily from PND21-PND 42 to pups
a

mg/kg bw/day unless otherwise specified (e.g. in diet),

*

doses are ppm in diet; NOAEL, no observed adverse effect level;

b

s.c., subcutaneous injection; i.p., intraperitoneal injection; i.v., intravenous injection

c

d, day(s); h, hour(s); m, month(s); wk, week(s); w/o, without; PND, postnatal days; GND, gestational day;

d

F, female; M, male;

e

Results are for all doses tested in each study, unless doses are indicated as numbers in parenthesis; all markers are from fasting blood samples unless otherwise specified; ↑, increase; ↓, decrease; = no change; w/, with; ♂, male; ♀, female; N/A, not available;

f

apoE3 & C57BL/6N strain were used;

g

GK (Goto-Kakizaki) rats are a spontaneous animal model of non-insulin-dependent diabetes without obesity;

Acronyms used: ACC-1, Acetyl-CoA carboxylase 1; pACC, phosphorylated ACC; pAkt, phosphorylated Akt; AMPKα, AMP-activated protein kinase alpha; pAMPKα, phosphorylated AMPKα; BW, body weight; cAMP, cyclic adenosine monophosphate; CaMKKβ, calcium/calmodulin-dependent protein kinase kinase β; CD36, cluster of differentiation 36; CEBPα, CCAAT/enhancer-binding protein α; cGMP, cyclic guanosine monophosphate; CPT1, carnitine palmitoyltransferase I; CRH, corticotropin-releasing hormone; FA, fatty acid; FFAs, free fatty acids; FABP4, fatty acid binding protein 4; F1,6D, fructose 1,6-bisphosphatase; GDH, glutamate dehydrogenase; GK, glucokinase; GK rat, Goto-Kakizaki rat; Glut2, glucose transporter 2; Glut4, glucose transporter 4; G6P, glucose-6-phosphatase; G6PD, glucose-6-phosphatase dehydrogenase; GP, glycogen phosphorylase; GS, glycogen synthase; GTT, glucose tolerance test; HbA1c, glycated hemoglobin; HDL, high density lipoprotein; HFD, high-fat diet; HK, hexokinase; HOMA-IR, homeostatic model assessment-insulin resistance; iNOS, inducible nitric oxide synthase; IL-6, interleukin 6; LDH, lactate dehydrogenase; LFD, low-fat diet; LPL, lipoprotein lipase; nNOS, neuronal nitric oxide synthase; PDE, phosphodiesterase inhibitor; PDK4, pyruvate dehydrogenase lipoamide kinase isozyme 4; PEPCK, phosphoenolpyruvate carboxykinase; PFK, phosphofructokinase; PGC-1α, peroxisome proliferator-activated receptor gamma co-activator 1α; PGM, phosphoglucomutas; PK, pyruvate kinase; PPARγ, peroxisome proliferator-activated receptor gamma; SCD-1, stearoyl-CoA desaturase-1; Sirt 1, NAD-dependent deacetylase sirtuin-1; SREBP1, sterol regulatory element-binding protein 1; TAT, tyrosine aminotransferase; TG, triglycerides; TNFα, tumor necrosis factor-α; VLDL, very low-density lipoprotein.

4.3.2. Effects of insecticides on body weight in rodents

Although it is known that overstimulation of the nervous system increases the energy demands that are potentially linked to reduced weight, a number of studies reported that exposure to insecticides can lead to increased body weight gain, while other studies found inconsistent results (summarized in Table 4) (Matin et al., 1990a; Matin et al., 1990b; Pournourmohammadi et al., 2007; Rezg et al., 2008). Exposures to organochlorine insecticides (DDE, HCB, and ɣ-HCH) have led to increased body weight gain in rodents (Table 4) (Chadwick et al., 1988; Howell et al., 2014; Howell et al., 2015; Villeneuve et al., 1977), including a study of parental trans-generational exposure to DDT linked to increased obesity rate in the offspring (Skinner et al., 2013). Similarly, others reported that perinatal exposure to DDT was linked to significant weight gain, but only in female offspring (La Merrill et al., 2014) or reduced weight was reported after dieldrin exposure (Shakoori et al., 1984).

Organophosphorus insecticides have also been demonstrated to have moderate obesogenic effects. Chronic exposure to chlopyrifos under manifestation of cholinergic toxicity was reported to increase body weight in mice (Peris-Sampedro et al., 2015) and rats (Meggs and Brewer, 2007), including developmental exposure of chlorpyrifos on weight gain in male, but not female offspring (Lassiter and Brimijoin, 2008). Others, however, reported reduced or no changes in weight after exposure to chlorpyrofos, dichlorvos, or dimethoate (Kamath and Rajini, 2007; Sarin and Gill, 1999; Slotkin et al., 2005).

Limited data are available for effects of pyrethroids and neonicotinoids on weight gain. Exposure to cypermethrin (a pyrethroid) or imidacloprid (a neonicotinoid) significantly decreased body weight in mice or rats (Arfat; et al., 2014; Bhardwaj et al., 2010; Ince et al., 2012), while no effects of deltamethrin on weigh in mice was reported (Armstrong et al., 2013). Recently, our group reported that low doses of orally administrated permethrin or imidacloprid (levels of NOAEL and ADI) potentiate weight gain in male mice only when high-fat diet was provided (Sun et al., 2016b; Xiao et al., 2016). Taken collectively, the effects of insecticide exposure on body weight change are rather limited and inconsistent, likely depending on various factors, such as dose and route of administration, species, sex, and treatment duration. With new reports on low doses of insecticide and dietary fat interaction, it is possible that effects of insecticide exposure will be significant, particularly for relatively long-term low-dose exposure, when combined with other known contributing factors of obesity.

5. Mechanisms of insecticide-induced change in glucose and lipid metabolisms

Recent studies indicate that insecticides are reported to influence various organs and tissues, such as endocrine organs, liver, pancreas, muscle, and adipose tissue, which may lead to altered glucose and lipid metabolisms (Abdollahi et al., 2004c; Hectors et al., 2011; Hernandez et al., 2013; Karami-Mohajeri and Abdollahi, 2011; Kuo et al., 2013; McKinlay et al., 2008; Mostafalou and Abdollahi, 2013; Rahimi and Abdollahi, 2007; Rezg et al., 2010b; Soltaninejad and Abdollahi, 2009). A number of mechanisms were suggested to be influenced by insecticides; including oxidative stress and endoplasmic reticulum stress.

5.1. Liver

The liver, as one of the principal organs in regulation of glucose homeostasis, contributes to blood glucose level by maintaining a balance between storage of glucose via glycolysis and glycogenesis and release of glucose via glycogenolysis and gluconeogenesis (Abdollahi et al., 2004a; Hers, 1990; Nordlie et al., 1999; Rezg et al., 2006; Yang et al., 2000). It was suggested that since insecticides are designed to target the nervous system, exposure to insecticides may attribute to increased liver glucose production to meet the increased energy demand caused by overstimulation of the nervous system (Teimouri et al., 2006). In fact, studies have demonstrated that exposure to certain insecticides can increase the activities of key enzymes involved in hepatic gluconeogenesis and glycogenolysis (Abdollahi et al., 2004a; Basiri et al., 2007; Bergman, 1997; Deotare and Chakrabarti, 1981; Kauffman et al., 1990; Matin et al., 1990a; Romero-Navarro et al., 2006; Teimouri et al., 2006). Organophosphorus insecticides were shown to increase hepatic phosphoenolpyruvate carboxykinase (a key enzyme for gluconeogenesis) and glycogen phosphorylase (a key enzyme for glycogenolysis) in both in vitro and in vivo (Abdollahi et al., 2004a; Basiri et al., 2007; Kauffman et al., 1990; Teimouri et al., 2006); however, others reported that insecticides increased hepatic glycogen levels by organochlorine (dieldrin) and organophosphorus (malathion) (Bhatia and Venkitasubramanian, 1972; Gupta, 1974). The inconsistent results obtained from different studies may be explained by initially increased blood glucose level caused by gluconeogenesis and glycogenolysis (the maximum increase was noted at 2 h after administration), followed by glycogenesis, and a return to normal blood glucose level 6–24 h after malathion treatment (Rezg et al., 2006; Rezg et al., 2007).

5.2. Pancreas

Excessive stimulation of the cholinergic receptors by insecticides can result in disturbance of insulin and glucagon secretions, potentially due to pancreas tissue damage. Thus, insecticides that influence either acetylcholine level or its receptors by inhibiting acetylcholine esterase (organophosphorus and carbamates) or by acting as agonists to nicotinic acetylcholine receptors (neonicotinoids) can all potentially stimulate insulin release from the pancreas. Studies have found that exposure to malathion, an organophosphorus insecticide, resulted in elevated blood insulin levels in rats (Pournourmohammadi et al., 2005; Pournourmohammadi et al., 2007). Another study, however, found that exposure to malathion resulted in decreased glucose-stimulated insulin secretion accompanied with patchy degenerative changes in the islets of Langerhans (Pournourmohammadi et al., 2007). Similarly, other human and animal studies also found that organophosphorus and carbamate insecticide can cause acute pancreatitis, which potentially influence insulin secretion (Dressel et al., 1978; Gokel et al., 2002; Goodale et al., 1993; Harputluoglu et al., 2003; Hsiao et al., 1996; Kandalaft et al., 1991; Lankisch et al., 1990; Marsh et al., 1988; Moore and James, 1981; Moritz et al., 1994; Panieri et al., 1997; Weizman and Sofer, 1992).

5.3. Muscle

Muscle is tightly linked with the nervous system by neuromuscular junctions, thus susceptible to insecticide-induced neurotoxic effects. Previous reports have exhibited that exposure to malathion, an organophosphorus insecticide, decreased glycogen content in muscles (Pournourmohammadi et al., 2005; Rezg et al., 2007). In particular, Pournourmohammadi et al. (Pournourmohammadi et al., 2005) reported that this was due to increased activities of glycogen phosphorylase and phosphofructokinase (PFK), which are the key enzymes regulating glycogenolysis and glycolysis, respectively. In addition, permethrin (a pyrethroid insecticide) and imidacloprid (a neonicotinoid insecticide) were previously shown to induce insulin resistance in C2C12 muscle cells via Akt signaling (Kim et al., 2013, 2014a).

5.4. Adipose tissue

It is suggested that many lipophilic insecticides, such as DDE, malathion, or permethrin, can be easily trapped and stored in adipose tissues (Karmaus et al., 2009; Morgan et al., 2007; Pournourmohammadi et al., 2007). In addition, in vitro studies using 3T3-L1 adipocytes demonstrated that DDT (Kim et al., 2016; Moreno-Aliaga and Matsumura, 2002), DDE (Kim et al., 2016; Mangum et al., 2015), imidacloprid (Park et al., 2013), and permethrin (Kim et al., 2014a) potentiated adipogenesis. Another study found that treatment of DDE to 3T3-L1 cells did not alter adipogenesis or lipolysis, but increased basal free fatty acid uptake and the release of leptin, resistin, and adiponectin, which may be potentially linked to increased risk of obesity and type 2 diabetes (Howell and Mangum, 2011). The same study also reported that oxychlordane and dieldrin (both organochlorines) increased basal free fatty acid uptake, but not insulin-stimulated glucose uptake in 3T3-L1 adipocytes (Howell and Mangum, 2011).

