1
|
Coppola L, Lori G, Tait S, Sogorb MA, Estevan C. Evaluation of developmental toxicity of chlorpyrifos through new approach methodologies: a systematic review. Arch Toxicol 2025; 99:935-981. [PMID: 39869190 PMCID: PMC11821739 DOI: 10.1007/s00204-024-03945-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025]
Abstract
Chlorpyrifos (CPF) is an organophosphorus pesticide of concern because many in vivo animal studies have demonstrated developmental toxicity exerted by this substance; however, despite its widespread use, evidence from epidemiological studies is still limited. In this study, we have collected all the information generated in the twenty-first century on the developmental toxicity of CPF using new approach methodologies. We have critically evaluated and integrated information coming from 70 papers considering human, rodent, avian and fish models. The comparison of the collected evidence with available adverse outcome pathways allows us to conclude that adverse outcomes observed in animals, such as memory and learning impairments as well as reduction in cognitive function, could involve several mechanisms of action including inhibition of acetylcholinesterase, overactivation of glutamate receptors and activation of mitogen-activated protein kinase, extracellular signal-regulated kinase 1/2, followed by both disruption of neurotransmitter release and increase in oxidative stress and apoptosis.
Collapse
Affiliation(s)
- L Coppola
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - G Lori
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - S Tait
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - M A Sogorb
- Bioengineering Institute, Miguel Hernández de Elche University, Elche, Spain
| | - C Estevan
- Applied Biology Department, Miguel Hernández de Elche University, Elche, Spain.
| |
Collapse
|
2
|
Li Y, Gao H. Investigation and validation of neurotransmitter receptor-related biomarkers for forecasting clinical outcomes and immunotherapeutic efficacy in breast cancer. Gene 2025; 937:149135. [PMID: 39615806 DOI: 10.1016/j.gene.2024.149135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE The prognostic role of neurotransmitters and their receptors in breast cancer (BC) has not been fully investigated. The aim of this study was to construct a survival model for the prognosis of BC patients based on neurotransmitter receptor-related genes (NRRGs). METHODS BC-related differentially expressed genes (DEGs) were screened and intersected with NRRGs. GO, KEGG and PPI analyses were performed. Univariate Cox, Least Absolute Shrinkage and Selection Operator (LASSO) and multivariate Cox regression analyses were used to construct prognostic models for biomarker expression levels. The model was validated using an external validation set. The receiver operating characteristic curves (ROC) for diagnostic value prediction and clinicopathologic characteristic nomogram were constructed. qRT-PCR was used for further in vitro validation experiments. RESULTS Forty-five overlapping genes were obtained by intersecting BC-related DEGs with 172 NRRGs. Univariate Cox, LASSO and multivariate Cox regression analyses were used to construct prognostic models for the expression levels of biomarkers including DLG3, SLC1A1, PSCA and PRKCZ. The feasibility of the model was validated by the GEO validation set. ROC curves were established for diagnostic value prediction. Patients in the high-risk group had a worse prognosis, higher TMB score, higher probability of gene mutation, and higher immune cell infiltration. RiskScore, M, N and Age were strongly correlated with survival. The mRNA expression levels of DLG3, PSCA and PRKCZ in the BC group were significantly higher than those in the control group. CONCLUSION Risk prediction model based on DLG3, SLC1A1, PSCA and PRKCZ, which are closely related to BC prognosis, was successfully constructed.
Collapse
Affiliation(s)
- Yili Li
- Department of Mammary Gland, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Mammary Gland, Chongqing Health Center for Women and Children, Chongqing, China
| | - Han Gao
- Department of Mammary Gland, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Mammary Gland, Chongqing Health Center for Women and Children, Chongqing, China.
| |
Collapse
|
3
|
Rimawi I, Yanai S, Turgeman G, Yanai J. Whole transcriptome analysis in offspring whose fathers were exposed to a developmental insult: a novel avian model. Sci Rep 2023; 13:16499. [PMID: 37779136 PMCID: PMC10543553 DOI: 10.1038/s41598-023-43593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023] Open
Abstract
Although the effects of paternal exposure to insults on the offspring received limited attention in the past, it is currently gaining interest especially after understanding the mechanisms which may mediate such exposure effects. In the current study, the well-controlled avian model (Fayoumi) was utilized to investigate the effects of paternal exposure to the developmental insult, chlorpyrifos on the offspring's gene expression via mRNA and small RNA sequencing. Numerous mRNA gene expression changes were detected in the offspring after paternal exposure to the developmental insult, especially in genes related to neurogenesis, learning and memory. qPCR analysis of several genes, that were significantly changed in mRNA sequencing, confirmed the results obtained in mRNA sequencing. On the other hand, small RNA sequencing did not identify significant microRNA genes expression changes in the offspring after paternal exposure to the developmental insult. The effects of the paternal exposure were more pronounced in the female offspring compared to the male offspring. The results identified expression alterations in major genes (some of which were pertinent to the functional changes observed in other forms of early developmental exposure) after paternal insult exposure and provided a direction for future studies involving the most affected genes.
