eISSN: 1509-572x
ISSN: 1641-4640
Folia Neuropathologica
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2019
vol. 57
 
Share:
Share:
Review paper

Rotenone: from modelling to implication in Parkinson’s disease

Khaled Radad
1, 2
,
Mubarak Al-Shraim
1
,
Ahmed Al-Emam
1, 3
,
Feixue Wang
4
,
Barbara Kranner
5
,
Wolf-Dieter Rausch
5
,
Rudolf Moldzio
5

1.
Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
2.
Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
3.
Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Egypt
4.
Department of Chinese Medicine, Xuanwu Hospital, Capital Medicine University, Beijing, China
5.
Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine, Vienna, Austria
Folia Neuropathol 2019; 57 (4): 317-326
Online publish date: 2019/11/18
Article file
Get citation
 
PlumX metrics:
 

Introduction

Rotenone, a naturally occurring substance, is derived from the roots, seeds and stems of some tropical plants including Derris, Tephrosia, Lonchocarpus and Mundulea species [26]. Rotenone was discovered hundreds of years ago in South America and Southeast Asia, and nowadays, it is an active ingredient of hundreds of pesticides and piscicides [31]. Most recently, rotenone has attained much attention since 2000 when Betarbet et al. reproduced the major features of Parkinson’s disease (PD) in rotenone-infused rats [3].
In this review, we aimed to address currently reported hazards of rotenone applications in humans, animals and environment, and recent updates on the rotenone model of PD and to discuss its implication in etiopathogenesis of PD.

Historical background

The use of rotenone goes back hundreds of years when Peruvian natives used crude extracts of rotenone-containing plants e.g. cubè plant to catch fish for eating. In the early 1900s, botanists looking for useful plants of commercial values in Peruvian jungles exported large quantities of plant roots and extracts with the major active ingredient being rotenone to the USA for use as insecticides on crops [31]. Rotenone was first isolated between the years 1895 and 1902. While travelling in French Guiana, French botanist Emmanuel Geoffroy isolated an active chemical compound that he called nicouline from the plant Lonchocarpus nicou. In 1902, Japanese chemist Nagai Nagayoshi isolated a pure crystalline compound from Derris elliptica and he called it rotenone. Nicouline and rotenone were recognized to be chemically the same by 1930 [39]. Since 1932, rotenone has been used extensively in fisheries management as a piscicide in North America. Nowadays, rotenone is recognized as the most environmentally benign pesticide and piscicide worldwide [46].

Physical and chemical characters

Rotenone is an odourless and colourless to brownish crystalline powder, which belongs to a class of compounds of related molecular structure referred to as isoflavones. Its empirical formula is C23H22O6 with a molecular weight of 394.41 and a melting point of 165-166°C. It is insoluble in water and on the other hand, it is very soluble in many organic solvents. Rotenone is very sensitive to light and oxygen, decomposes to less toxic products, e.g. rotenolone. The rate of decomposition of rotenone is dependent upon several factors such as temperature, pH, sunlight, depth, dose and presence of organic debris [16].

Toxicokinetics and metabolism

Rotenone is absorbed in relatively different amounts through the gastrointestinal tract, lungs and skin. Low and incomplete absorption of rotenone occurs in the gastrointestinal tract. Fats and oils enhance the absorption of rotenone from the intestines. Rotenone dust can find its way to the lungs through inhalation; experimental inhalation of rotenone dust in rats and dogs resulted in earlier clinical signs than following oral ingestion. Dermal absorption of rotenone is negligible. Application of a single dose of 5 g/kg rotenone on rabbit’s skin resulted in no systemic toxicity or mortalities. Rotenone is metabolized by NADP-linked hepatic microsomal enzymes in the liver. Unabsorbed rotenone is excreted by the faecal route and about 20% of the oral doses are excreted in the urine after 24 h. Several metabolites of rotenone metabolism such as rotenolone, and hydroxyl- and dihydroxyrotenone are identified in the blood, urine, faeces and liver. These metabolites can serve as a biomarker of rotenone exposure [20,24].

Current applications of rotenone

Fisheries management and marine research

Rotenone is still extensively used as a selective piscicide for fisheries management in the USA, Canada and more than 30 countries worldwide. This includes controlling of undesirable fish, eradication of harmful and exotic fish, quantification of fish population, controlling of fish diseases, and restoration of threatened and endangered fish species [46]. Also, rotenone is considered a unique and efficient tool for sampling fishes for marine research [61]. In 2007, the Environmental Protection Agency (EPA) allowed re-registration of all piscicidal uses of rotenone as none of them posed any adverse effects on the humans and environment [76].

Controlling pests

After its discovery in 1848 and for more than 150 years, rotenone had been commonly used as a broad-spectrum insecticide for controlling a wide range of pests in numerous crops and home garden, and lice, ticks and mites in veterinary practices and animal husbandry. Currently, food uses of rotenone have been cancelled in the USA, Canada and the EU. However it continues to be used legally in many other countries [46].