5.5. Endocrine organs and brain

Currently more than 101 pesticides have been listed as proven or possible endocrine disruptors by the Pesticide Action Network UK (Mostafalou and Abdollahi, 2013). Some of the insecticides disrupt the endocrine system by mimicking the action of estrogen. For example, certain organochlorines and carbamates were reported to inhibit androgen receptors; some organophosphorus insecticides were reported to increase the expression of estrogen responsive genes; and certain pyrethroids potentiate the action of estrogen (McKinlay et al., 2008). The endocrine disrupting activities of pesticides may potentiate the risk of obesity and type 2 diabetes and other related diseases (Alonso-Magdalena et al., 2011; Gore et al., 2015).

Along with altered glucose homeostasis, organophosphorus compounds have been demonstrated to elevate catecholamine levels (Abdollahi et al., 2003a; Abdollahi et al., 2003b; Fukuyama and Adie, 1963; Gowda et al., 1983; Matin et al., 1990b; Pournourmohammadi et al., 2005; Ramu et al., 1973; Sungur and Guven, 2001), which are abolished by adrenalectomy (Matin et al., 1990a; Matin et al., 1990b). This suggests a role of adrenal glands in organophosphorus-induced disturbance of glucose homeostasis.

Pyrethroids were reported to inhibit progesterone action, organophosphorus insecticides were shown to inhibit thyroid hormone receptor (McKinlay et al., 2008), and exposure to an organophosphorus, diazinon, was previously reported to increase the serum testosterone levels (Alahyary et al., 2008). Organochlorine and carbamate insecticides showed anti-androgenic effects by inhibition of binding natural ligand to androgen binding receptors (Mostafalou and Abdollahi, 2013). All these hormones are known to influence insulin sensitivity and glucose homeostasis (Bruns and Kemnitz, 2004; Fernandez-Real et al., 2006; Polderman et al., 1994). It was suggested that insecticides were able to act as an agonist or an antagonist towards aryl hydrocarbon receptors and certain nuclear receptors, such as retinoic acid receptors, pregame X receptors, and peroxisome proliferator-activated receptors (Kojima et al., 2010; Lemaire et al., 2005).

Studies have reported decreased brain glycogen content with increased activity of glycogen phosphorylase by organophosphorus insecticide, malathion (Matin and Husain, 1987; Matin et al., 1990a) and dichlorvos (Sarin and Gill, 1999). It was suggested that malathion could interfere with oxygen uptake and promoted glycogenolysis and glycolysis in favor of anaerobic conditions to counteract the neurotoxic effects in rats (Matin and Husain, 1987). Malathion was also shown to increase lactic acid concentration without altering pyruvate content in the brain (Matin et al., 1990b). Others reported that an organochlorine insecticide (dieldrin) was previously reported to cause apoptosis and neural degeneration by increasing acetylation of core histone H3 and H4 in neuronal cells (Song et al., 2010). Limited studies reported that exposure to insecticides could affect feeding behavior by acting on the neural circuits (Mostafalou and Abdollahi, 2013). When low doses of imidacloprid or permethrin (at or lower than NOAEL) were administered in mice, no significant effects of either insecticide on food intake were observed (Sun et al., 2016b; Xiao et al., 2015; Xiao et al., 2016).

5.6. Cellular Responses

5.6.1. Oxidative stress

Oxidative stress was often suggested to be associated with insecticide-induced toxicity in vitro and in vivo (Abdollahi et al., 2004b; Bagchi et al., 1995; Begum and Rajini, 2011; Kalender et al., 2005; Mostafalou and Abdollahi, 2012; Mostafalou and Abdollahi, 2013; Mostafalou et al., 2012; Slaninova et al., 2009; Soltaninejad and Abdollahi, 2009; Teimouri et al., 2006; Yang and Dettbarn, 1996). It is also known that oxidative stress is linked with obesity and type 2 diabetes (Furukawa et al., 2004). Thus, many mechanistic studies have suggested that insecticides disrupt glucose and lipid metabolisms via oxidative stress-mediated mechanisms, such as lipid peroxidation, mitochondrial dysfunction, inhibition of paraoxonase and glucose-6-phosphate dehydrogenase (G6PD), and nitrosative stress (Abdollahi et al., 2004b; Abdollahi et al., 2004c; Akhgari et al., 2003; Karami-Mohajeri and Abdollahi, 2011; Matsuoka et al., 1997; Mostafalou and Abdollahi, 2013; Ranjbar et al., 2002; Rezg et al., 2008; Shadnia et al., 2005; Soltaninejad and Abdollahi, 2009).

Glucose plays a critical role as an antioxidant against free radicals via the pentose phosphate pathway, in which nicotinamide adenine dinucleotide phosphate (NADP) is reduced to NADPH, which is subsequently used for the reduction of oxidized glutathione (GSH) from disulfides of GSH and cellular proteins (Teimouri et al., 2006). The increased demand of glucose to counteract the increased reactive oxygen species induced by insecticides has been suggested to cause hyperglycemia, leading to stimulated hepatic glycogenolysis and gluconeogenesis pathways (Teimouri et al., 2006). In fact, some studies found that antioxidants could attenuate insecticide-induced hyperglycemia (Basiri et al., 2007; Yousef et al., 2006).

Abnormal mitochondrial respiratory chain functions can cause disturbance in intracellular energy homeostasis and has been involved in many diseases, including diabetes (Abdul-Ghani and DeFronzo, 2008; Kim et al., 2008; Lowell and Shulman, 2005; Ma et al., 2012). Exposure to insecticides can cause muscle fasciculation, which greatly increase the oxygen flow into corresponding tissues and organs (Abdollahi et al., 2004b; Abdollahi et al., 2004c; Basiri et al., 2007; Mostafalou and Abdollahi, 2012). And this increased oxygen flow caused by insecticides may lead to elevated oxidative phosphorylation in mitochondria, which subsequently increase the production of reactive oxygen species as byproducts. In addition, some insecticides (rotenone or pyridaben) are known to disrupt mitochondrial respiratory chain reaction, mainly by inhibiting Complex I, II, III and V electron transport chain (Gomez et al., 2007).

Studies have demonstrated that pancreatic beta cell failure induced by insecticide exposure could be the result of underlying mitochondrial dysfunction in the pancreas (Lee, 2011; Lim et al., 2009; Mostafalou and Abdollahi, 2013). It is reported that the pancreas is more susceptible to reactive oxygen species than other tissues because of its relative low expression of defensive enzymes against reactive oxygen species (Grankvist et al., 1981; Ho and Bray, 1999; Kakkar et al., 1998).

Besides their inhibiting effect on acetylcholine esterase activity, organophosphorus compounds were reported to inhibit other esterase, such as the paraoxonase, a key enzyme involved in hydrolysis of oxons (active organophosphorus metabolites), which may potentially increase oxidative stress (Mackness et al., 1991).

cAMP and cGMP signaling may also play an important role in insecticide-induced oxidative stress. Previous studies have found that increased cyclic nucleotides using phosphodiesterase inhibitors could protect against organophosphorus-induced lipid peroxidation in rat submandibular saliva (Abdollahi et al., 2003a) and liver cells (Abdollahi et al., 2003b). Similarly, another report supported that increased intracellular levels of cAMP and cGMP by intraperitoneal administration of phosphodiesterase inhibitors could exert protective effects against organophosphorus-induced hyperglycemia and oxidative/nitrosative stress in Langerhans islets cells in rat (Ghafour-Rashidi et al., 2007).

5.6.2. Endoplasmic reticulum (ER) stress

Recent studies have found ER stress play key roles in development of several chronic diseases, including obesity and type 2 diabetes (Cao and Kaufman, 2013; Hotamisligil, 2010; Hummasti and Hotamisligil, 2010; Ozcan et al., 2004; van der Kallen et al., 2009). There are a few studies reporting a correlation between several types of insecticides and ER stress. An organochlorine (endosulfan), carbamates (formetanate, methomyl, pyrimicarb), and a pyrethroid (bifenthrin) were reported to increase 78 kDa glucose-regulated protein GRP78, also known as binding immunoglobulin protein (BiP), which is one of the ER stress markers in human pulmonary A549 cells (Skandrani et al., 2006a; Skandrani et al., 2006b). Another pyrethroids insecticide, deltamethrin, was reported to induce ER stress in SK-N-AS neuroblastoma cells via elevation of intracellular calcium level and activation of eukaryotic translation initiation factor 2 α (eIF2α) (Hossain and Richardson, 2011). Currently there is no study directly determining the role of ER stress caused by insecticides and development of obesity and type 2 diabetes.

6. Conclusion

Previous studies have largely focused on organochlorine and organophosphorus insecticides, with less on carbamates, pyrethroids, and neonicotinoids and the link between development of obesity and type 2 diabetes. The human and animal studies included in this review generally suggest that exposure to insecticides increases the risk of developing obesity and type 2 diabetes with some inconsistent results. This inconsistency may have contributed from other factors, such as dose, duration of exposure, methods used to determine variables, diet, and experimental design especially for animal studies. Although limited, current knowledge suggest that there is great need to determine the effects of low level exposure to insecticides, at or lower than NOAEL, and interaction with other known factors, such as sex and diet, that contributing in development of obesity and type 2 diabetes. In addition, knowledge on specifics of various exposures of new types of insecticides is still very limited, and there is a great need to develop proper strategy for evaluating the exposure levels for insecticides, particularly for those known to be metabolized and eliminated rather quickly in the biological system. Lastly, although we have not focused on the developmental exposure of insecticides in the current review, it would be significant to understand developmental exposure of insecticides and health implication of the next generation. Overall, we conclude that the majority of studies reporting exposures to insecticides are associated with increased risk of developing obesity and type 2 diabetes. More clinical and epidemiological studies, as well as mechanistic studies are needed to understand the insecticide-induced disturbances in glucose and lipid metabolisms, which may contribute to the development of obesity and type 2 diabetes.

Supplementary Material

1
2
3

Highlights.

  • Exposure to insecticides are positively associated with the risk of obesity and type 2 diabetes.

  • Exposure to insecticides may disturb glucose and lipid metabolisms.

  • Insecticide may induce oxidative and/or endoplasmic reticulum stresses.