Collapse
Affiliation(s)
- Issam Rimawi
- The Ross Laboratory for Studies in Neural Birth Defects, Department of Medical Neurobiology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Sunny Yanai
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gadi Turgeman
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Joseph Yanai
- The Ross Laboratory for Studies in Neural Birth Defects, Department of Medical Neurobiology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel.
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
4
|
Anyachor CP, Dooka DB, Orish CN, Amadi CN, Bocca B, Ruggieri F, Senofonte M, Frazzoli C, Orisakwe OE. Mechanistic considerations and biomarkers level in nickel-induced neurodegenerative diseases: An updated systematic review. IBRO Neurosci Rep 2022; 13:136-146. [PMID: 35989698 PMCID: PMC9382260 DOI: 10.1016/j.ibneur.2022.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/30/2022] [Indexed: 10/27/2022] Open
Abstract
The environment has been implicated to be a strong determinant of brain health with higher risk of neurodegeneration. The drastic rise in the prevalence of neurodegenerative diseases (NDDs) including Alzheimer's disease (AD), Parkinson's disease (PD), autism spectrum disorder (ASD), multiple sclerosis (MS) etc., supports the idea that environmental factors may play a major role in NDDs aetiology. Nickel is one of the listed environmental metals reported to pose a serious threat to human health. This paper reported available studies on nickel level in NDDs covering both animal and human studies. Different databases were searched for articles reporting the main neurotoxicity mechanisms and the concentration of nickel in fluids and tissues of NDDs patients compared to controls. Data were extracted and synthesized by ensuring the articles were related to nickel and NDDs. Various mechanisms were reported as oxidative stress, disturbances in mitochondrial membrane potential, trace elements homeostasis destabilization, etc. Nickel was found elevated in biological fluids as blood, serum/plasma and CSF and in the brain of NDDs, as a consequence of unintentional exposure thorough nickel-contaminated air, food, water, and skin contact. In addition, after exposure to nickel, the concentration of markers of lipid peroxidation were increased, while some antioxidant defence systems decreased. Thus, the reduction in the exposure to nickel contaminant may hold a promise in reducing the incidence of NDDs.
Collapse
Affiliation(s)
- Chidinma Promise Anyachor
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Donatus Baridoo Dooka
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Chinna Nneka Orish
- Department of Anatomy, College of Health Sciences University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Cecilia Nwadiuto Amadi
- Department of Experimental Pharmacology & Toxicology, Faculty of Pharmacy, University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Beatrice Bocca
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Flavia Ruggieri
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Marta Senofonte
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Frazzoli
- Department for Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, Rome Viale Regina Elena, 29900161 Roma, Italy
| | - Orish E. Orisakwe
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
- Department of Anatomy, College of Health Sciences University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| |
Collapse
|
5
|
Modulation of CREB and its associated upstream signaling pathways in pesticide-induced neurotoxicity. Mol Cell Biochem 2022; 477:2581-2593. [PMID: 35596844 PMCID: PMC9618525 DOI: 10.1007/s11010-022-04472-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/04/2022] [Indexed: 11/13/2022]
Abstract
Human beings are exposed to various environmental xenobiotics throughout their life consisting of a broad range of physical and chemical agents that impart bodily harm. Among these, pesticide exposure that destroys insects mainly by damaging their central nervous system also exerts neurotoxic effects on humans and is implicated in the etiology of several degenerative disorders. The connectivity between CREB (cAMP Response Element Binding Protein) signaling activation and neuronal activity is of broad interest and has been thoroughly studied in various diseased states. Several genes, as well as protein kinases, are involved in the phosphorylation of CREB, including BDNF (Brain-derived neurotrophic factor), Pi3K (phosphoinositide 3-kinase), AKT (Protein kinase B), RAS (Rat Sarcoma), MEK (Mitogen-activated protein kinase), PLC (Phospholipase C), and PKC (Protein kinase C) that play an essential role in neuronal plasticity, long-term potentiation, neuronal survival, learning, and memory formation, cognitive function, synaptic transmission, and suppressing apoptosis. These elements, either singularly or in a cascade, can result in the modulation of CREB, making it a vulnerable target for various neurotoxic agents, including pesticides. This review provides insight into how these various intracellular signaling pathways converge to bring about CREB activation and how the activated or deactivated CREB levels can affect the gene expression of the upstream molecules. We also discuss the various target genes within the cascade vulnerable to different types of pesticides. Thus, this review will facilitate future investigations associated with pesticide neurotoxicity and identify valuable therapeutic targets.