Implication of rotenone use on humans, animals and environment

Rotenone toxicity in humans

The World Health Organization (WHO) classifies rotenone as a moderately hazardous agent (a class II pesticide). Rotenone poisoning in humans is uncommon as: (1) the estimated oral LD50 of rotenone in humans is much higher (300-500 mg/kg b.w.) to expose to it and upon exposure, rotenone is efficiently metabolized in the gut and so little or no rotenone goes to the blood stream [17]. Moreover, the most effective route of exposure, the intravenous one, is difficult to happen and absorption of rotenone through lungs and skin is negligible [20]; (2) Quick decomposition of environmental rotenone to less toxic products decreases the opportunities of human exposure to toxic rotenone [31]; (3) EPA has cancelled all food uses of rotenone since 2006 and only allowed re-registration of its piscicidal uses with strict regulations [46]. Therefore, fatalities of rotenone in humans were only reported following accidental or intended poisoning. In this context, Holland [29] reported that natives in Papua Guinea were seen to eat the roots of plants known to contain rotenone as a method of deliberate suicide. Three fatalities were reported following ingestion of commercially available rotenone-containing formulations in a 3.5-year-old girl [9], a 47-year-old woman [78] and a 49-year-old Tamil man [12]. The three cases showed a similar course of signs including vomiting, irregular respiration, unconsciousness, hypotension and circulatory failure, and they eventually died after 5 h – 3 days [12]. Implication of rotenone in PD development will be discussed in a separate section.

Rotenone toxicity in animals

Rotenone toxicity is moderate and widely varies between and within animal species. The oral LD50 values of rotenone are approx. 1.5 mg/kg b.w., 60-135 mg/kg b.w. and 350 mg/kg b.w. in rabbits, rats and mice, respectively. Systemic uptake of rotenone results in higher toxicity than by the oral route. The estimated intravenous and intraperitoneal LD50 values in rabbits and mice are 0.35-0.65 mg/kg b.w. and 2.8 mg/kg b.w., respectively [20]. In general, common clinical signs of rotenone poisoning in animals include pharyngitis, gastric pain, vomiting, muscle tremors, chronic convulsions and respiratory stimulation followed by depression. It can also lead to severe signs of hypoglycaemia, liver failure, alterations in arterial blood gases and acid base balance, and hypercapnia and hypoxemia due to seizures and respiratory depression. Death can result from cardio-respiratory failure [20]. Compared to mammalian species, rotenone is highly toxic to fish as it is rapidly absorbed from the gastrointestinal tract and directly uptaken through gills to the blood stream [20].

Effects of rotenone on the environment

Because rotenone breaks down quickly by exposure to light and temperature, its impact on the environment is considered to be low. It has low mobility in soil and only travels less than one inch through most soils. It does not leach far into underlying sediment and therefore, it does not affect groundwater supplies [8].

Rotenone model of Parkinson’s disease

In 1985, rotenone was first used by Heikkila et al. to model PD by stereotaxic administration of 5 mg of this mitochondrial complex I inhibitor into the rat brain [25]. Drawbacks of Heikkila’s study were the use of a higher dose of rotenone, i.e. approximately 500,000-fold higher than the half maximal inhibitory concentration (IC50) of 10 nM and the stereotaxic route of administration which is an impracticable route for rotenone exposure. Another trial of using rotenone to model PD was in 1997 when Ferrante et al. [13] administered 18 mg/kg/day intravenously to rats. In addition to the higher dose, the results were nonspecific brain lesions and peripheral toxicity [13]. In 2000, Betarbet et al. succeeded to reproduce the two pathological hallmarks of PD, i.e. the loss of dopaminergic neurons and the formation of Lewy-like bodies in the surviving dopaminergic neurons, as well as some of the parkinsonian motor deficits by systemic administration of rotenone in rats [3]. Following Betarbet et al.’s study, the rotenone model has been widely used by many researchers to investigate both the mechanisms that underlie dopaminergic cell death and to test new potential symptomatic and neuroprotective therapies in PD [34,59]. This is due to: (1) its extreme lipophilicity that enables it to cross cellular membranes independent of any transporter producing systemic inhibition of mitochondrial complex I [19]; (2) its implication in many pathogenic pathways that mediate dopaminergic cell death including oxidative stress, -synuclein phosphorylation and aggregation, and Lewy pathology, DJ-1 acidification and translocation, proteasomal dysfunction and nigral iron accumulation [4]; (3) its capability of reproducing some of the non-motor symptoms of PD, most notably disruption of gastrointestinal and olfactory discrimination [34]; (4) its usefulness in assessing some end-points such as -synuclein accumulation, ubiquitin-proteasome function and GIT dysfunction in addition to preservation of dopaminergic neurons and related motor functions which are the most commonly used endpoints in neuroprotective studies [7]. Beside the intravenous route of rotenone administration used in Betarbet’s study, researchers also adopted oral, subcutaneous and intraperitoneal routes to deliver rotenone in different animal models of PD. Intraperitoneal rotenone administration was reported to produce highly reproducible PD-like lesions including L-dopa-responsive locomotor deficits and loss of dopaminergic neurons in substantia nigra associated with -synuclein pathology. These features make it a well-suited model for the assessment of pathogenic pathways and experimental therapeutic intervention [7]. The subcutaneous route of rotenone administration has recently attracted some attention as it is more convenient, simple and efficacious in reproducing features of PD in animal models [81]. Use of the oral route is primarily intending to test the effect of rotenone on gastric motility and enteric the nervous system as its absorption from the gastrointestinal tract is low and incomplete to produce systemic mitochondrial inhibition [72]. Rotenone was also used to study PD in some in vitro cellular models such as primary mesencephalic cell culture, neuroblastoma cell line (SH-SY5Y) and pheochromocytoma cells (PC12). Apart of motor clinical signs, these rotenone in vitro models of PD presented most of the cellular and molecular pathology that occurs in PD patients, most notably dopaminergic cell loss (Fig. 1) [55] and formation of protein aggregates containing -synuclein [80]. Unlike MPP+ which specifically damages dopaminergic neurons, rotenone was shown to injure other neuronal populations in primary mesencephalic cell culture. Similar results were obtained by our laboratory for some other neurotoxins like domoic acid [56] and acrylamide [57].