Acknowledgments

Authors would like to thank Ms. Brooke T. Pearson and Jayne M. Storkson for help in writing manuscript. This project is supported in part by NIH funding R21ES023676.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abdollahi M, Bahreini-Moghadam A, Emami B, Fooladian F, Zafari K. Increasing intracellular cAMP and cGMP inhibits cadmium-induced oxidative stress in rat submandibular saliva. Comp Biochem Physiol C Toxicol Pharmacol. 2003a;135C(3):331–336. doi: 10.1016/s1532-0456(03)00120-0. [DOI] [PubMed] [Google Scholar]
  2. Abdollahi M, Chan TS, Subrahmanyam V, O'Brien PJ. Effects of phosphodiesterase 3,4,5 inhibitors on hepatocyte cAMP levels, glycogenolysis, gluconeogenesis and susceptibility to a mitochondrial toxin. Mol Cell Biochem. 2003b;252(1–2):205–211. doi: 10.1023/a:1025568714217. [DOI] [PubMed] [Google Scholar]
  3. Abdollahi M, Donyavi M, Pournourmohammadi S, Saadat M. Hyperglycemia associated with increased hepatic glycogen phosphorylase and phosphoenolpyruvate carboxykinase in rats following subchronic exposure to malathion. Comp Biochem Physiol C Toxicol Pharmacol. 2004a;137(4):343–347. doi: 10.1016/j.cca.2004.03.009. [DOI] [PubMed] [Google Scholar]
  4. Abdollahi M, Mostafalou S, Pournourmohammadi S, Shadnia S. Oxidative stress and cholinesterase inhibition in saliva and plasma of rats following subchronic exposure to malathion. Comp Biochem Physiol C Toxicol Pharmacol. 2004b;137(1):29–34. doi: 10.1016/j.cca.2003.11.002. [DOI] [PubMed] [Google Scholar]
  5. Abdollahi M, Ranjbar A, Shadnia S, Nikfar S, Rezaie A. Pesticides and oxidative stress: a review. Med Sci Monit. 2004c;10(6):RA141–147. [PubMed] [Google Scholar]
  6. Abdul-Ghani MA, DeFronzo RA. Mitochondrial dysfunction, insulin resistance, and type 2 diabetes mellitus. Curr Diab Rep. 2008;8(3):173–178. doi: 10.1007/s11892-008-0030-1. [DOI] [PubMed] [Google Scholar]
  7. Abou-Donia MB. Organophosphorus ester-induced chronic neurotoxicity. Arch Environ Health. 2003;58(8):484–497. doi: 10.3200/AEOH.58.8.484-497. [DOI] [PubMed] [Google Scholar]
  8. ADA. The Cost of Diabetes. American Diabetes Association; 2013. http://www.diabetes.org/advocacy/news-events/cost-of-diabetes.html?referrer= https://www.google.com/ [Google Scholar]
  9. Aguirre GA, De Ita JR, de la Garza RG, Castilla-Cortazar I. Insulin-like growth factor-1 deficiency and metabolic syndrome. J Transl Med. 2016;14:3. doi: 10.1186/s12967-015-0762-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Airaksinen R, Rantakokko P, Eriksson JG, Blomstedt P, Kajantie E, Kiviranta H. Association between type 2 diabetes and exposure to persistent organic pollutants. Diabetes Care. 2011;34(9):1972–1979. doi: 10.2337/dc10-2303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Akhgari M, Abdollahi M, Kebryaeezadeh A, Hosseini R, Sabzevari O. Biochemical evidence for free radical-induced lipid peroxidation as a mechanism for subchronic toxicity of malathion in blood and liver of rats. Hum Exp Toxicol. 2003;22(4):205–211. doi: 10.1191/0960327103ht346oa. [DOI] [PubMed] [Google Scholar]
  12. Al-Attar AM, Abu Zeid IM. Effect of tea (Camellia sinensis) and olive (Olea europaea L.) leaves extracts on male mice exposed to diazinon. Biomed Res Int. 2013 doi: 10.1155/2013/461415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Al-Othman A, Yakout S, Abd-Alrahman SH, Al-Daghri NM. Strong associations between the pesticide hexachlorocyclohexane and type 2 diabetes in Saudi Adults. Int J Env Res Pub He. 2014;11(9):8984–8995. doi: 10.3390/ijerph110908984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Al-Othman AA, Abd-Alrahman SH, Al-Daghri NM. DDT and its metabolites are linked to increased risk of type 2 diabetes among Saudi adults: a cross-sectional study. Environ Sci Pollut R. 2015;22(1):379–386. doi: 10.1007/s11356-014-3371-0. [DOI] [PubMed] [Google Scholar]
  15. Alahyary P, Poor MI, Azarbaijani FF, Nejati V. The potential toxicity of diazinon on physiological factors in male rat. Pak J Biol Sci. 2008;11(1):127–130. doi: 10.3923/pjbs.2008.127.130. [DOI] [PubMed] [Google Scholar]
  16. Alonso-Magdalena P, Quesada I, Nadal A. Endocrine disruptors in the etiology of type 2 diabetes mellitus. Nat Rev Endocrinol. 2011;7(6):346–353. doi: 10.1038/nrendo.2011.56. [DOI] [PubMed] [Google Scholar]
  17. Amanvermez R, Baydin A, Yardan T, Basol N, Gunay M. Emergency laboratory abnormalities in suicidal patients with acute organophosphate poisoning. Turk J Biochem. 2010;35(1):29–34. [Google Scholar]
  18. Arfat Y, Mahmood N, Tahir MU, Rashid M, Anjum S, Zhao F, Li D-J, Sun Y-L, Hu L, Zhihao C, Yin C, Shang P, Qian A-R. Effect of imidacloprid on hepatotoxicity and nephrotoxicity in male albino mice. Toxicol Rep. 2014;1:554–561. doi: 10.1016/j.toxrep.2014.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Armstrong LE, Driscoll MV, Donepudi AC, Xu J, Baker A, Aleksunes LM, Richardson JR, Slitt AL. Effects of developmental deltamethrin exposure on white adipose tissue gene expression. J Biochem Mol Toxicol. 2013;27(2):165–171. doi: 10.1002/jbt.21477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Arrebola JP, Pumarega J, Gasull M, Fernandez MF, Martin-Olmedo P, Molina-Molina JM, Fernandez-Rodriguez M, Porta M, Olea N. Adipose tissue concentrations of persistent organic pollutants and prevalence of type 2 diabetes in adults from Southern Spain. Environ Res. 2013;122:31–37. doi: 10.1016/j.envres.2012.12.001. [DOI] [PubMed] [Google Scholar]
  21. Avsarogullari L, Ikizceli I, Sungur M, Sozuer E, Akdur O, Yucei M. Acute amitraz poisoning in adults: clinical features, laboratory findings, and management. Clin Toxicol (Phila) 2006;44(1):19–23. doi: 10.1080/15563650500357545. [DOI] [PubMed] [Google Scholar]
  22. Azandjeme CS, Delisle H, Fayomi B, Ayotte P, Djrolo F, Houinato D, Bouchard M. High serum organochlorine pesticide concentrations in diabetics of a cotton producing area of the Benin Republic (West Africa) Environ Int. 2014;69:1–8. doi: 10.1016/j.envint.2014.04.002. [DOI] [PubMed] [Google Scholar]
  23. Badrane N, Askour M, Berechid K, Abidi K, Dendane T, Zeggwagh AA. Severe oral and intravenous insecticide mixture poisoning with diabetic ketoacidosis: a case report. BMC Res Notes. 2014;7:485. doi: 10.1186/1756-0500-7-485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bagchi D, Bagchi M, Hassoun EA, Stohs SJ. In vitro and in vivo generation of reactive oxygen species, DNA damage and lactate dehydrogenase leakage by selected pesticides. Toxicology. 1995;104(1–3):129–140. doi: 10.1016/0300-483x(95)03156-a. [DOI] [PubMed] [Google Scholar]
  25. Basiri S, Esmaily H, Vosough-Ghanbari S, Mohammadirad A, Yasa N, Abdollahi M. Improvement by Satureja khuzestanica essential oil of malathion-induced red blood cells acetylcholinesterase inhibition and altered hepatic mitochondrial glycogen phosphorylase and phosphoenolpyruvate carboxykinase activities. Pestic Biochem Physiol. 2007;89(2):124–129. [Google Scholar]
  26. Begum K, Rajini PS. Augmentation of hepatic and renal oxidative stress and disrupted glucose homeostasis by monocrotophos in streptozotocin-induced diabetic rats. Chem-Biol Interact. 2011;193(3):240–245. doi: 10.1016/j.cbi.2011.07.003. [DOI] [PubMed] [Google Scholar]
  27. Berdanier CD, de Dennis SK. Effect of exercise on the responses of rats to DDT. J Toxicol Environ Health. 1977;2(3):651–656. doi: 10.1080/15287397709529466. [DOI] [PubMed] [Google Scholar]
  28. Bergman RN. New concepts in extracellular signaling for insulin action: the single gateway hypothesis. Recent Prog Horm Res. 1997;52:359–385. discussion 385-357. [PubMed] [Google Scholar]
  29. Bhardwaj S, Srivastava MK, Kapoor U, Srivastava LP. A 90 days oral toxicity of imidacloprid in female rats: morphological, biochemical and histopathological evaluations. Food Chem Toxicol. 2010;48(5):1185–1190. doi: 10.1016/j.fct.2010.02.009. [DOI] [PubMed] [Google Scholar]
  30. Bhatia SC, Venkitasubramanian TA. Mechanism of dieldrin-induced fat accumulation in rat liver. J Agric Food Chem. 1972;20(5):993–996. doi: 10.1021/jf60183a040. [DOI] [PubMed] [Google Scholar]
  31. Boada LD, Lara PC, Alvarez-Len EE, Losada A, Zurnbado ML, Limihana-Canhal JM, Apolinario R, Serra-Majem L, Luzardo OP. Serum levels of insulin-like growth factor-I in relation to organochlorine pesticides exposure. Growth Horm IGF Res. 2007;17(6):506–511. doi: 10.1016/j.ghir.2007.05.004. [DOI] [PubMed] [Google Scholar]
  32. Bruns CM, Kemnitz JW. Sex hormones, insulin sensitivity, and diabetes mellitus. ILAR J. 2004;45(2):160–169. doi: 10.1093/ilar.45.2.160. [DOI] [PubMed] [Google Scholar]
  33. Burns JS, Williams PL, Korrick SA, Hauser R, Sergeyev O, Revich B, Lam T, Lee MM. Association between chlorinated pesticides in the serum of prepubertal Russian boys and longitudinal biomarkers of metabolic function. Am J Epidemiol. 2014;180(9):909–919. doi: 10.1093/aje/kwu212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Calore EE, Sesso A, Puga FR, Cavaliere MJ, Calore NM, Weg R. Sarcoplasmic lipase and non-specific esterase inhibition in myofibers of rats intoxicated with the organophosphate isofenphos. Exp Toxicol Pathol. 1999;51(1):27–33. doi: 10.1016/S0940-2993(99)80054-2. [DOI] [PubMed] [Google Scholar]
  35. Cao SS, Kaufman RJ. Targeting endoplasmic reticulum stress in metabolic disease. Expert Opin Ther Targets. 2013;17(4):437–448. doi: 10.1517/14728222.2013.756471. [DOI] [PubMed] [Google Scholar]