Collapse
|
6
|
Nisa FY, Rahman MA, Hossen MA, Khan MF, Khan MAN, Majid M, Sultana F, Haque MA. Role of neurotoxicants in the pathogenesis of Alzheimer's disease: a mechanistic insight. Ann Med 2021; 53:1476-1501. [PMID: 34433343 PMCID: PMC8405119 DOI: 10.1080/07853890.2021.1966088] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most conspicuous chronic neurodegenerative syndrome, which has become a significant challenge for the global healthcare system. Multiple studies have corroborated a clear association of neurotoxicants with AD pathogenicity, such as Amyloid beta (Aβ) proteins and neurofibrillary tangles (NFTs), signalling pathway modifications, cellular stress, cognitive dysfunctions, neuronal apoptosis, neuroinflammation, epigenetic modification, and so on. This review, therefore, aimed to address several essential mechanisms and signalling cascades, including Wnt (wingless and int.) signalling pathway, autophagy, mammalian target of rapamycin (mTOR), protein kinase C (PKC) signalling cascades, cellular redox status, energy metabolism, glutamatergic neurotransmissions, immune cell stimulations (e.g. microglia, astrocytes) as well as an amyloid precursor protein (APP), presenilin-1 (PSEN1), presenilin-2 (PSEN2) and other AD-related gene expressions that have been pretentious and modulated by the various neurotoxicants. This review concluded that neurotoxicants play a momentous role in developing AD through modulating various signalling cascades. Nevertheless, comprehension of this risk agent-induced neurotoxicity is far too little. More in-depth epidemiological and systematic investigations are needed to understand the potential mechanisms better to address these neurotoxicants and improve approaches to their risk exposure that aid in AD pathogenesis.Key messagesInevitable cascade mechanisms of how Alzheimer's Disease-related (AD-related) gene expressions are modulated by neurotoxicants have been discussed.Involvement of the neurotoxicants-induced pathways caused an extended risk of AD is explicited.Integration of cell culture, animals and population-based analysis on the clinical severity of AD is addressed.
Collapse
Affiliation(s)
- Fatema Yasmin Nisa
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Atiar Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Amjad Hossen
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Mohammad Forhad Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Md. Asif Nadim Khan
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Mumtahina Majid
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Farjana Sultana
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Moyano P, Garcia JM, García J, Pelayo A, Muñoz-Calero P, Frejo MT, Flores A, Del Pino J. Aryl Hydrocarbon Receptor Activation Produces Heat Shock Protein 90 and 70 Overexpression, Prostaglandin E2/Wnt/β-Catenin Signaling Disruption, and Cell Proliferation in MCF-7 and MDA-MB-231 Cells after 24 h and 14 Days of Chlorpyrifos Treatment. Chem Res Toxicol 2021; 34:2019-2023. [PMID: 34424684 PMCID: PMC9132385 DOI: 10.1021/acs.chemrestox.1c00258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The biocide chlorpyrifos (CPF) was
described to increase breast
cancer risk in humans, to produce breast cancer in animals, and to
induce cell proliferation in MCF-7 and MDA-MB-231 cells after 1 and
14 days of treatment. The entire mechanisms related to these CPF actions
remain unknown. CPF induced cell proliferation in MCF-7 and MDA-MB-231
cells after 1 and 14 days of treatment by AhR activation through the
PGE2/Wnt/β-catenin pathway and HSP90 and HSP70 overexpression.
Our results reveal new information on CPF toxic mechanisms induced
in human breast cancer cell lines, which could assist in elucidating
its involvement in breast cancer.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - José Manuel Garcia
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jimena García
- Department of Pharmacology, Health Sciences School, Alfonso X University, 28691 Madrid, Spain
| | - Adela Pelayo
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Pilar Muñoz-Calero
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28040 Madrid, Spain
| | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Andrea Flores
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
8
|
Giambò F, Leone GM, Gattuso G, Rizzo R, Cosentino A, Cinà D, Teodoro M, Costa C, Tsatsakis A, Fenga C, Falzone L. Genetic and Epigenetic Alterations Induced by Pesticide Exposure: Integrated Analysis of Gene Expression, microRNA Expression, and DNA Methylation Datasets. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18168697. [PMID: 34444445 PMCID: PMC8394939 DOI: 10.3390/ijerph18168697] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
Environmental or occupational exposure to pesticides is considered one of the main risk factors for the development of various diseases. Behind the development of pesticide-associated pathologies, there are both genetic and epigenetic alterations, where these latter are mainly represented by the alteration in the expression levels of microRNAs and by the change in the methylation status of the DNA. At present, no studies have comprehensively evaluated the genetic and epigenetic alterations induced by pesticides; therefore, the aim of the present study was to identify modifications in gene miRNA expression and DNA methylation useful for the prediction of pesticide exposure. For this purpose, an integrated analysis of gene expression, microRNA expression, and DNA methylation datasets obtained from the GEO DataSets database was performed to identify putative genes, microRNAs, and DNA methylation hotspots associated with pesticide exposure and responsible for the development of different diseases. In addition, DIANA-miRPath, STRING, and GO Panther prediction tools were used to establish the functional role of the putative biomarkers identified. The results obtained demonstrated that pesticides can modulate the expression levels of different genes and induce different epigenetic alterations in the expression levels of miRNAs and in the modulation of DNA methylation status.
Collapse
Affiliation(s)
- Federica Giambò
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Gian Marco Leone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.M.L.); (G.G.); (R.R.); (A.C.)