Mechanisms of action of rotenone

Complex I inhibition and production of reactive oxygen species

Mitochondria generate ATP via oxidative phosphorylation complexes that are present in their inner membranes. These complexes are known as NADH-ubiquinone oxidoreductase (complex I), succinate dehydrogenase (complex II), ubiquinol-cytochrome c oxidoreductase (complex III), cytochrome c oxidase (complex IV) and ATP synthase (complex V). The process of oxidative phosphorylation is initiated by oxidation of NADH by the complex I enzyme. This results in transferring two electrons, which reduces ubiquinone to ubiquinol. Ubiquinol is re-oxidized by the complex III enzyme and transfers electrons to reduce molecular oxygen to water at complex IV. As a result, the redox energy released during this process is used to transfer protons from the mitochondrial matrix to the periplasmic space that generates proton-motive force across the inner mitochondrial membrane at complex I, III, and IV. This proton-motive force is used by complex V to produce ATP from ADP and inorganic phosphate [64]. Therefore, complex I is the major entry point for electrons to the respiratory chain and the rate-limiting step in overall respiration. However it is still the most complex and least understood component of the mitochondrial oxidative phosphorylation system [63,77].
Inhibition of complex I is the strongest action of the pesticide rotenone [26]. This effect of rotenone dates back to the 1960s [73] when Palmer et al. [53] found that rotenone inhibited electron transfer from the iron-sulfur centres in complex I to ubiquinone leading to blockade of oxidative phosphorylation with limited synthesis of ATP in submitochondrial particles. Singer and Ramsay [68] described two binding sites for rotenone that must be occupied to completely inhibit NADH oxidation: one is located in the corner between the two arms of the l-shaped protein complex [40] and the other is in the hydrophobic domains in the membrane bond arm of the protein which may be responsible for the formation of superoxide radicals. The sensitivity of complex I to rotenone was reported to be different in-between species and ages. In this context, Ueno et al. [75] found that the effect of rotenone is not equal in all species and considerable differences in various taxa are shown. Lenaz et al. [41] reported that mitochondria are not identical and their activity alters during aging. For instance, Genova et al. [15] observed a decrease in complex I sensitivity to rotenone in liver, heart and muscle mitochondria of 24 more than 4-month-old rats.
Rotenone inhibition of electrons transfer from the Fe-S centres in complex I to ubiquinone results in reducing oxidation of NADH and ATP formation [62]. In addition to decreasing ATP production, electrons that leak at complex I can reduce oxygen that was not reduced at complex IV to reactive oxygen species (ROS) such as superoxide and hydrogen peroxide [67]. Elevation of ROS production by rotenone was observed in some experimental models. For instance, Radad et al. [58] found that treatment of primary mesencephalic cell culture with 20 nM of rotenone significantly increased formation of superoxide radical (Fig. 1) and overall ROS (Fig. 1) as measured by dihydroethidium and C-DCDHF-DA fluorescent dyes, respectively. Li et al. [43] reported that rotenone was able to increase ROS production both in isolated mitochondria from HL-60 cells and the cultured HL-60 cells themselves. Besides production of ROS, rotenone was reported to decrease the activities of antioxidant enzymes. In this context, Ojha et al. [52] reported that rotenone administration to rats significantly reduced the activity of superoxide dismutase (SOD) and catalase (CAT), and depleted glutathione (GSH) concentrations. Elevation of ROS production and reducing activity of antioxidant enzymes lead to oxidative stress, the process that mediates most of the rotenone-induced insults.

Inflammatory mechanisms

Inflammation was reported as one of important underlying mechanisms that mediate rotenone-induced damage to neuronal cells. In this context, it was shown that rotenone increased the release of proinflammatory cytokines such as interleukin-1 (IL-1), IL-6 and tumor necrosis factor  (TNF-) in the BV2 cells [44] and in rat’s brain tissues [33,52,65,74]. Microglial activation was reported to play a central pillar in rotenone-induced neuroinflammation. Zaitone et al. [79] found that rotenone upregulates genes encoding CD11bc, a microglial surface antigen, in mice brain. Activated microglia was shown to release cytotoxic inflammatory cytokines such as IL-1 and TNF- [14]. TNF- activates intracellular death signalling pathways such as nuclear factor B (NF-B), c-Jun N-terminal kinase (JNK) and p38 pathways, and increases cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expressions which further amplify inflammatory cascades [28]. Activated microglia was also seen to lead to neuronal damage through phagocytic activities without increasing pro-inflammatory cytokines in primary neuronal/glial cultures prepared from rat cerebella [11]. Prevention of such rotenone-induced neuronal loss by inhibition of microglial phagocytic activities indicates that rotenone neurotoxicity is at least partially mediated by microglial phagocytosis [11]. On the other hand, Klintworth et al. [38] showed that rotenone did not act directly on microglia in cultures prepared from C57BL/6 mice and rotenone-induced microglial activation may occur as a result of neuronal damage or due to releasing of some factors by other neurons or cells.