  36. Casida JE. Neonicotinoid metabolism: compounds, substituents, pathways, enzymes, organisms, and relevance. J Agric Food Chem. 2011;59(7):2923–2931. doi: 10.1021/jf102438c. [DOI] [PubMed] [Google Scholar]
  37. Casida JE, Durkin KA. Neuroactive insecticides: targets, selectivity, resistance, and secondary effects. Annu Rev Entomol. 2013;58:99–117. doi: 10.1146/annurev-ento-120811-153645. [DOI] [PubMed] [Google Scholar]
  38. Casida JE, Quistad GB. Organophosphate toxicology: safety aspects of nonacetylcholinesterase secondary targets. Chem Res Toxicol. 2004;17(8):983–998. doi: 10.1021/tx0499259. [DOI] [PubMed] [Google Scholar]
  39. CDC. National diabetes statistics report. Centers for Disease Control and Prevention; 2014. https://www.cdc.gov/diabetes/data/statistics/2014statisticsreport.html. [Google Scholar]
  40. Chadwick RW, Cooper RL, Chang J, Rehnberg GL, McElroy WK. Possible antiestrogenic activity of lindane in female rats. J Biochem Toxicol. 1988;3:147–158. doi: 10.1002/jbt.2570030303. [DOI] [PubMed] [Google Scholar]
  41. Choudhari PD, Chakrabarti CH. Effect of acephate (orthene), an organophosphorus insecticide, on lipid metabolism in albino rats. Indian J Exp Biol. 1984;22(1):45–49. [PubMed] [Google Scholar]
  42. Coats JR. Mechanisms of toxic action and structure-activity relationships for organochlorine and synthetic pyrethroid insecticides. Environ Health Perspect. 1990;87:255–262. doi: 10.1289/ehp.9087255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Codru N, Schymura MJ, Negoita S, Akwesasne Task Force on, E. Rej R, Carpenter DO. Diabetes in relation to serum levels of polychlorinated biphenyls and chlorinated pesticides in adult Native Americans. Environ Health Perspect. 2007;115(10):1442–1447. doi: 10.1289/ehp.10315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Cox S, Niskar AS, Narayan KMV, Marcus M. Prevalence of self-reported diabetes and exposure to organochlorine pesticides among Mexican Americans: Hispanic Health and Nutrition Examination Survey, 1982–1984. Environ Health Perspect. 2007;115(12):1747–1752. doi: 10.1289/ehp.10258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Cremer JE, Seville MP. Comparative effects of two pyrethroids, deltamethrin and cismethrin, on plasma catecholamines and on blood glucose and lactate. Toxicol Appl Pharmacol. 1982;66(1):124–133. doi: 10.1016/0041-008x(82)90067-9. [DOI] [PubMed] [Google Scholar]
  46. Davies TG, Field LM, Usherwood PN, Williamson MS. DDT, pyrethrins, pyrethroids and insect sodium channels. IUBMB Life. 2007;59(3):151–162. doi: 10.1080/15216540701352042. [DOI] [PubMed] [Google Scholar]
  47. Debost-Legrand A, Warembourg C, Massart C, Chevrier C, Bonvallot N, Monfort C, Rouget F, Bonnet F, Cordier S. Prenatal exposure to persistent organic pollutants and organophosphate pesticides, and markers of glucose metabolism at birth. Environ Res. 2016;146:207–217. doi: 10.1016/j.envres.2016.01.005. [DOI] [PubMed] [Google Scholar]
  48. Deotare ST, Chakrabarti CH. Effect of acephate (orthene) on tissue levels of thiamine, pyruvic acid, lactic acid, glycogen and blood sugar. Indian J Physiol Pharmacol. 1981;25(3):259–264. [PubMed] [Google Scholar]
  49. Dirinck E, Jorens PG, Covaci A, Geens T, Roosens L, Neels H, Mertens I, Van Gaal L. Obesity and persistent organic pollutants: possible obesogenic effect of organochlorine pesticides and polychlorinated biphenyls. Obesity (Silver Spring) 2011;19(4):709–714. doi: 10.1038/oby.2010.133. [DOI] [PubMed] [Google Scholar]
  50. Dirinck EL, Dirtu AC, Govindan M, Covaci A, Van Gaal LF, Jorens PG. Exposure to persistent organic pollutants: relationship with abnormal glucose metabolism and visceral adiposity. Diabetes Care. 2014;37(7):1951–1958. doi: 10.2337/dc13-2329. [DOI] [PubMed] [Google Scholar]
  51. Dressel TD, Goodale RL, Arneson MA, Borner JW. Pancreatitis as a complication of anticholinesterase insecticide intoxication. Ann Surg. 1978;189(2):199–204. doi: 10.1097/00000658-197902000-00011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Everett CJ, Frithsen IL, Diaz VA, Koopman RJ, Simpson WM, Mainous AG. Association of a polychlorinated dibenzo-p-dioxin, a polychlorinated biphenyl, and DDT with diabetes in the 1999–2002 National Health and Nutrition Examination Survey. Environ Res. 2007;103(3):413–418. doi: 10.1016/j.envres.2006.11.002. [DOI] [PubMed] [Google Scholar]
  53. Everett CJ, Matheson EM. Biomarkers of pesticide exposure and diabetes in the 1999–2004 national health and nutrition examination survey. Environ Int. 2010;36(4):398–401. doi: 10.1016/j.envint.2010.02.010. [DOI] [PubMed] [Google Scholar]
  54. Faerch K, Hojlund K, Vind BF, Vaag A, Dalgard C, Nielsen F, Grandjean P. Increased serum concentrations of persistent organic pollutants among prediabetic individuals: potential role of altered substrate oxidation patterns. J Clin Endocr Metab. 2012;97(9):E1705–E1713. doi: 10.1210/jc.2012-1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Fernandez-Real JM, Lopez-Bermejo A, Castro A, Casamitjana R, Ricart W. Thyroid function is intrinsically linked to insulin sensitivity and endothelium-dependent vasodilation in healthy euthyroid subjects. J Clin Endocr Metab. 2006;91(9):3337–3343. doi: 10.1210/jc.2006-0841. [DOI] [PubMed] [Google Scholar]
  56. Fukuyama GS, Adie PA. Blood levels of adrenaline and noradrenaline during anticholinesterase poisoning. Arch Int Pharmacodyn Ther. 1963;146:56–64. [PubMed] [Google Scholar]
  57. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114(12):1752–1761. doi: 10.1172/JCI21625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Gasull M, Pumarega J, Tellez-Plaza M, Castell C, Tresserras R, Lee DH, Porta M. Blood concentrations of persistent organic pollutants and prediabetes and diabetes in the general population of Catalonia. Environ Sci Technol. 2012;46(14):7799–7810. doi: 10.1021/es300712g. [DOI] [PubMed] [Google Scholar]
  59. Gawade L, Dadarkar SS, Husain R, Gatne M. A detailed study of developmental immunotoxicity of imidacloprid in Wistar rats. Food Chem Toxicol. 2013;51:61–70. doi: 10.1016/j.fct.2012.09.009. [DOI] [PubMed] [Google Scholar]
  60. Gervais JA, Luukinen B, Buhl K, Stone D. "Imidacloprid technical fact sheet" (PDF) National Pesticide Information Center 2012 [Google Scholar]
  61. Ghafour-Rashidi Z, Dermenaki-Farahani E, Aliahmadi A, Esmaily H, Mohammadirad A, Ostad SN, Abdollahi M. Protection by cAMP and cGMP phosphodiesterase inhibitors of diazinon-induced hyperglycemia and oxidative/nitrosative stress in rat Langerhans islets cells: Molecular evidence for involvement of non-cholinergic mechanisms. Pestic Biochem Physiol. 2007;87(3):261–270. [Google Scholar]
  62. Glynn AW, Granath F, Aune M, Atuma S, Darnerud PO, Bjerselius R, Vainio H, Weiderpass E. Organochlorines in Swedish women: determinants of serum concentrations. Environ Health Perspect. 2003;111(3):349–355. doi: 10.1289/ehp.5456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Gokel Y, Gulalp B, Acikalin A. Parotitis due to organophosphate intoxication. J Toxicol Clin Toxicol. 2002;40(5):563–565. doi: 10.1081/clt-120014648. [DOI] [PubMed] [Google Scholar]
  64. Gomez C, Bandez MJ, Navarro A. Pesticides and impairment of mitochondrial function in relation with the parkinsonian syndrome. Front Biosci. 2007;12:1079–1093. doi: 10.2741/2128. [DOI] [PubMed] [Google Scholar]
  65. Goodale RL, Manivel JC, Borner JW, Liu S, Judge J, Li C, Tanaka T. Organophosphate sensitizes the human pancreas to acinar cell injury - an ultrastructural-study. Pancreas. 1993;8(2):171–175. doi: 10.1097/00006676-199303000-00006. [DOI] [PubMed] [Google Scholar]
  66. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J, Zoeller RT. Executive summary to EDC-2: The endocrine society's second scientific statement on endocrine-disrupting chemicals. Endocr Rev. 2015;36(6):593–602. doi: 10.1210/er.2015-1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Gowda H, Uppal RP, Garg BD. Effect of malathion on adrenal activity, liver glycogen & blood glucose in rats. Indian J Med Res. 1983;78:847–851. [PubMed] [Google Scholar]
  68. Grankvist K, Marklund SL, Taljedal IB. CuZn-superoxide dismutase, Mn-superoxide dismutase, catalase and glutathione peroxidase in pancreatic islets and other tissues in the mouse. Biochem J. 1981;199(2):393–398. doi: 10.1042/bj1990393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Gupta M, Mukherjee S, Gupta SD, Dolui AK, Dey SN, Roy DK. Changes of lipid spectrum in different tissues of Furadan-treated mice. Toxicology. 1986;38(1):69–79. doi: 10.1016/0300-483x(86)90173-3. [DOI] [PubMed] [Google Scholar]
  70. Gupta PK. Malathion induced biochemical changes in rat. Acta Pharmacol Toxicol (Copenh) 1974;35(3):191–194. doi: 10.1111/j.1600-0773.1974.tb00738.x. [DOI] [PubMed] [Google Scholar]
  71. Hansen MR, Jors E, Lander F, Condarco G, Schlunssen V. Is cumulated pyrethroid exposure associated with prediabetes? A cross-sectional study. J Agromedicine. 2014;19(4):417–426. doi: 10.1080/1059924X.2014.945708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Harputluoglu MM, Kantarceken B, Karincaoglu M, Aladag M, Yildiz R, Ates M, Yildirim B, Hilmioglu F. Acute pancreatitis: an obscure complication of organophosphate intoxication. Hum Exp Toxicol. 2003;22(6):341–343. doi: 10.1191/0960327103ht347cr. [DOI] [PubMed] [Google Scholar]