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.M.L.); (G.G.); (R.R.); (A.C.)
| | - Roberta Rizzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.M.L.); (G.G.); (R.R.); (A.C.)
| | - Alessia Cosentino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.M.L.); (G.G.); (R.R.); (A.C.)
| | - Diana Cinà
- Health Management of the “Cannizzaro” Emergency Hospital of Catania, 95126 Catania, Italy;
- Clinical Pathology and Clinical Molecular Biology Unit, “Garibaldi Centro” Hospital, ARNAS Garibaldi, 95123 Catania, Italy
| | - Michele Teodoro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Occupational Medicine Section, University of Messina, 98125 Messina, Italy; (M.T.); (C.F.)
| | - Chiara Costa
- Clinical and Experimental Medicine Department, University of Messina, 98125 Messina, Italy;
| | - Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Concettina Fenga
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Occupational Medicine Section, University of Messina, 98125 Messina, Italy; (M.T.); (C.F.)
| | - Luca Falzone
- Epidemiology and Biostatistics Unit, National Cancer Institute-IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-095-478-1278
| |
Collapse
|
9
|
Moyano P, García JM, García J, Pelayo A, Muñoz-Calero P, Frejo MT, Anadon MJ, Naval MV, Flores A, Mirat VA, Del Pino J. Chlorpyrifos induces cell proliferation in MCF-7 and MDA-MB-231 cells, through cholinergic and Wnt/β-catenin signaling disruption, AChE-R upregulation and oxidative stress generation after single and repeated treatment. Food Chem Toxicol 2021; 152:112241. [PMID: 33930485 DOI: 10.1016/j.fct.2021.112241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/18/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
Chlorpyrifos (CPF) biocide, is associated with breast cancer. The processes underlying this association have not been elucidated to date. CPF increases MCF-7 and MDA-MB-231 cell proliferation after acute and long-term treatment, partially through KIAA1363 overexpression and aryl-hydrocarbon receptor activation but also through estrogen receptor-alpha activation after 24 h exposure in MCF-7 cells, suggesting other mechanisms may be involved. CPF induces reactive oxygen species (ROS) generation, acetylcholine accumulation, and overexpression of acetylcholinesterase-R/S (AChE-R/S) variants, while it also alters the Wnt/β-catenin pathway, both in vitro and in vivo, in processes different from cancer. These latter mechanisms are also linked to cell proliferation and could mediate this effect induced by CPF. Our results show that CPF (0.01-100 μM), following one-day and fourteen-days treatment, respectively, induced ROS generation and lipid peroxidation, and acetylcholine accumulation due to AChE inhibition, Wnt/β-catenin up- or downregulation depending on the CPF treatment concentration, and AChE-R and AChE-S overexpression, with the latter being mediated through GSK-3β activity alteration. Finally, CPF promoted cell division through ACh and ROS accumulation, AChE-R overexpression, and Wnt/β-catenin signaling disruption. Our results provide novel information on the effect of CPF on human breast cancer cell lines that may help to explain its involvement in breast cancer.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain
| | - José Manuel García
- Department of Pharmacology and Toxicology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Jimena García
- Department of Pharmacology, Health Sciences School, Alfonso X University, 28691, Madrid, Spain
| | - Adela Pelayo
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain
| | | | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Maria Jose Anadon
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Maria Victoria Naval
- Department of Pharmacology, Pharmacognosy and Botany, Pharmacy School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Andrea Flores
- Department of Pharmacology and Toxicology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Vega Alejandra Mirat
- Department of Pharmacology and Toxicology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Medicine School, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
10
|
|
11
|
Gestational exposures to organophosphorus insecticides: From acute poisoning to developmental neurotoxicity. Neuropharmacology 2020; 180:108271. [PMID: 32814088 DOI: 10.1016/j.neuropharm.2020.108271] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/03/2020] [Accepted: 08/10/2020] [Indexed: 11/22/2022]
Abstract
For over three-quarters of a century, organophosphorus (OP) insecticides have been ubiquitously used in agricultural, residential, and commercial settings and in public health programs to mitigate insect-borne diseases. Their broad-spectrum insecticidal effectiveness is accounted for by the irreversible inhibition of acetylcholinesterase (AChE), the enzyme that catalyzes acetylcholine (ACh) hydrolysis, in the nervous system of insects. However, because AChE is evolutionarily conserved, OP insecticides are also toxic to mammals, including humans, and acute OP intoxication remains a major public health concern in countries where OP insecticide usage is poorly regulated. Environmental exposures to OP levels that are generally too low to cause marked inhibition of AChE and to trigger acute signs of intoxication, on the other hand, represent an insidious public health issue worldwide. Gestational exposures to OP insecticides are particularly concerning because of the exquisite sensitivity of the developing brain to these insecticides. The present article overviews and discusses: (i) the health effects and therapeutic management of acute OP poisoning during pregnancy, (ii) epidemiological studies examining associations between environmental OP exposures during gestation and health outcomes of offspring, (iii) preclinical evidence that OP insecticides are developmental neurotoxicants, and (iv) potential mechanisms underlying the developmental neurotoxicity of OP insecticides. Understanding how gestational exposures to different levels of OP insecticides affect pregnancy and childhood development is critical to guiding implementation of preventive measures and direct research aimed at identifying effective therapeutic interventions that can limit the negative impact of these exposures on public health.