Microtubules depolarizing effects

Rotenone was shown to depolarize microtubules in some in vitro studies. For instance, Passmore et al. [54] showed that treatment of COS-7 cells with higher rotenone concentrations resulted in a complete depolarization of microtubules. Bisbal et al. [5] reported that rotenone decreased microtubule stability in cultured hippocampal neurons. This depolarizing effect of rotenone on microtubules was suggested to be attributed to binding to tubulin, the protein that polymerizes to microtubules [70]. Protection of microtubules against rotenone-induced depolarization by the microtubule-stabilizing drug taxol confirmed the depolarizing effect of rotenone on microtubules [54,60]. As a microtubule-depolarizing agent, rotenone can result in (1) arresting mitosis and inhibition of cell proliferation [70], (2) inhibition of axon growth by releasing Lfc, a specific GEF for RhoA, from depolarized microtubules [5] and (3) disruption of vesicular transport of dopamine along microtubules leading to accumulation of dopamine in the dopaminergic soma. This can result in increasing oxidative stress due to oxidation of cytosolic dopamine [60]. Moreover, induction of microtubule depolarization in COS-7 cells by rotenone was shown to alter peroxisome morphology and distribution, and on the other hand, treatment of COS-7 cells with rotenone after stabilization of microtubules by paclitaxel resulted in no effects on peroxisome. Peroxisomes are closely linked to the mitochondria and they both maintain a redox-sensitive relationship [54].

Autophagy inhibition

Autophagy is a highly regulated intracellular catabolic process that mediates the degradation of unnecessary materials and dysfunctional organelles in eukaryotic cells [21]. The process of autophagy is essential in maintaining cellular haemostasis and protection against various physiological and pathological stresses, and on the other hand, can lead to some pathological processes [49]. Rotenone was reported to inhibit the autophagy system both in in vitro and in vivo experimental models [32,36]. The raised question now is whether rotenone inhibits autophagy process by decreasing autophagy initiation or autophagy-lysosomal pathway (ALP). The answer to this question is still controversial. Suppression of Beclin 1 expression which acts during initiation of autophagosome formation in rotenone-treated SH-SY5Y neuroblastoma cells indicates that rotenone inhibits autophagy process earlier at the initiation step [32,36]. On the other hand, accumulation of autophagic vacuoles as indicated by increasing the expression of microtubule associated protein-light chain 3-II (LC3-II) in rotenone-treated SH-SY5Y neuroblastoma cells and C57BL/6 mice [82] indicates that rotenone either stimulated autophagic vacuoles formation or decreasing ALP. LC3-II is an accepted and selective marker of autophagic vacuoles. Mader et al. [49] showed that autophagic accumulation by rotenone in SH-SY5Y neuroblastoma cells resulted not from autophagy induction but rather from a block in the lysosomal degradation of autophagic vacuoles. This is because treatment of SH-SY5Y neuroblastoma cells with bafilomycin A1 prior to rotenone resulted in no increase in levels of LC3-II. Bafilomycin A1 is a selective inhibitor of vacuolar-type V-ATPase that completely blocks degradation of autophagic vacuoles through inhibition of autophagic vacuole-lysosome fusion [49].

Rotenone-induced cell death

Apoptotic cell death

The pesticide rotenone was shown to induce apoptotic cell death in some cellular and animal experimental models. For example, Ahmadi et al. [2] reported that exposure of primary dopaminergic neuronal cell culture to rotenone resulted in increasing the number of apoptotic tyrosine hydroxylase positive neurons (TH+) and that was correlated with upregulation of caspase-3 immunoreactivity. Lin et al. [45] showed that chronic rotenone intoxication resulted in apoptotic cell death in rat’s striatum. Using neuroblastoma SH-SY5Y cells, it was shown that rotenone induced apoptotic cell death through activation of caspases [37,48], and p38 and JNK pathways [50], and upregulation of Bax and downregulation of Bcl-2 [10]. In PC12 cells, Hirata et al. [27] showed that rotenone induced apoptosis through activation of JNK and p38 mitogen-activated protein kinase (MAPK). Increasing mitochondrial ROS is implicated in rotenone-induced apoptotic cell death [43]. In this context, the authors showed that rotenone-induced apoptosis was inhibited by antioxidants such as GSH, N-acetylcysteine and vitamin C [43]. Moreover, they observed that HT1080 cells overexpressing magnesium superoxide dismutase were more resistant to rotenone-induced apoptosis than untreated control cells [43].

Necrotic cell death

Not only apoptosis, rotenone was also reported to induce necrotic cell death. In this context, Kamalden et al. [35] found that rotenone induced degeneration of RGC-5 cells by activation of mitogen-activated kinase and not caspase-dependent apoptosis. Hong et al. [30] found that rotenone induced necrotic cell death in PC12 cells. Recently, Callizot et al. [6] have shown that rotenone treatment resulted in necrotic cell death of primary rat dopaminergic neurons at higher concentrations. According to Skulachev [69], rotenone seems to induce necrotic cell death through ATP depletion. Generally, rotenone was reported to cause apoptosis at low concentrations and necrosis at high concentrations [22].

Necroptotic cell death

In addition to apoptotic and necrotic cell death, rotenone was reported to lead to necroptosis in primary rat dopaminergic neurons as measured by the upregulation of RIPK3 after 24 h of exposure [6]. Upregulation of RIPK3 is an indicator of necroptotic cell death [47].