  73. Hectors TLM, Vanparys C, van der Ven K, Martens GA, Jorens PG, Van Gaal LF, Covaci A, De Coen W, Blust R. Environmental pollutants and type 2 diabetes: a review of mechanisms that can disrupt beta cell function. Diabetologia. 2011;54(6):1273–1290. doi: 10.1007/s00125-011-2109-5. [DOI] [PubMed] [Google Scholar]
  74. Hernandez AF, Parron T, Tsatsakis AM, Requena M, Alarcon R, Lopez-Guarnido O. Toxic effects of pesticide mixtures at a molecular level: Their relevance to human health. Toxicology. 2013;307:136–145. doi: 10.1016/j.tox.2012.06.009. [DOI] [PubMed] [Google Scholar]
  75. Hers HG. Mechanisms of blood glucose homeostasis. J Inherit Metab Dis. 1990;13(4):395–410. doi: 10.1007/BF01799497. [DOI] [PubMed] [Google Scholar]
  76. Ho E, Bray TM. Antioxidants, NFkappaB activation, and diabetogenesis. Proc Soc Exp Biol Med. 1999;222(3):205–213. doi: 10.1046/j.1525-1373.1999.d01-137.x. [DOI] [PubMed] [Google Scholar]
  77. Hossain MM, Richardson JR. Mechanism of pyrethroid pesticide-induced apoptosis: role of calpain and the ER stress pathway. Toxicol Sci. 2011;122(2):512–525. doi: 10.1093/toxsci/kfr111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 2010;140(6):900–917. doi: 10.1016/j.cell.2010.02.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Howell G, 3rd, Mangum L. Exposure to bioaccumulative organochlorine compounds alters adipogenesis, fatty acid uptake, and adipokine production in NIH3T3-L1 cells. Toxicol In Vitro. 2011;25(1):394–402. doi: 10.1016/j.tiv.2010.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Howell GE, 3rd, Mulligan C, Meek E, Chambers JE. Effect of chronic p,p'-dichlorodiphenyldichloroethylene (DDE) exposure on high fat diet-induced alterations in glucose and lipid metabolism in male C57BL/6H mice. Toxicology. 2015;328:112–122. doi: 10.1016/j.tox.2014.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Howell GE, Meek E, Kilic J, Mohns M, Mulligan C, Chambers JE. Exposure to p,p '-dichlorodiphenyldichloroethylene (DDE) induces fasting hyperglycemia without insulin resistance in male C57BL/6H mice. Toxicology. 2014;320:6–14. doi: 10.1016/j.tox.2014.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Hsiao CT, Yang CC, Deng JF, Bullard MJ, Liaw SJ. Acute pancreatitis following organophosphate intoxication. J Toxicol Clin Toxicol. 1996;34(3):343–347. doi: 10.3109/15563659609013800. [DOI] [PubMed] [Google Scholar]
  83. Hummasti S, Hotamisligil GS. Endoplasmic reticulum stress and inflammation in obesity and diabetes. Circ Res. 2010;107(5):579–591. doi: 10.1161/CIRCRESAHA.110.225698. [DOI] [PubMed] [Google Scholar]
  84. Husain K, Ansari RA. Influence of cholinergic and adrenergic blocking drugs on hyperglycemia and brain glycogenolysis in diazinon-treated animals. Can J Physiol Pharmacol. 1988;66(9):1144–1147. doi: 10.1139/y88-188. [DOI] [PubMed] [Google Scholar]
  85. Ibrahim NA, El-Gamal BA. Effect of diazinon, an organophosphate insecticide, on plasma lipid constituents in experimental animals. J Biochem Mol Biol. 2003;36(5):499–504. doi: 10.5483/bmbrep.2003.36.5.499. [DOI] [PubMed] [Google Scholar]
  86. Ince S, Kucukkurt I, Demirel HH, Turkmen R, Sever E. Thymoquinone attenuates cypermethrin induced oxidative stress in Swiss albino mice. Pestic Biochem Physiol. 2012;104(3):229–235. [Google Scholar]
  87. Jamshidi HR, Ghahremani MH, Ostad SN, Sharifzadeh M, Dehpour AR, Abdollahi M. Effects of diazinon on the activity and gene expression of mitochondrial glutamate dehydrogenase from rat pancreatic Langerhans islets. Pestic Biochem Physiol. 2009;93(1):23–27. [Google Scholar]
  88. Joshi AKR, Rajini PS. Reversible hyperglycemia in rats following acute exposure to acephate, an organophosphorus insecticide: Role of gluconeogenesis. Toxicology. 2009;257(1–2):40–45. doi: 10.1016/j.tox.2008.12.006. [DOI] [PubMed] [Google Scholar]
  89. Kakkar R, Mantha SV, Radhi J, Prasad K, Kalra J. Increased oxidative stress in rat liver and pancreas during progression of streptozotocin-induced diabetes. Clin Sci (Lond) 1998;94(6):623–632. doi: 10.1042/cs0940623. [DOI] [PubMed] [Google Scholar]
  90. Kalender S, Ogutcu A, Uzunhisarcili M, Acikgoz F, Durak D, Ulusoy Y, Kalender Y. Diazinon-induced hepatotoxicity and protective effect of vitamin E on some biochemical indices and ultrastructural changes. Toxicology. 2005;211(3):197–206. doi: 10.1016/j.tox.2005.03.007. [DOI] [PubMed] [Google Scholar]
  91. Kamath V, Rajini PS. Altered glucose homeostasis and oxidative impairment in pancreas of rats subjected to dimethoate intoxication. Toxicology. 2007;231(2–3):137–146. doi: 10.1016/j.tox.2006.11.072. [DOI] [PubMed] [Google Scholar]
  92. Kandalaft K, Liu S, Manivel C, Borner JW, Dressel TD, Sutherland DE, Goodale RL. Organophosphate increases the sensitivity of human exocrine pancreas to acetylcholine. Pancreas. 1991;6(4):398–403. doi: 10.1097/00006676-199107000-00004. [DOI] [PubMed] [Google Scholar]
  93. Karami-Mohajeri S, Abdollahi M. Toxic influence of organophosphate, carbamate, and organochlorine pesticides on cellular metabolism of lipids, proteins, and carbohydrates: a systematic review. Hum Exp Toxicol. 2011;30(9):1119–1140. doi: 10.1177/0960327110388959. [DOI] [PubMed] [Google Scholar]
  94. Karmaus W, Osuch JR, Eneli I, Mudd LM, Zhang J, Mikucki D, Haan P, Davis S. Maternal levels of dichlorodiphenyl-dichloroethylene (DDE) may increase weight and body mass index in adult female offspring. Occup Environ Med. 2009;66(3):143–149. doi: 10.1136/oem.2008.041921. [DOI] [PubMed] [Google Scholar]
  95. Kauffman FC, Davis LH, Whittaker M. Activation of glycogen phosphorylase in rat pheochromocytoma PC12 cells and isolated hepatocytes by organophosphates. Biochem Pharmacol. 1990;39(2):347–354. doi: 10.1016/0006-2952(90)90034-i. [DOI] [PubMed] [Google Scholar]
  96. Kim J, Park Y, Yoon KS, Clark JM, Park Y. Imidacloprid, a neonicotinoid insecticide, induces insulin resistance. J Toxicol Sci. 2013;38(5):655–660. doi: 10.2131/jts.38.655. [DOI] [PubMed] [Google Scholar]
  97. Kim J, Park Y, Yoon KS, Clark JM, Park Y. Permethrin alters adipogenesis in 3T3-L1 adipocytes and causes insulin resistance in C2C12 myotubes. J Biochem Mol Toxicol. 2014a;28(9):418–424. doi: 10.1002/jbt.21580. [DOI] [PubMed] [Google Scholar]
  98. Kim J, Sun Q, Yue Y, Yoon KS, Whang KY, Marshall Clark J, Park Y. 4,4'-Dichlorodiphenyltrichloroethane (DDT) and 4,4'-dichlorodiphenyldichloroethylene (DDE) promote adipogenesis in 3T3-L1 adipocyte cell culture. Pestic Biochem Physiol. 2016;131:40–45. doi: 10.1016/j.pestbp.2016.01.005. [DOI] [PubMed] [Google Scholar]
  99. Kim JA, Wei Y, Sowers JR. Role of mitochondrial dysfunction in insulin resistance. Circ Res. 2008;102(4):401–414. doi: 10.1161/CIRCRESAHA.107.165472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Kim KS, Lee YM, Kim SG, Lee IK, Lee HJ, Kim JH, Kim J, Moon HB, Jacobs DR, Jr, Lee DH. Associations of organochlorine pesticides and polychlorinated biphenyls in visceral vs. subcutaneous adipose tissue with type 2 diabetes and insulin resistance. Chemosphere. 2014b;94:151–157. doi: 10.1016/j.chemosphere.2013.09.066. [DOI] [PubMed] [Google Scholar]
  101. Kojima H, Takeuchi S, Nagai T. Endocrine-disrupting potential of pesticides via nuclear receptors and aryl hydrocarbon receptor. J Health Sci. 2010;56(4):374–386. [Google Scholar]
  102. Kuo CC, Moon K, Thayer KA, Navas-Acien A. Environmental chemicals and type 2 diabetes: an updated systematic review of the epidemiologic evidence. Curr Diab Rep. 2013;13(6):831–849. doi: 10.1007/s11892-013-0432-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. La Merrill M, Karey E, Moshier E, Lindtner C, La Frano MR, Newman JW, Buettner C. Perinatal exposure of mice to the pesticide DDT impairs energy expenditure and metabolism in adult female offspring. PLoS One. 2014;9(7):e103337. doi: 10.1371/journal.pone.0103337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Langer P, Ukropec J, Kocan A, Drobna B, Radikova Z, Huckova M, Imrich R, Gasperikova D, Klimes I, Trnovec T. Obesogenic and diabetogenic impact of high organochlorine levels (HCB, p,p'-DDE, PCBs) on inhabitants in the highly polluted Eastern Slovakia. Endocr Regul. 2014;48(1):17–24. doi: 10.4149/endo_2014_01_17. [DOI] [PubMed] [Google Scholar]
  105. Lankisch PG, Muller CH, Niederstadt H, Brand A. Painless acute pancreatitis subsequent to anticholinesterase insecticide (parathion) intoxication. Am J Gastroenterol. 1990;85(7):872–875. [PubMed] [Google Scholar]
  106. Lassiter TL, Brimijoin S. Rats gain excess weight after developmental exposure to the organophosphorothionate pesticide, chlorpyrifos. Neurotoxicol Teratol. 2008;30(2):125–130. doi: 10.1016/j.ntt.2007.10.004. [DOI] [PubMed] [Google Scholar]
  107. Lassiter TL, Ryde IT, Mackillop EA, Brown KK, Levin ED, Seidler FJ, Slotkin TA. Exposure of neonatal rats to parathion elicits sex-selective reprogramming of metabolism and alters the response to a high-fat diet in adulthood. Environ Health Perspect. 2008;116(11):1456–1462. doi: 10.1289/ehp.11673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Lee DH, Lee IK, Jin SH, Steffes M, Jacobs DR. Association between serum concentrations of persistent organic pollutants and insulin resistance among nondiabetic adults. Diabetes Care. 2007a;30(3):622–628. doi: 10.2337/dc06-2190. [DOI] [PubMed] [Google Scholar]