Collapse
|
12
|
Hayman NT, Hentschel BT, Renick VC, Anderson TW. Combined effects of flow speed and sub-lethal insecticide exposure on predator-prey interactions between the California killifish and an infaunal polychaete. ECOTOXICOLOGY (LONDON, ENGLAND) 2019; 28:117-131. [PMID: 30547329 DOI: 10.1007/s10646-018-2005-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Hydrodynamics and pollution affect estuarine populations, but their ecological effects have rarely been studied in combination. We conducted two laboratory experiments to quantify whether predator-prey interactions between California killifish, Fundulus parvipinnis, and the polychaete Polydora cornuta vary with flow speed and chlorpyrifos exposure. In one experiment, only F. parvipinnis was exposed to chlorpyrifos; in the other, only P. cornuta was exposed. The flume included a 300-cm2 area of sediment with 24 P. cornuta in a central patch (98 cm2). We videotaped groups of three killifish for 50 min at one of four flow speeds (6, 9, 12, or 15 cm/s) and recorded the proportion of bites directed at the prey patch. Unexposed killifish directed 70% of their bites at the prey patch at 6 cm/s, and prey-patch selection decreased as flow increased. Killifish exposed to chlorpyrifos directed 41% of their bites at the prey patch at 6 cm/s with reduced prey-patch selection relative to unexposed fish at 9 and 12 cm/s. At 15 cm/s, both exposed and unexposed fish displayed non-selective biting. Worms were videotaped to quantify their deposit- and suspension-feeding activities. Exposing worms to chlorpyrifos reduced total feeding activity by ~30%. Suspension feeding was more common at faster flow speeds, but the time worms spent suspension feeding relative to deposit feeding was unaffected by chlorpyrifos. No behavioral changes were noted in either species when the other was exposed to chlorpyrifos. This study highlights how hydrodynamic conditions can alter the relative importance of a toxicant's effects on predator-prey interactions.
Collapse
Affiliation(s)
- Nicholas T Hayman
- Department of Biology and Coastal and Marine Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182-4614, USA.
| | - Brian T Hentschel
- Department of Biology and Coastal and Marine Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182-4614, USA
| | - Violet C Renick
- Department of Biology and Coastal and Marine Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182-4614, USA
| | - Todd W Anderson
- Department of Biology and Coastal and Marine Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182-4614, USA
| |
Collapse
|
13
|
Lamtai M, Chaibat J, Ouakki S, Zghari O, Mesfioui A, El Hessni A, Rifi EH, Marmouzi I, Essamri A, Ouichou A. Effect of Chronic Administration of Nickel on Affective and Cognitive Behavior in Male and Female Rats: Possible Implication of Oxidative Stress Pathway. Brain Sci 2018; 8:brainsci8080141. [PMID: 30065183 PMCID: PMC6119950 DOI: 10.3390/brainsci8080141] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/22/2018] [Accepted: 07/02/2018] [Indexed: 01/24/2023] Open
Abstract
Nickel (Ni) toxicity has been reported to produce biochemical and behavioral dysfunction. The present study was undertaken to examine whether Ni chronic administration can induce alterations of affective and cognitive behavior and oxidative stress in male and female rats. Twenty-four rats, for each gender, divided into control and three test groups (n = 6), were injected intraperitoneally with saline (0.9% NaCl) or NiCl2 (0.25 mg/kg, 0.5 mg/kg and 1 mg/kg) for 8 weeks. After treatment period, animals were tested in the open-field, elevated plus maze tests for anxiety-like behavior, and forced swimming test for depression-like behavior. The Morris Water Maze was used to evaluate the spatial learning and memory. The hippocampus of each animal was taken for biochemical examination. The results showed that Ni administration dose dependently increased anxiety-like behavior in both tests. A significant increase in depression-like symptoms was also exhibited by Ni treated rats. In the Morris Water Maze test, the spatial learning and memory were significantly impaired just in males treated with 1 mg/kg of Ni. With regard to biochemical analysis, activity of catalase (CAT) and superoxide dismutase (SOD) were significantly decreased, while the levels of nitric oxide (NO) and lipid peroxidation (LPO) in the hippocampus were significantly increased in the Ni-treated groups. Consequently, chronic Ni administration induced behavioral and biochemical dysfunctions.
Collapse
Affiliation(s)
- Mouloud Lamtai
- Unit of Nervous and Endocrine Physiology, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - Jihane Chaibat
- Unit of Nervous and Endocrine Physiology, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - Sihame Ouakki
- Unit of Nervous and Endocrine Physiology, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - Oussama Zghari
- Unit of Nervous and Endocrine Physiology, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - Abdelhalem Mesfioui
- Unit of Nervous and Endocrine Physiology, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - Aboubaker El Hessni
- Unit of Nervous and Endocrine Physiology, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - El-Housseine Rifi
- Laboratory of Synthesis Organic and Extraction Processes, Department of Chemistry, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - Ilias Marmouzi
- Laboratoire de Pharmacologie et Toxicologie, équipe de Pharmacocinétique, Faculté de Médecine et de Pharmacie, University Mohammed V in Rabat, BP 6203, Rabat Instituts, Rabat 10100, Morocco.
| | - Azzouz Essamri
- Laboratory of Agro-Resources and Process Engineering, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| | - Ali Ouichou
- Unit of Nervous and Endocrine Physiology, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Science, University Ibn Tofail, Kenitra 14000, Morocco.