Implication of rotenone in Parkinson disease

For a long time, it has been known that there is an increased risk of PD among people who live in rural areas compared to those who live in cities and this would be attributed to some kind of environmental factors including pesticides [66]. In 2000, successful reproduction of most behavioural, biochemical and pathological features of PD in systemically rotenone-treated rats by Betarbet et al. raised significant concerns about rotenone contribution to PD [3]. Following Betarbet et al.’s study, researchers showed that rotenone can produce a number of pathological processes such as inflammation [46], apoptotic cell death [2], autophagic impairment [32] and microtubule depolarization [54] in rodents similar to those occurring in parkinsonian human brains. Besides CNS pathology, rotenone was also found to cause progressive functional and pathological changes in the enteric nervous system of rodents mimicking changes found in human PD [18]. All together strengthen the association relationship between the pesticide rotenone and PD, and support Greenamyre’s belief that rotenone can produce PD in humans as it did in experimental animals [1]. Supporting animal experiments, some epidemiological studies have linked exposure to pesticides with an increased risk of PD in humans [51]. Of which, Tanner et al. [71] in a case-control study nested in the Agricultural Health Study (AHS) found that PD was positively associated with exposure to two groups of pesticides defined experimentally by impairing mitochondrial function (rotenone) and increasing oxidative stress (paraquat). On the other hand, short environmental half-life and limited bioavailability of rotenone, and its detoxification in the gut by enzymatic, bacterial and hydrolytic reactions make its relationship to PD questionable [23,42].

Conclusions

Rotenone is still used as a non-specific broad-spectrum insecticide and as a piscicide worldwide. Data from experimental and epidemiological studies show a clear association between rotenone exposure and a higher risk of PD. However the causal relationship between rotenone and PD is still questionable. Rotenone in in vitro and in vivo models is an invaluable tool for investigating most molecular and cellular pathology that occur in PD patients and therefore can efficiently test new treatment strategies that would be beneficial for patients with PD.

Acknowledgements

The authors extend their appreciation to the Deanship of Scientific Research at King Khalid University for funding this work through research groups program under grant number R.G.P. 1/125/40.

Disclosure

The authors report no conflict of interest.