  109. Lee DH, Lee IK, Porta M, Steffes M, Jacobs DR. Relationship between serum concentrations of persistent organic pollutants and the prevalence of metabolic syndrome among non-diabetic adults: results from the National Health and Nutrition Examination Survey 1999–2002. Diabetologia. 2007b;50(9):1841–1851. doi: 10.1007/s00125-007-0755-4. [DOI] [PubMed] [Google Scholar]
  110. Lee DH, Lee IK, Song K, Steffes M, Toscano W, Baker BA, Jacobs DR. A strong dose-response relation between serum concentrations of persistent organic pollutants and diabetes - Results from the National Health and Examination Survey 1999–2002. Diabetes Care. 2006;29(7):1638–1644. doi: 10.2337/dc06-0543. [DOI] [PubMed] [Google Scholar]
  111. Lee DH, Lind PM, Jacobs DR, Jr, Salihovic S, van Bavel B, Lind L. Polychlorinated biphenyls and organochlorine pesticides in plasma predict development of type 2 diabetes in the elderly: the prospective investigation of the vasculature in Uppsala Seniors (PIVUS) study. Diabetes Care. 2011a;34(8):1778–1784. doi: 10.2337/dc10-2116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Lee DH, Porta M, Jacobs DR, Jr, Vandenberg LN. Chlorinated persistent organic pollutants, obesity, and type 2 diabetes. Endocr Rev. 2014a;35(4):557–601. doi: 10.1210/er.2013-1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Lee DH, Steffes MW, Sjodin A, Jones RS, Needham LL, Jacobs DR. Low dose of some persistent organic pollutants predicts type 2 diabetes: a nested case-control study. Environ Health Persp. 2010;118(9):1235–1242. doi: 10.1289/ehp.0901480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Lee DH, Steffes MW, Sjodin A, Jones RS, Needham LL, Jacobs DR., Jr Low dose organochlorine pesticides and polychlorinated biphenyls predict obesity, dyslipidemia, and insulin resistance among people free of diabetes. PLoS One. 2011b;6(1):e15977. doi: 10.1371/journal.pone.0015977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Lee HK. Mitochondrial dysfunction and insulin resistance: the contribution of dioxin-like substances. Diabetes Metab J. 2011;35(3):207–215. doi: 10.4093/dmj.2011.35.3.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Lee YM, Kim KS, Kim SA, Hong NS, Lee SJ, Lee DH. Prospective associations between persistent organic pollutants and metabolic syndrome: A nested case-control study. Sci Total Environ. 2014b;496:219–225. doi: 10.1016/j.scitotenv.2014.07.039. [DOI] [PubMed] [Google Scholar]
  117. Lemaire G, Balaguer P, Michel S, Rahmani R. Activation of retinoic acid receptor-dependent transnocription by orgachlorine pesticides. Toxicol Appl Pharm. 2005;202(1):38–49. doi: 10.1016/j.taap.2004.06.004. [DOI] [PubMed] [Google Scholar]
  118. Lim S, Ahn SY, Song IC, Chung MH, Jang HC, Park KS, Lee KU, Pak YK, Lee HK. Chronic exposure to the herbicide, atrazine, causes mitochondrial dysfunction and insulin resistance. PLoS One. 2009;4(4):e5186. doi: 10.1371/journal.pone.0005186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Lowell BB, Shulman GI. Mitochondrial dysfunction and type 2 diabetes. Science. 2005;307(5708):384–387. doi: 10.1126/science.1104343. [DOI] [PubMed] [Google Scholar]
  120. Ma ZA, Zhao Z, Turk J. Mitochondrial dysfunction and beta-cell failure in type 2 diabetes mellitus. Exp Diabetes Res. 2012;2012:703538. doi: 10.1155/2012/703538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Mackness MI, Harty D, Bhatnagar D, Winocour PH, Arrol S, Ishola M, Durrington PN. Serum paraoxonase activity in familial hypercholesterolaemia and insulin-dependent diabetes mellitus. Atherosclerosis. 1991;86(2–3):193–199. doi: 10.1016/0021-9150(91)90215-o. [DOI] [PubMed] [Google Scholar]
  122. Malekirad AA, Faghih M, Mirabdollahi M, Kiani M, Fathi A, Abdollahi M. Neurocognitive, mental health, and glucose disorders in farmers exposed to organophosphorus pesticides. Arh Hig Rada Toksikol. 2013;64(1):1–8. doi: 10.2478/10004-1254-64-2013-2296. [DOI] [PubMed] [Google Scholar]
  123. Mangum LH, Howell GE, III, Chambers JE. Exposure to p,p '-DDE enhances differentiation of 3T3-L1 preadipocytes in a model of sub-optimal differentiation. Toxicol Lett. 2015;238(2):65–71. doi: 10.1016/j.toxlet.2015.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Marsh WH, Vukov GA, Conradi EC. Acute pancreatitis after cutaneous exposure to an organophosphate insecticide. Am J Gastroenterol. 1988;83(10):1158–1160. [PubMed] [Google Scholar]
  125. Matin MA, Husain K. Cerebral glycogenolysis and glycolysis in malathion-treated hyperglycaemic animals. Biochem Pharmacol. 1987;36(11):1815–1817. doi: 10.1016/0006-2952(87)90243-7. [DOI] [PubMed] [Google Scholar]
  126. Matin MA, Husain K, Khan SN. Modification of diazinon-induced changes in carbohydrate-metabolism by adrenalectomy in rats. Biochem Pharmacol. 1990a;39(11):1781–1786. doi: 10.1016/0006-2952(90)90125-5. [DOI] [PubMed] [Google Scholar]
  127. Matin MA, Sattar S, Husain K. Modification of malathion induced neurochemical changes by adrenalectomy in rats. Mol Chem Neuropathol. 1990b;13(1–2):119–128. doi: 10.1007/BF03159913. [DOI] [PubMed] [Google Scholar]
  128. Matsuoka T, Kajimoto Y, Watada H, Kaneto H, Kishimoto M, Umayahara Y, Fujitani Y, Kamada T, Kawamori R, Yamasaki Y. Glycation-dependent, reactive oxygen species-mediated suppression of the insulin gene promoter activity in HIT cells. J Clin Invest. 1997;99(1):144–150. doi: 10.1172/JCI119126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. McKinlay R, Plant JA, Bell JNB, Voulvoulis N. Endocrine disrupting pesticides: implications for risk assessment. Environ Int. 2008;34(2):168–183. doi: 10.1016/j.envint.2007.07.013. [DOI] [PubMed] [Google Scholar]
  130. Meggs WJ, Brewer KL. Weight gain associated with chronic exposure to chlorpyrifos in rats. J Med Toxicol. 2007;3(3):89–93. doi: 10.1007/BF03160916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Meller D, Fraser I, Kryger M. Hyperglycemia in anticholinesterase poisoning. Can Med Assoc J. 1981;124(6):745–748. [PMC free article] [PubMed] [Google Scholar]
  132. Mendez MA, Garcia-Esteban R, Guxens M, Vrijheid M, Kogevinas M, Goni F, Fochs S, Sunyer J. Prenatal organochlorine compound exposure, rapid weight gain, and overweight in infancy. Environ Health Perspect. 2011;119(2):272–278. doi: 10.1289/ehp.1002169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Montgomery MP, Kamel F, Saldana TM, Alavanja CR, Sandler DP. Incident diabetes and pesticide exposure among licensed pesticide applicators: Agricultural Health Study, 1993–2003. Am J Epidemiol. 2008;167(10):1235–1246. doi: 10.1093/aje/kwn028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Moore PG, James OF. Acute pancreatitis induced by acute organophosphate poisoning. Postgrad Med J. 1981;57(672):660–662. doi: 10.1136/pgmj.57.672.660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Moreno-Aliaga MJ, Matsumura F. Effects of 1,1,1-trichloro-2,2-bis(p-chlorophenyl)-ethane (p,p'-DDT) on 3T3-L1 and 3T3-F442A adipocyte differentiation. Biochem Pharmacol. 2002;63(5):997–1007. doi: 10.1016/s0006-2952(01)00933-9. [DOI] [PubMed] [Google Scholar]
  136. Morgan MK, Sheldon LS, Croghan CW, Jones PA, Chuang JC, Wilson NK. An observational study of 127 preschool children at their homes and daycare centers in Ohio: Environmental pathways to cis- and trans-permethrin exposure. Environ Res. 2007;104(2):266–274. doi: 10.1016/j.envres.2006.11.011. [DOI] [PubMed] [Google Scholar]
  137. Moritz F, Droy JM, Dutheil G, Melki J, Bonmarchand G, Leroy J. Acute-pancreatitis after carbamate insecticide intoxication. Intens Care Med. 1994;20(1):49–50. doi: 10.1007/BF02425057. [DOI] [PubMed] [Google Scholar]
  138. Mostafalou S, Abdollahi M. The role of environmental pollution of pesticides in human diabetes. Int J Pharmacol. 2012;8(2):139–140. [Google Scholar]
  139. Mostafalou S, Abdollahi M. Pesticides and human chronic diseases: evidences, mechanisms, and perspectives. Toxicol Appl Pharmacol. 2013;268(2):157–177. doi: 10.1016/j.taap.2013.01.025. [DOI] [PubMed] [Google Scholar]
  140. Mostafalou S, Eghbal MA, Nili-Ahmadabadi A, Baeeri M, Abdollahi M. Biochemical evidence on the potential role of organophosphates in hepatic glucose metabolism toward insulin resistance through inflammatory signaling and free radical pathways. Toxicol Ind Health. 2012;28(9):840–851. doi: 10.1177/0748233711425073. [DOI] [PubMed] [Google Scholar]
  141. Narendra M, Kavitha G, Helah Kiranmai A, Raghava Rao N, Varadacharyulu NC. Chronic exposure to pyrethroid-based allethrin and prallethrin mosquito repellents alters plasma biochemical profile. Chemosphere. 2008;73(3):360–364. doi: 10.1016/j.chemosphere.2008.05.070. [DOI] [PubMed] [Google Scholar]
  142. Nauen R, Bretschneider T. New modes of action of insecticides. Pestic Outlook. 2002;13 [Google Scholar]
  143. NCHS. Prevalence of Obesity in the United States, 2009–2010. National Center for Health Statistics, Centers for Disease Control and Prevention; 2012. https://www.cdc.gov/nchs/data/databriefs/db82.htm. [Google Scholar]
  144. Nordlie RC, Foster JD, Lange AJ. Regulation of glucose production by the liver. Annu Rev Nutr. 1999;19:379–406. doi: 10.1146/annurev.nutr.19.1.379. [DOI] [PubMed] [Google Scholar]