| |
Collapse
|
14
|
Burke RD, Todd SW, Lumsden E, Mullins RJ, Mamczarz J, Fawcett WP, Gullapalli RP, Randall WR, Pereira EFR, Albuquerque EX. Developmental neurotoxicity of the organophosphorus insecticide chlorpyrifos: from clinical findings to preclinical models and potential mechanisms. J Neurochem 2017; 142 Suppl 2:162-177. [PMID: 28791702 DOI: 10.1111/jnc.14077] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 12/20/2022]
Abstract
Organophosphorus (OP) insecticides are pest-control agents heavily used worldwide. Unfortunately, they are also well known for the toxic effects that they can trigger in humans. Clinical manifestations of an acute exposure of humans to OP insecticides include a well-defined cholinergic crisis that develops as a result of the irreversible inhibition of acetylcholinesterase (AChE), the enzyme that hydrolyzes the neurotransmitter acetylcholine (ACh). Prolonged exposures to levels of OP insecticides that are insufficient to trigger signs of acute intoxication, which are hereafter referred to as subacute exposures, have also been associated with neurological deficits. In particular, epidemiological studies have reported statistically significant correlations between prenatal subacute exposures to OP insecticides, including chlorpyrifos, and neurological deficits that range from cognitive impairments to tremors in childhood. The primary objectives of this article are: (i) to address the short- and long-term neurological issues that have been associated with acute and subacute exposures of humans to OP insecticides, especially early in life (ii) to discuss the translational relevance of animal models of developmental exposure to OP insecticides, and (iii) to review mechanisms that are likely to contribute to the developmental neurotoxicity of OP insecticides. Most of the discussion will be focused on chlorpyrifos, the top-selling OP insecticide in the United States and throughout the world. These points are critical for the identification and development of safe and effective interventions to counter and/or prevent the neurotoxic effects of these chemicals in the developing brain. This is an article for the special issue XVth International Symposium on Cholinergic Mechanisms.
Collapse
Affiliation(s)
- Richard D Burke
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Spencer W Todd
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Eric Lumsden
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Roger J Mullins
- Department of Diagnostic Radiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jacek Mamczarz
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - William P Fawcett
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rao P Gullapalli
- Department of Diagnostic Radiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - William R Randall
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Edna F R Pereira
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Edson X Albuquerque
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Abreu-Villaça Y, Levin ED. Developmental neurotoxicity of succeeding generations of insecticides. ENVIRONMENT INTERNATIONAL 2017; 99:55-77. [PMID: 27908457 PMCID: PMC5285268 DOI: 10.1016/j.envint.2016.11.019] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/17/2016] [Accepted: 11/17/2016] [Indexed: 05/19/2023]
Abstract
Insecticides are by design toxic. They must be toxic to effectively kill target species of insects. Unfortunately, they also have off-target toxic effects that can harm other species, including humans. Developmental neurotoxicity is one of the most prominent off-target toxic risks of insecticides. Over the past seven decades several classes of insecticides have been developed, each with their own mechanisms of effect and toxic side effects. This review covers the developmental neurotoxicity of the succeeding generations of insecticides including organochlorines, organophosphates, pyrethroids, carbamates and neonicotinoids. The goal of new insecticide development is to more effectively kill target species with fewer toxic side effects on non-target species. From the experience with the developmental neurotoxicity caused by the generations of insecticides developed in the past advice is offered how to proceed with future insecticide development to decrease neurotoxic risk.
Collapse
Affiliation(s)
- Yael Abreu-Villaça
- Departamento de Ciências Fisiologicas, Universidade do Estado do Rio de Janeiro (UERJ), RJ, Brazil
| | - Edward D Levin
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
16
|
Rezvanfar MA, Hodjat M, Abdollahi M. Growing knowledge of using embryonic stem cells as a novel tool in developmental risk assessment of environmental toxicants. Life Sci 2016; 158:137-160. [DOI: 10.1016/j.lfs.2016.05.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/27/2016] [Accepted: 05/17/2016] [Indexed: 12/19/2022]
|
17
|
The neuroprotective effects of taurine against nickel by reducing oxidative stress and maintaining mitochondrial function in cortical neurons. Neurosci Lett 2015; 590:52-7. [PMID: 25637701 DOI: 10.1016/j.neulet.2015.01.065] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/16/2015] [Accepted: 01/26/2015] [Indexed: 11/20/2022]
Abstract
Previous studies have indicated that oxidative stress and mitochondrial dysfunction are involved in the toxicity of nickel. Taurine is recognized as an efficient antioxidant and is essential for mitochondrial function. To investigate whether taurine could protect against the neurotoxicity of nickel, we exposed primary cultured cortical neurons to various concentrations of nickel chloride (NiCl2; 0.5mM, 1mM and 2mM) for 24h or to 1mM NiCl2 for various periods (0 h, 12h, 24h and 48 h). Our results showed that taurine efficiently reduced lactate dehydrogenase (LDH) release induced by NiCl2. Along with this protective effect, taurine pretreatment not only significantly reversed the increase of ROS production and mitochondrial superoxide concentration, but also attenuated the decrease of superoxide dismutase (SOD) activity and glutathione (GSH) concentration in neurons exposed to NiCl2 for 24h. Moreover, nickel exposure reduced ATP production, disrupted the mitochondrial membrane potential and decreased mtDNA content. These types of oxidative damage in the mitochondria were efficiently ameliorated by taurine pretreatment. Taken together, our results indicate that the neuroprotective effects of taurine against the toxicity of nickel might largely depend on its roles in reducing oxidative stress and improving mitochondrial function. Taurine may have great pharmacological potential in treating the adverse effects of nickel in the nervous system.