References

1. Adam D. Pesticide use linked to Parkinson’s disease. Nature 2000; 408: 125.
2. Ahmadi FA, Linseman DA, Grammatopoulos TN, Jones SM, Bouchard RJ, Freed CR, Heidenreich KA, Zawada WM. The pesticide rotenone induces caspase-3-mediated apoptosis in ventral mesencephalic dopaminergic neurons. J Neurochem 2003; 87: 914-921.
3. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 2000; 3: 1301-1306.
4. Betarbet R, Canet-Aviles RM, Sherer TB, Mastroberardino PG, McLendon C, Kim JH, Lund S, Na HM, Taylor G, Bence NF, Kopito R,Seo BB, Yagi T, Yagi A, Klinefelter G, Cookson MR, Greenamyre JT. Intersecting pathways to neurodegeneration in Parkinson’s disease: effects of the pesticide rotenone on DJ-1, alpha-synuclein, and the ubiquitin-proteasome system. Neurobiol Dis 2006; 22: 404-420.
5. Bisbal M, Remedi M, Quassollo G, Cáceres A, Sanchez M. Rotenone inhibits axonogenesis via an Lfc/RhoA/ROCK pathway in cultured hippocampal neurons. J Neurochem 2018; 146: 570-584.
6. Callizot N, Combes M, Henriques A, Poindron P. Necrosis, apoptosis, necroptosis, three modes of action of dopaminergic neuron neurotoxins. PLoS One 2019; 14: e0215277.
7. Cannon JR, Tapias V, Na HM, Honick AS, Drolet RE, Greenamyre JT. A highly reproducible rotenone model of Parkinson’s disease. Neurobiol Dis 2009; 34: 279-290.
8. Dawson VK, Gingerich WH, Davis RA, Gilderhus PA. Rotenone persistence in freshwater ponds: effects of temperature and sediment adsorption. North Am J Fisheries Manag 1991; 11: 226-231.
9. De Wilde AR, Heyndrickx A, Carton D. A case of fatal rotenone poisoning in a child. J Forensic Sci 1986; 31: 1492-1498.
10. Deng YT, Huang HC, Lin JK. Rotenone induces apoptosis in MCF-7 human breast cancer cell-mediated ROS through JNK and p38 signaling. Mol Carcinog 2010; 49: 141-151.
11. Emmrich JV, Hornik TC, Neher JJ, Brown GC. Rotenone induces neuronal death by microglial phagocytosis of neurons. FEBS J 2013; 280: 5030-5038.
12. Faber K, Augsburger M, Sporkert F, Laffer R, Ceschi A. Rapidly fatal poisoning with an insecticide containing rotenone. Clin Toxicol 2014; 52: 431-432.
13. Ferrante RJ, Browne SE, Shinobu LA, Bowling AC, Baik MJ, MacGarvey U, Kowall NW, Brown RH Jr, Beal MF. Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J Neurochem 1997; 69: 2064-2074.
14. Gao HM, Liu B, Hong JS. Critical role for microglial NADPH oxidase in rotenone-induced degeneration of dopaminergic neurons. J Neurosci 2003; 23: 6181-6187.
15. Genova ML, Bovina C, Marchetti M, Pallotti F, Tietz C, Biagini G, Pugnaloni A, Viticchi C, Gorini A, Villa RF, Lenaz G. Decrease of rotenone inhibition is a sensitive parameter of complex I damage in brain non-synaptic mitochondria of aged rats. FEBS Lett 1997; 410: 467-469.
16. Gilderhus PA, Allen JL, Dawson VK. Persistence of rotenone in ponds at different temperatures. North Am J Fisheries Manag 1986; 6: 129.
17. Gosselin RE, Smith RP, Hodge HC. Clinical toxicology of commercial products. 5th ed. Williams & Wilkins, Baltimore/London 1984; 111-366.
18. Greene JG, Noorian AR, Srinivasan S. Delayed gastric emptying and enteric nervous system dysfunction in the rotenone model of Parkinson’s disease. Exp Neurol 2009; 218: 154-161.
19. Grivennikova VG, Maklashina EO, Gavrikova EV, Vinogradov AD. Interaction of the mitochondrial NADH-ubiquinone reductase with rotenone as related to the enzyme active/inactive transition. Biochim Biophys Acta 1997; 1319: 223-232.
20. Gupta RC, Mitatovic D. Insecticides. In: Biomarkers in toxicology. Elsevier Inc 2014; pp. 389-407.
21. Han B, Wang L, Fu F, Wang Z, Zhang L, Qi GJ, Wang T. Hydroxysafflor yellow A promotes -synuclein clearance via regulating autophagy in rotenone-induced Parkinson’s disease mice. Folia Neuropathol 2018; 56: 133-140.
22. Hartley A, Stone JM, Heron C, Cooper JM, Schapira AH. Complex I inhibitors induce dose-dependent apoptosis in PC12 cells: relevance to Parkinson’s disease. J Neurochem 1994; 63: 1987-1990.
23. Hatcher JM, Pennell KD, Miller GW. Parkinson’s disease and pesticides: a toxicological perspective. Trends Pharmacol Sci 2008; 29: 322-329.
24. Hayes WJ. Pesticides studied in man. Williams & Wilkins, Baltimore/London 1982; pp. 81-86.
25. Heikkila RE, Nicklas WJ, Vyas I, Duvoisin RC. Dopaminergic toxicity of rotenone and the 1-methyl-4-phenylpyridinium ion after their stereotaxic administration to rats: implication for the mechanism of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity. Neurosci Lett 1985; 62: 389-394.
26. Heinz S, Freyberger A, Lawrenz B, Schladt L, Schmuck G, Ellinger-Ziegelbauer H. Mechanistic Investigations of the Mitochondrial Complex I Inhibitor Rotenone in the Context of Pharmacological and Safety Evaluation. Sci Rep 2017; 7: 45465.
27. Hirata Y, Meguro T, Kiuchi, K. Differential effect of nerve growth factor on dopaminergic neurotoxin-induced apoptosis, J Neurochem 2006; 99: 416-425.
28. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol 2009; 8: 382-397.
29. Holland EA. Suicide by ingestion of derris root sp in New Ireland. Trans R Soc Trop Med Hyg 1938; 32: 293-294.
30. Hong Y, Nie H, Wu D, Wei X, Ding X, Ying W. NAD(+) treatment prevents rotenone-induced apoptosis and necrosis of differentiated PC12 cells. Neurosci Lett 2014; 560: 46-50.