  145. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Gorgun C, Glimcher LH, Hotamisligil GS. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306(5695):457–461. doi: 10.1126/science.1103160. [DOI] [PubMed] [Google Scholar]
  146. Pal S, Blais JM, Robidoux MA, Haman F, Krummel E, Seabert TA, Imbeault P. The association of type 2 diabetes and insulin resistance/secretion with persistent organic pollutants in two First Nations communities in northern Ontario. Diabetes Metab. 2013;39(6):497–504. doi: 10.1016/j.diabet.2013.01.006. [DOI] [PubMed] [Google Scholar]
  147. Panieri E, Krige JE, Bornman PC, Linton DM. Severe necrotizing pancreatitis caused by organophosphate poisoning. J Clin Gastroenterol. 1997;25(2):463–465. doi: 10.1097/00004836-199709000-00015. [DOI] [PubMed] [Google Scholar]
  148. Park Y, Kim Y, Kim J, Yoon KS, Clark J, Lee J, Park Y. Imidacloprid, a neonicotinoid insecticide, potentiates adipogenesis in 3T3-L1 adipocytes. J Agric Food Chem. 2013;61(1):255–259. doi: 10.1021/jf3039814. [DOI] [PubMed] [Google Scholar]
  149. Patel CJ, Bhattacharya J, Butte AJ. An environment-wide association Study (EWAS) on type 2 diabetes mellitus. PLoS One. 2010;5(5):e10746. doi: 10.1371/journal.pone.0010746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Peris-Sampedro F, Cabre M, Basaure P, Reverte I, Domingo JL, Teresa Colomina M. Adulthood dietary exposure to a common pesticide leads to an obese-like phenotype and a diabetic profile in apoE3 mice. Environ Res. 2015;142:169–176. doi: 10.1016/j.envres.2015.06.036. [DOI] [PubMed] [Google Scholar]
  151. Philibert A, Schwartz H, Mergler D. An exploratory study of diabetes in a First Nation community with respect to serum concentrations of p,p'-DDE and PCBs and fish consumption. Int J Environ Res Public Health. 2009;6(12):3179–3189. doi: 10.3390/ijerph6123179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Polderman KH, Gooren LJ, Asscheman H, Bakker A, Heine RJ. Induction of insulin resistance by androgens and estrogens. J Clin Endocrinol Metab. 1994;79(1):265–271. doi: 10.1210/jcem.79.1.8027240. [DOI] [PubMed] [Google Scholar]
  153. Pournourmohammadi S, Farzami B, Ostad SN, Azizi E, Abdollahi M. Effects of malathion subchronic exposure on rat skeletal muscle glucose metabolism. Environ Toxicol Pharmacol. 2005;19(1):191–196. doi: 10.1016/j.etap.2004.07.002. [DOI] [PubMed] [Google Scholar]
  154. Pournourmohammadi S, Ostad SN, Azizi E, Ghahremani MH, Farzami B, Minaie B, Larijani B, Abdollahi M. Induction of insulin resistance by malathion: Evidence for disrupted islets cells metabolism and mitochondrial dysfunction. Pestic Biochem Physiol. 2007;88(3):346–352. [Google Scholar]
  155. Raafat N, Abass MA, Salem HM. Malathion exposure and insulin resistance among a group of farmers in Al-Sharkia governorate. Clin Biochem. 2012;45(18):1591–1595. doi: 10.1016/j.clinbiochem.2012.07.108. [DOI] [PubMed] [Google Scholar]
  156. Rahimi R, Abdollahi M. A review on the mechanisms involved in hyperglycemia induced by organophosphorus pesticides. Pestic Biochem Physiol. 2007;88(2):115–121. [Google Scholar]
  157. Ramu A, Drexler H. Hyperglycemia in acute malathion intoxication in rats. Isr J Med Sci. 1973;9(5):635–639. [PubMed] [Google Scholar]
  158. Ramu A, Slonim AE, London M, Eyal F. Hyperglycemia in acute malathion poisoning. Isr J Med Sci. 1973;9(5):631–634. [PubMed] [Google Scholar]
  159. Ranjbar A, Pasalar P, Abdollahi M. Induction of oxidative stress and acetylcholinesterase inhibition in organophosphorous pesticide manufacturing workers. Hum Exp Toxicol. 2002;21(4):179–182. doi: 10.1191/0960327102ht238oa. [DOI] [PubMed] [Google Scholar]
  160. Ray DE, Cremer JE. Action of decamethrin (a synthetic pyrethroid) on the rat. Pestic Biochem Physiol. 1979;10(3):333–340. [Google Scholar]
  161. Reena K, Ajay K, Sharma CB. Haematological changes induced by dimethoate in rat. Arh Hig Rada Toksikol. 1989;40(1):23–27. [PubMed] [Google Scholar]
  162. Rezg R, Mornagui B, Benahmed M, Chouchane SG, Belhajhmida N, Abdeladhim M, Kamoun A, El-fazaa S, Gharbi N. Malathion exposure modulates hypothalamic gene expression and induces dyslipedemia in Wistar rats. Food Chem Toxicol. 2010a;48(6):1473–1477. doi: 10.1016/j.fct.2010.03.013. [DOI] [PubMed] [Google Scholar]
  163. Rezg R, Mornagui B, El-Arbi M, Kamoun A, El-Fazaa S, Gharbi N. Effect of subchronic exposure to malathion on glycogen phosphorylase and hexokinase activities in rat liver using native PAGE. Toxicology. 2006;223(1–2):9–14. doi: 10.1016/j.tox.2006.02.020. [DOI] [PubMed] [Google Scholar]
  164. Rezg R, Mornagui B, El-Fazaa S, Gharbi N. Caffeic acid attenuates malathion induced metabolic disruption in rat liver, involvement of acetylcholinesterase activity. Toxicology. 2008;250(1):27–31. doi: 10.1016/j.tox.2008.05.017. [DOI] [PubMed] [Google Scholar]
  165. Rezg R, Mornagui B, El-Fazaa S, Gharbi N. Organophosphorus pesticides as food chain contaminants and type 2 diabetes: a review. Trends Food Sci Tech. 2010b;21(7):345–357. [Google Scholar]
  166. Rezg R, Mornagui B, Kamoun A, El-Fazaa S, Gharbi N. Effect of subchronic exposure to malathion on metabolic parameters in the rat. C R Biol. 2007;330(2):143–147. doi: 10.1016/j.crvi.2006.11.002. [DOI] [PubMed] [Google Scholar]
  167. Rignell-Hydbom A, Lidfeldt J, Kiviranta H, Rantakokko P, Samsioe G, Agardh CD, Rylander L. Exposure to p,p'-DDE: a risk factor for type 2 diabetes. PLoS One. 2009;4(10):e7503. doi: 10.1371/journal.pone.0007503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Rignell-Hydbom A, Rylander L, Hagmar L. Exposure to persistent organochlorine pollutants and type 2 diabetes mellitus. Hum Exp Toxicol. 2007;26(5):447–452. doi: 10.1177/0960327107076886. [DOI] [PubMed] [Google Scholar]
  169. Risher JF, Mink FL, Stara JF. The toxicologic effects of the carbamate insecticide aldicarb in mammals: a review. Environ Health Perspect. 1987;72:267–281. doi: 10.1289/ehp.8772267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Rodrigues MA, Puga FR, Chenker E, Mazanti MT. Short-term effect of malathion on rats' blood glucose and on glucose utilization by mammalian cells in vitro. Ecotoxicol Environ Saf. 1986;12(2):110–113. doi: 10.1016/0147-6513(86)90046-1. [DOI] [PubMed] [Google Scholar]
  171. Romero-Navarro G, Lopez-Aceves T, Rojas-Ochoa A, Fernandez Mejia C. Effect of dichlorvos on hepatic and pancreatic glucokinase activity and gene expression, and on insulin mRNA levels. Life Sci. 2006;78(9):1015–1020. doi: 10.1016/j.lfs.2005.06.010. [DOI] [PubMed] [Google Scholar]
  172. Rylander L R-HA, Hagmar L. A cross-sectional study of the association between persistent organochlorine pollutants and diabetes. Environ Health. 2005;4(28) doi: 10.1186/1476-069X-4-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Saldana TM, Basso O, Hoppin JA, Baird DD, Knott C, Blair A, Alavanja MCR, Sandler DP. Pesticide exposure and self-reported gestational diabetes mellitus in the agricultural health study. Diabetes Care. 2007;30(3):529–534. doi: 10.2337/dc06-1832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Sarin S, Gill KD. Dichlorvos induced alterations in glucose homeostasis: possible implications on the state of neuronal function in rats. Mol Cell Biochem. 1999;199(1–2):87–92. doi: 10.1023/a:1006930511459. [DOI] [PubMed] [Google Scholar]
  175. Shadnia S, Azizi E, Hosseini R, Khoei S, Fouladdel S, Pajoumand A, Jalali N, Abdollahi M. Evaluation of oxidative stress and genotoxicity in organophosphorus insecticide formulators. Hum Exp Toxicol. 2005;24(9):439–445. doi: 10.1191/0960327105ht549oa. [DOI] [PubMed] [Google Scholar]
  176. Shakoori AR, Ali SS, Saleem MA. Effects of six months' feeding of cypermethrin on the blood and liver of albino rats. J Biochem Toxicol. 1988;3:59–71. doi: 10.1002/jbt.2570030107. [DOI] [PubMed] [Google Scholar]
  177. Shakoori AR, Rasul YG, Ali SS. The effect of long term administration of dieldrin on biochemical components in blood serum of albino rats. Folia Biol (Krakow) 1984;32(3):213–222. [PubMed] [Google Scholar]
  178. Shapiro GD, Dodds L, Arbuckle TE, Ashley-Martin J, Ettinger AS, Fisher M, Taback S, Bouchard MF, Monnier P, Dallaire R, Morisset AS, Fraser W. Exposure to organophosphorus and organochlorine pesticides, perfluoroalkyl substances, and polychlorinated biphenyls in pregnancy and the association with impaired glucose tolerance and gestational diabetes mellitus: The MIREC Study. Environ Res. 2016;147:71–81. doi: 10.1016/j.envres.2016.01.040. [DOI] [PubMed] [Google Scholar]
  179. Shen P, Hsieh TH, Yue Y, Sun Q, Clark JM, Park Y. Deltamethrin increases the fat accumulation in 3T3-L1 adipocytes and Caenorhabditis elegans. Food Chem Toxicol. 2017;101:149–156. doi: 10.1016/j.fct.2017.01.015. [DOI] [PubMed] [Google Scholar]
  180. Skandrani D, Gaubin Y, Beau B, Murat JC, Vincent C, Croute F. Effect of selected insecticides on growth rate and stress protein expression in cultured human A549 and SH-SY5Y cells. Toxicol In Vitro. 2006a;20(8):1378–1386. doi: 10.1016/j.tiv.2006.06.001. [DOI] [PubMed] [Google Scholar]
  181. Skandrani D, Gaubin Y, Vincent C, Beau B, Claude Murat J, Soleilhavoup JP, Croute F. Relationship between toxicity of selected insecticides and expression of stress proteins (HSP, GRP) in cultured human cells: effects of commercial formulations versus pure active molecules. Biochim Biophys Acta. 2006b;1760(1):95–103. doi: 10.1016/j.bbagen.2005.09.015. [DOI] [PubMed] [Google Scholar]