Collapse
|
18
|
Alteration of hedgehog signaling by chronic exposure to different pesticide formulations and unveiling the regenerative potential of recombinant sonic hedgehog in mouse model of bone marrow aplasia. Mol Cell Biochem 2014; 401:115-31. [DOI: 10.1007/s11010-014-2299-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/27/2014] [Indexed: 10/24/2022]
|
19
|
Allen EMG, Florang VR, Davenport LL, Jinsmaa Y, Doorn JA. Cellular localization of dieldrin and structure-activity relationship of dieldrin analogues in dopaminergic cells. Chem Res Toxicol 2013; 26:1043-54. [PMID: 23763672 DOI: 10.1021/tx300458b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The incidence of Parkinson's disease (PD) correlates with environmental exposure to pesticides, such as the organochlorine insecticide, dieldrin. Previous studies found an increased concentration of the pesticide in the striatal region of the brains of PD patients and also that dieldrin adversely affects cellular processes associated with PD. These processes include mitochondrial function and reactive oxygen species production. However, the mechanism and specific cellular targets responsible for dieldrin-mediated cellular dysfunction and the structural components of dieldrin contributing to its toxicity (toxicophore) have not been fully defined. In order to identify the toxicophore of dieldrin, a structure-activity approach was used, with the toxicity profiles of numerous analogues of dieldrin (including aldrin, endrin, and cis-aldrin diol) assessed in PC6-3 cells. The MTT and lactate dehydrogenase (LDH) assays were used to monitor cell viability and membrane permeability after treatment with each compound. Cellular assays monitoring ROS production and extracellular dopamine metabolite levels were also used. Structure and stereochemistry for dieldrin were found to be very important for toxicity and other end points measured. Small changes in structure for dieldrin (e.g., comparison to the stereoisomer endrin) yielded significant differences in toxicity. Interestingly, the cis-diol metabolite of dieldrin was found to be significantly more toxic than the parent compound. Disruption of dopamine catabolism yielded elevated levels of the neurotoxin, 3,4-dihydroxyphenylacetaldehyde, for many organochlorines. Comparisons of the toxicity profiles for each dieldrin analogue indicated a structure-specific effect important for elucidating the mechanisms of dieldrin neurotoxicity.
Collapse
Affiliation(s)
- Erin M G Allen
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, The University of Iowa , Iowa City, Iowa 52242, United States
| | | | | | | | | |
Collapse
|
20
|
Gao X, Lin H, Ray R, Ray P. Toxicogenomic studies of human neural cells following exposure to organophosphorus chemical warfare nerve agent VX. Neurochem Res 2013; 38:916-34. [PMID: 23440544 DOI: 10.1007/s11064-013-0996-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/25/2013] [Accepted: 01/31/2013] [Indexed: 02/01/2023]
Abstract
Organophosphorus (OP) compounds represent an important group of chemical warfare nerve agents that remains a significant and constant military and civilian threat. OP compounds are considered acting primarily via cholinergic pathways by binding irreversibly to acetylcholinesterase, an important regulator of the neurotransmitter acetylcholine. Many studies over the past years have suggested that other mechanisms of OP toxicity exist, which need to be unraveled by a comprehensive and systematic approach such as genome-wide gene expression analysis. Here we performed a microarray study in which cultured human neural cells were exposed to 0.1 or 10 μM of VX for 1 h. Global gene expression changes were analyzed 6, 24, and 72 h post exposure. Functional annotation and pathway analysis of the differentially expressed genes has revealed many genes, networks and canonical pathways that are related to nervous system development and function, or to neurodegenerative diseases such as Alzheimer's disease, Huntington's disease, and Parkinson's disease. In particular, the neuregulin pathway impacted by VX exposure has important implications in many nervous system diseases including schizophrenia. These results provide useful information valuable in developing suitable antidotes for more effective prevention and treatment of, as well as in developing biomarkers for, VX-induced chronic neurotoxicity.