31. Islam MB. Botanical insecticides, deterrents, and repellents in modern agriculture and an increasingly regulated world. Annu Rev Entomol 2006; 51: 45-66.
32. Jang W, Kim HJ, Li H, Jo KD, Lee MK, Yang HO. The Neuroprotective effect of erythropoietin on rotenone-induced neurotoxicity in SH-SY5Y Cells through the induction of autophagy. Mol Neurobiol 2016; 53: 3812-3821.
33. Javed H, Azimullah S, Abul Khair SB, Ojha S, Haque ME. Neuroprotective effect of nerolidol against neuroinflammation and oxidative stress induced by rotenone. BMC Neurosci 2016; 17: 58.
34. Johnson ME, Stringer A, Bobrovskaya L. Rotenone induces gastrointestinal pathology and microbiota alterations in a rat model of Parkinson’s disease. Neurotoxicology 2018; 65: 174-185.
35. Kamalden TA, Ji D, Osborne NN. Rotenone-induced death of RGC-5 cells is caspase independent, involves the JNK and p38 pathways and is attenuated by specific green tea flavonoids. Neurochem Res 2012; 37: 1091-1101.
36. Kang SY, Lee SB, Kim HJ, Kim HT, Yang HO, Jang W. Autophagic modulation by rosuvastatin prevents rotenone-induced neurotoxicity in an in vitro model of Parkinson’s disease. Neurosci Lett 2017; 642: 20-26.
37. Kitamura Y, Inden M, Miyamura A, Kakimura J, Taniguchi T, Shimohama S. Possible involvement of both mitochondria- and endoplasmic reticulum-dependent caspase pathways in rotenone-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neurosci Lett 2002; 333: 25-28.
38. Klintworth H, Garden G, Xia Z. Rotenone and paraquat do not directly activate microglia or induce inflammatory cytokine release. Neurosci Lett 2009; 462: 1-5.
39. La Forge FB, Haller HL, Smith LE. The Determination of the structure of rotenone. Chem Rev 1933; 18: 181-213.
40. Lambert AJ, Brand MD. Inhibitors of the quinone-binding site allow rapid superoxide production from mitochondrial NADH:ubiquinone oxidoreductase (complex I). J Biol Chem 2004; 279: 39414-39420.
41. Lenaz G, Bovina C, Castelluccio C, Fato R, Formiggini G, Genova ML, Marchetti M, Pich MMe, Pallotti F, Castelli GP, Biagini G. Mitochondrial complex I defects in aging. Molecular and Cellular Biochemistry 174; 1997: 329-333.
42. Li AA, Mink PJ, McIntosh LJ, Teta MJ, Finley B. Evaluation of epidemiologic and animal data associating pesticides with Parkinson’s disease. J Occup Environ Med 2005; 47: 1059-1087.
43. Li N, Ragheb K, Lawler G, Sturgis J, Rajwa B, Melendez JA, Robinson JP. Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive oxygen species production. J Biol Chem 2003; 278: 8516-8525.
44. Liang Y, Zhou T, Chen Y, Lin D, Jing X, Peng S, Zheng D, Zeng Z, Lei M, Wu X, Huang K, Yang L, Xiao S, Liu J, Tao E. Rifampicin inhibits rotenone-induced microglial inflammation via enhancement of autophagy. Neurotoxicology 2017; 63: 137-145.
45. Lin TK, Cheng CH, Chen SD, Liou CW, Huang CR, Chuang YC. Mitochondrial dysfunction and oxidative stress promote apoptotic cell death in the striatum via cytochrome c/caspase-3 signaling cascade following chronic rotenone intoxication in rats. Int J Mol Sci 2012; 13: 8722-8739.
46. Ling N. Rotenone a review of its toxicity and use for fisheries management. Sci Conservation 2002; 211: 40.
47. Liu S, Li Y, Choi HMC, Sarkar C, Koh EY, Wu J, Lipinski MM. Lysosomal damage after spinal cord injury causes accumulation of RIPK1 and RIPK3 proteins and potentiation of necroptosis. Cell Death Dis 2018; 9: 476.
48. Ma J, Gao SS, Yang HJ, Wang M, Cheng BF, Feng ZW, Wang L. neuroprotective effects of proanthocyanidins, natural flavonoids derived from plants, on rotenone-induced oxidative stress and apoptotic cell death in human neuroblastoma SH-SY5Y cells. Front Neurosci 2018; 31: 369.
49. Mader BJ, Pivtoraiko VN, Flippo HM, Klocke BJ, Roth KA, Mangieri LR, Shacka JJ. Rotenone inhibits autophagic flux prior to inducing cell death. ACS Chem Neurosci 2012; 3: 1063-1072.
50. Newhouse K, Hsuan SL, Chang SH, Cai B, Wang Y, Xia Z. Rotenone-induced apoptosis is mediated by p38 and JNK MAP kinases in human dopaminergic SH-SY5Y cells. Toxicol Sci 2004; 79: 137-1346.
51. Nisticò R, Mehdawy B, Piccirilli S, Mercuri N. Paraquat- and rotenone-induced models of Parkinson’s disease. Int J Immunopathol Pharmacol 2011; 24: 313-322.
52. Ojha S, Javed H, Azimullah S, Abul Khair SB, Haque ME. Glycyrrhizic acid attenuates neuroinflammation and oxidative stress in rotenone model of Parkinson’s disease. Neurotox Res 2016; 29: 275-287.
53. Palmer G, Horgan DJ, Tisdale H, Singer TP, Beinert H. Studies on the respiratory chain-linked reduced nicotinamide adenine dinucleotide dehydrogenase. XIV. Location of the sites of inhibition of rotenone, barbiturates, and piericidin by means of electron paramagnetic resonance spectroscopy. J Biol Chem 1968; 243: 844-847.
54. Passmore JB, Pinho S, Gomez-Lazaro M, Schrader M. The respiratory chain inhibitor rotenone affects peroxisomal dynamics via its microtubule-destabilizing activity. Histochem Cell Biol 2017; 148: 331-341.
55. Radad K, Moldzio R, Rausch WD. Rapamycin protects dopaminergic neurons against rotenone-induced cell death in primarymesencephalic cell culture. Folia Neuropathol 2015; 53: 250-261.
56. Radad K, Al-Shraim M, Al-Emam A, Moldzio R, Rausch WD. Neurotoxic effects of domoic acid on dopaminergic neurons in primary mesencephalic cell culture. Folia Neuropathol 2018; 56: 39-48.