  182. Skinner MK, Manikkam M, Tracey R, Guerrero-Bosagna C, Haque M, Nilsson EE. Ancestral dichlorodiphenyltrichloroethane (DDT) exposure promotes epigenetic transgenerational inheritance of obesity. Bmc Medicine. 2013;11 doi: 10.1186/1741-7015-11-228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Slaninova A, Smutna M, Modra H, Svobodova Z. A review: oxidative stress in fish induced by pesticides. Neuro Endocrinol Lett. 2009;30(Suppl 1):2–12. [PubMed] [Google Scholar]
  184. Slotkin TA, Brown KK, Seidler FJ. Developmental exposure of rats to chlorpyrifos elicits sex-selective hyperlipidemia and hyperinsulinemia in adulthood. Environ Health Perspect. 2005;113(10):1291–1294. doi: 10.1289/ehp.8133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Soderlund DM, Clark JM, Sheets LP, Mullin LS, Piccirillo VJ, Sargent D, Stevens JT, Weiner ML. Mechanisms of pyrethroid neurotoxicity: implications for cumulative risk assessment. Toxicology. 2002;171(1):3–59. doi: 10.1016/s0300-483x(01)00569-8. [DOI] [PubMed] [Google Scholar]
  186. Soltaninejad K, Abdollahi M. Current opinion on the science of organophosphate pesticides and toxic stress: A systematic review. Med Sci Monitor. 2009;15(3):Ra75–Ra90. [PubMed] [Google Scholar]
  187. Son HK, Kim SA, Kang JH, Chang YS, Park SK, Lee SK, Jacobs DR, Lee DH. Strong associations between low-dose organochlorine pesticides and type 2 diabetes in Korea. Environ Int. 2010;36(5):410–414. doi: 10.1016/j.envint.2010.02.012. [DOI] [PubMed] [Google Scholar]
  188. Song C, Kanthasamy A, Anantharam V, Sun F, Kanthasamy AG. Environmental neurotoxic pesticide increases histone acetylation to promote apoptosis in dopaminergic neuronal cells: relevance to epigenetic mechanisms of neurodegeneration. Mol Pharmacol. 2010;77(4):621–632. doi: 10.1124/mol.109.062174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Sparks TC. Insecticide discovery: An evaluation and analysis. Pestic Biochem Physiol. 2013;107(1):8–17. doi: 10.1016/j.pestbp.2013.05.012. [DOI] [PubMed] [Google Scholar]
  190. Sparks TC, Nauen R. IRAC: Mode of action classification and insecticide resistance management. Pestic Biochem Physiol. 2015;121:122–128. doi: 10.1016/j.pestbp.2014.11.014. [DOI] [PubMed] [Google Scholar]
  191. Sun Q, Qi W, Yang J, Yoon KS, Clark JM, Park Y. Fipronil promotes adipogenesis via AMPK alpha-mediated pathway in 3T3-L1 adipocytes. Food Chem Toxicol. 2016a;92:217–223. doi: 10.1016/j.fct.2016.04.011. [DOI] [PubMed] [Google Scholar]
  192. Sun Q, Xiao X, Kim Y, Kim D, Yoon KS, Clark JM, Park Y. Imidacloprid promotes high fat diet-induced adiposity and insulin resistance in male C57BL/6J mice. J Agric Food Chem. 2016b;64(49):9293–9306. doi: 10.1021/acs.jafc.6b04322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Sungur M, Guven M. Intensive care management of organophosphate insecticide poisoning. Crit Care. 2001;5(4):211–215. doi: 10.1186/cc1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Swaminathan K, Sundaram M, Prakash P, Subbiah S. Diabetic ketoacidosis: an uncommon manifestation of pesticide poisoning. Diabetes Care. 2013;36(1):e4. doi: 10.2337/dc12-1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Teeyapant P, Ramchiun S, Polputpisatkul D, Uttawichai C, Parnmen S. Serum concentrations of organochlorine pesticides p,p'-DDE in adult Thai residents with background levels of exposure. J Toxicol Sci. 2014;39(1):121–127. doi: 10.2131/jts.39.121. [DOI] [PubMed] [Google Scholar]
  196. Teichert-Kuliszewska K, Szymczyk T. Changes in rat carbohydrate metabolism after acute and chronic treatment with dichlorvos. Toxicol Appl Pharmacol. 1979;47(2):323–330. doi: 10.1016/0041-008x(79)90327-2. [DOI] [PubMed] [Google Scholar]
  197. Teimouri F, Amirkabirian N, Esmaily H, Mohammadirad A, Aliahmadi A, Abdollahi M. Alteration of hepatic cells glucose metabolism as a non-cholinergic detoxication mechanism in counteracting diazinon-induced oxidative stress. Hum Exp Toxicol. 2006;25(12):697–703. doi: 10.1177/0960327106075064. [DOI] [PubMed] [Google Scholar]
  198. The European Commision. Decision by the European Commission to restrict the use of imidacloprid, thiamethoxam and clothianidin (Regulation EU No. 485/2013) 2016 http://eur-lex.europa.eu/eli/reg_impl/2013/485/oj.
  199. Turyk M, Anderson H, Knobeloch L, Imm P, Persky V. Organochlorine exposure and incidence of diabetes in a cohort of great lakes sport fish consumers. Environ Health Persp. 2009;117(7):1076–1082. doi: 10.1289/ehp.0800281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Turyk M, Fantuzzi G, Persky V, Freels S, Lambertino A, Pini M, Rhodes DH, Anderson HA. Persistent organic pollutants and biomarkers of diabetes risk in a cohort of Great Lakes sport caught fish consumers. Environ Res. 2015;140:335–344. doi: 10.1016/j.envres.2015.03.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Ueyama J, Kamijima M, Asai K, Mochizuki A, Wang D, Kondo T, Suzuki T, Takagi K, Takagi K, Kanazawa H, Miyamoto K, Wakusawa S, Hasegawa T. Effect of the organophosphorus pesticide diazinon on glucose tolerance in type 2 diabetic rats. Toxicol Lett. 2008;182(1–3):42–47. doi: 10.1016/j.toxlet.2008.08.004. [DOI] [PubMed] [Google Scholar]
  202. Ukropec J, Radikova Z, Huckova M, Koska J, Kocan A, Sebokova E, Drobna B, Trnovec T, Susienkova K, Labudova V, Gasperikova D, Langer P, Klimes I. High prevalence of prediabetes and diabetes in a population exposed to high levels of an organochlorine cocktail. Diabetologia. 2010;53(5):899–906. doi: 10.1007/s00125-010-1683-2. [DOI] [PubMed] [Google Scholar]
  203. USDA. Pesticide Data Program Annual Summary. United States Department of Agriculture; 2014. [Google Scholar]
  204. van der Kallen CJ, van Greevenbroek MM, Stehouwer CD, Schalkwijk CG. Endoplasmic reticulum stress-induced apoptosis in the development of diabetes: is there a role for adipose tissue and liver? Apoptosis. 2009;14(12):1424–1434. doi: 10.1007/s10495-009-0400-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. van Emden HF, Peakall DB. Beyond silent spring. Springer; 1996. [Google Scholar]
  206. Van Larebeke N, Sioen I, Hond ED, Nelen V, Van de Mieroop E, Nawrot T, Bruckers L, Schoeters G, Baeyens W. Internal exposure to organochlorine pollutants and cadmium and self-reported health status: a prospective study. Int J Hyg Environ Health. 2015;218(2):232–245. doi: 10.1016/j.ijheh.2014.11.002. [DOI] [PubMed] [Google Scholar]
  207. Veerappan M, Hwang I, Pandurangan M. Effect of cypermethrin, carbendazim and their combination on male albino rat serum. Int J Clin Exp Pathol. 2012;93(5):361–369. doi: 10.1111/j.1365-2613.2012.00828.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Villeneuve DC, Van Logten MJ, Den Tonkelaar EM, Greve PA, Vos JG, Speijers GJA, Van Esch GJ. Effect of food deprivation on low level hexa chloro benzene exposure in rats. Sci Total Environ. 1977;8(2):179–186. doi: 10.1016/0048-9697(77)90076-6. [DOI] [PubMed] [Google Scholar]
  209. Wang J, Zhu Y, Cai X, Yu J, Yang X, Cheng J. Abnormal glucose regulation in pyrethroid pesticide factory workers. Chemosphere. 2011a;82(7):1080–1082. doi: 10.1016/j.chemosphere.2010.10.065. [DOI] [PubMed] [Google Scholar]
  210. Wang JS, Zhu YQ, Cai XA, Yu JM, Yang XP, Cheng JX. Abnormal glucose regulation in pyrethroid pesticide factory workers. Chemosphere. 2011b;82(7):1080–1082. doi: 10.1016/j.chemosphere.2010.10.065. [DOI] [PubMed] [Google Scholar]
  211. Weizman Z, Sofer S. Acute pancreatitis in children with anticholinesterase insecticide intoxication. Pediatrics. 1992;90(2):204–206. [PubMed] [Google Scholar]
  212. WHO. Diabetes fact sheet. World Health Organization; 2013. https://web.archive.org/web/20130826174444/ http://www.who.int/mediacentre/factsheets/fs312/en/ [Google Scholar]
  213. Xiao X, Kim Y, Kim D, Yoon KS, Clark JM, Park Y. The Toxicologist: Supplement to Toxicological Sciences. 1. Vol. 144. Society of Toxicology; 2015. Exposure to permethrin alters glucose metabolism in response to high fat diet in female C57BL/6J mice. 2015, Abstract no. 2154. [Google Scholar]
  214. Xiao X, Sun Q, Kim Y, Kim D, Yoon KS, Clark JM, Park Y. The Toxicologist: Supplement to Toxicological Sciences. 1. Vol. 150. Society of Toxicology; 2016. Exposure to permethrin increases body fat mass and alters glucose metabolism in response to high fat diet in male C57BL/6J mice. 2016, Abstract no. 2889. [Google Scholar]
  215. Yang MC, McLean AJ, Rivory LP, Le Couteur DG. Hepatic disposition of neurotoxins and pesticides. Pharmacol Toxicol. 2000;87(6):286–291. doi: 10.1034/j.1600-0773.2000.pto870608.x. [DOI] [PubMed] [Google Scholar]
  216. Yang ZP, Dettbarn WD. Diisopropylphosphorofluoridate-induced cholinergic hyperactivity and lipid peroxidation. Toxicol Appl Pharmacol. 1996;138(1):48–53. doi: 10.1006/taap.1996.0096. [DOI] [PubMed] [Google Scholar]
  217. Yau DT, Mennear JH. The inhibitory effect of DDT on insulin secretion in mice. Toxicol Appl Pharmacol. 1977;39(1):81–88. doi: 10.1016/0041-008x(77)90179-x. [DOI] [PubMed] [Google Scholar]
  218. Yousef MI, Awad TI, Mohamed EH. Deltamethrin-induced oxidative damage and biochemical alterations in rat and its attenuation by Vitamin E. Toxicology. 2006;227(3):240–247. doi: 10.1016/j.tox.2006.08.008. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2
3

RESOURCES