57. Khaled Radad, Mubarak Al-Shraim, Ahmed Al-Emam, Rudolf Moldzio, Rausch WD. Neurotoxic effects of acrylamide on dopaminergic neurons in primary mesencephalic cell culture. Folia Neuropathol 2019; 57: 196-204.
58. Radad K, Rausch WD, Gille G. Rotenone induces cell death in primary dopaminergic culture by increasing ROS production and inhibiting mitochondrial respiration. Neurochem Int 2006; 49: 379-386.
59. Rasheed MZ, Andrabi SS, Salman M, Tabassum H, Shaquiquzzaman M, Parveen S, Parvez S. Melatonin improves behavioral and biochemical outcomes in a rotenone-induced rat model of Parkinson’s disease. J Environ Pathol Toxicol Oncol 2018; 37: 139-150.
60. Ren Y, Liu W, Jiang H, Jiang Q, Feng J. Selective vulnerability of dopaminergic neurons to microtubule depolymerization. J Biol Chem 2005; 280: 34105-34112.
61. Robertson DR, Smith-Vaniz WF. Rotenone: an essential but demonized tool for assessing marine fish diversity. BioScience 2008; 58: 165-170.
62. Schuler F, Casida JE. The insecticide target in the PSST subunit of complex I. Pest Manag Sci 2001; 57: 932-940.
63. Sharma LK, Lu J, Bai Y. Mitochondrial respiratory complex I: structure, function and implication in human diseases. Curr Med Chem 2009; 16: 1266-1277.
64. Sharma N, Jamwal S, Kumar P. Beneficial effect of antidepressants against rotenone induced Parkinsonism like symptoms in rats. Pathophysiology 2016; 23: 123-134.
65. Sharma S, Kumar P, Deshmukh R. Neuroprotective potential of spermidine against rotenone induced Parkinson’s disease in rats. Neurochem Int 2018; 116: 104-111.
66. Sherer T, Testa CM, Seo BB, Richardson JR, Kim JH, Miller GW, Yagi T, Matsun-Yagi A, Greenamyre T. Mechanism of toxicity in rotenone models of Parkinson’s disease. J Neurosci 2003; 34: 10756-10764.
67. Sherer TB, Trimmer PA, Borland K, Parks JK, Bennett JP Jr, Tuttle JB. Chronic reduction in complex I function alters calcium signaling in SH-SY5Y neuroblastoma cells. Brain Res 2001; 891: 94-105.
68. Singer TP, Ramsay RR. The reaction sites of rotenone and ubiquinone with mitochondrial NADH dehydrogenase. Biochim Biophys Acta 1994; 1187: 198-202.
69. Skulachev VP. Bioenergetic aspects of apoptosis, necrosis and mitoptosis. Apoptosis 2006; 11: 473-485.
70. Srivastava P, Panda D. Rotenone inhibits mammalian cell proliferation by inhibiting microtubule assembly through tubulin binding. FEBS J 2007; 274: 4788-4801.
71. Tanner CM, Kamel F, Ross GW, Hoppin JA, Goldman SM, Korell M, Marras C, Bhudhikanok GS, Kasten M, Chade AR, Comyns K, Richards MB, Meng C, Priestley B, Fernandez HH, Cambi F, Umbach DM, Blair A, Sandler DP, Langston JW. Rotenone, paraquat, and Parkinson’s disease. Environ Health Perspect 2011; 119: 866-872.
72. Tasselli M, Chaumette T, Paillusson S, Monnet Y, Lafoux A, Huchet-Cadiou C, Aubert P, Hunot S, Derkinderen P, Neunlist M. Effects of oral administration of rotenone on gastrointestinal functions in mice. Neurogastroenterol Motil 2013; 25: e183-193.
73. Teeter ME, Baginsky ML, Hatefi Y. Ectopic inhibition of the complexes of the electron transport system by rotenone, piericidin A, demerol and antimycin A. Biochim Biophys Acta 1969; 172: 331-333.
74. Thakur P, Nehru B. Anti-inflammatory properties rather than anti-oxidant capability is the major mechanism of neuroprotection by sodium salicylate in a chronic rotenone model of Parkinson’s disease. Neuroscience 2013; 231: 420-431.
75. Ueno H, Miyoshi H, Inoue M, Niidome Y, Iwamura H. Structural factors of rotenone required for inhibition of various NADH-ubiquinone oxidoreductases. Biochim Biophys Acta 1996; 1276: 195-202.
76. US EPA. Re-registration eligibility decision for rotenone. US EPA Case #0255, 2007.
77. Wirth C, Brandt U, Hunte C, Zickermann V. Structure and function of mitochondrial complex I. Biochim Biophys Acta 2016; 1857: 902-914.
78. Wood DM, Alsahaf H, Streete P, Dargan PI, Jones AL. Fatality after deliberate ingestion of the pesticide rotenone: a case report. Crit Care 2005; 9: 280-284.
79. Zaitone SA, Ahmed E, Elsherbiny NM, Mehanna ET, El-Kherbetawy MK, ElSayed MH, Alshareef DM, Moustafa YM. Caffeic acid improves locomotor activity and lessens inflammatory burden in a mouse model of rotenone-induced nigral neurodegeneration: Relevance to Parkinson’s disease therapy. Pharmacol Rep 2018; 71: 32-41.
80. Zhang M, Deng YN, Zhang JY, Liu J, Li YB, Su H, Qu QM. SIRT3 Protects rotenone-induced injury in SH-SY5Y cells by promoting autophagy through the LKB1-AMPK-mTOR pathway. Aging Dis 2018; 9: 273-286.
81. Zhang ZN, Zhang JS, Xiang J, Yu ZH, Zhang W, Cai M, Li XT, Wu T, Li WW, Cai DF. Subcutaneous rotenone rat model of Parkinson’s disease: dose exploration study. Brain Res 2017; 1655: 104-113.
82. Zhou Q, Chen B, Wang X, Wu L, Yang Y, Cheng X, Hu Z, Cai X, Yang J, Sun X, Lu W, Yan H, Chen J, Ye J, Shen J, Cao P. Sulforaphane protects against rotenone-induced neurotoxicity in vivo: Involvement of the mTOR, Nrf2, and autophagy pathways. Sci Rep 2016; 24: 32206.
Copyright: © 2019 Mossakowski Medical Research Centre Polish Academy of Sciences and the Polish Association of Neuropathologists. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.