Collapse
Affiliation(s)
- Xiugong Gao
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA.
| | | | | | | |
Collapse
|
21
|
Li T, Zhao H, Hung GC, Han J, Tsai S, Li B, Zhang J, Puri RK, Lo SC. Differentially expressed genes and pathways induced by organophosphates in human neuroblastoma cells. Exp Biol Med (Maywood) 2013; 237:1413-23. [PMID: 23354400 DOI: 10.1258/ebm.2012.012178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Organophosphates (OPs) are toxic chemicals commonly used as pesticides and herbicides. Some OPs are highly toxic to humans and have been used in warfare and terrorist attacks. In order to elucidate the molecular mechanisms of injury caused by OPs, the differentially expressed genes were analyzed in human SK-N-SH neuroblastoma cells induced by three OPs. The SK-N-SH cells were treated with one of the three OPs, chlorpyrifos, dichlorvos or methamidophos at LC20 (high-dose), the concentration causing 20% cell death, as well as 1/20 of LC20 (low-dose), a sub-lethal concentration with no detectable cell death, for 24 h. The genome-wide gene changes were identified by Agilent Microarray System, and analyzed by microarray analysis tools. The analysis revealed neuroblastoma cells treated with the high doses of all three OPs markedly activated cell apoptosis and inhibited cell growth and proliferation genes, which would most likely lead to the process of cell death. Interestingly, the analysis also revealed significant decrease in expressions of many genes in a specific spliceosome pathway in cells treated with the low doses of all three different OPs. The change of spliceosome pathway may represent an important mechanism of injury in neuronal cells exposed to low doses of various OPs. In addition to unraveling a potentially different form of OP pathogenesis, this finding could provide a new diagnostic marker in assessing OP-associated injury in cells or tissues. In addition, these results could also contribute to the development of new prevention and/or therapeutic regimens against OP toxicity.
Collapse
Affiliation(s)
- Tianwei Li
- Tissue Safety Laboratory Program, Center for Biologics Evaluation and Research, Food and Drug Administration, NIH Building 29B, 29 Lincoln Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Magnarelli G, Fonovich T. Protein phosphorylation pathways disruption by pesticides. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abc.2013.35050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Estevan C, Vilanova E, Sogorb MA. Chlorpyrifos and its metabolites alter gene expression at non-cytotoxic concentrations in D3 mouse embryonic stem cells under in vitro differentiation: considerations for embryotoxic risk assessment. Toxicol Lett 2012; 217:14-22. [PMID: 23220036 DOI: 10.1016/j.toxlet.2012.11.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 11/27/2012] [Accepted: 11/29/2012] [Indexed: 11/15/2022]
Abstract
The effects of organophosphate insecticide chlorpyrifos (CPF) on development are currently under discussion. CPF and its metabolites, chlorpyrifos-oxon (CPO) and 3,5,6-trichloro-2-pyridinol (TClP), were more cytotoxic for D3 mouse embryonic stem cells than for differentiated fibroblasts 3T3 cells. Exposure to 10 μM CPF and TClP and 100 μM CPO for 12 h significantly altered the in vitro expression of biomarkers of differentiation in D3 cells. Similarly, exposure to 20 μM CPF and 25 μM CPO and TClP for 3 days also altered the expression of the biomarkers in the same model. These exposures caused no significant reduction in D3 viability with mild inhibition of acetylcholinesterase and neuropathy target esterase by CPF and severe inhibition by CPO. We conclude that certain in vivo exposure scenarios are possible, which cause inhibition of acetylcholinesterase but without clinical symptoms that reach high enough systemic CPF concentrations able to alter the expression of genes involved in cellular differentiation with potentially hazard effects on development. Conversely, the risk for embryotoxicity by CPO and TClP was very low because the required exposure would induce severe cholinergic syndrome.
Collapse
Affiliation(s)
- Carmen Estevan
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, 03202 Elche, Spain.
| | | | | |
Collapse
|
24
|
Chen S, Liu C, Peng C, Liu H, Hu M, Zhong G. Biodegradation of chlorpyrifos and its hydrolysis product 3,5,6-trichloro-2-pyridinol by a new fungal strain Cladosporium cladosporioides Hu-01. PLoS One 2012; 7:e47205. [PMID: 23056611 PMCID: PMC3466218 DOI: 10.1371/journal.pone.0047205] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 09/10/2012] [Indexed: 12/02/2022] Open
Abstract
Intensive use of chlorpyrifos has resulted in its ubiquitous presence as a contaminant in surface streams and soils. It is thus critically essential to develop bioremediation methods to degrade and eliminate this pollutant from environments. We present here that a new fungal strain Hu-01 with high chlorpyrifos-degradation activity was isolated and identified as Cladosporium cladosporioides based on the morphology and 5.8S rDNA gene analysis. Strain Hu-01 utilized 50 mg·L−1 of chlorpyrifos as the sole carbon of source, and tolerated high concentration of chlorpyrifos up to 500 mg·L−1. The optimum degradation conditions were determined to be 26.8°C and pH 6.5 based on the response surface methodology (RSM). Under these conditions, strain Hu-01 completely metabolized the supplemented chlorpyrifos (50 mg·L−1) within 5 d. During the biodegradation process, transient accumulation of 3,5,6-trichloro-2-pyridinol (TCP) was observed. However, this intermediate product did not accumulate in the medium and disappeared quickly. No persistent accumulative metabolite was detected by gas chromatopraphy-mass spectrometry (GC-MS) analysis at the end of experiment. Furthermore, degradation kinetics of chlorpyrifos and TCP followed the first-order model. Compared to the non-inoculated controls, the half-lives (t1/2) of chlorpyrifos and TCP significantly reduced by 688.0 and 986.9 h with the inoculum, respectively. The isolate harbors the metabolic pathway for the complete detoxification of chlorpyrifos and its hydrolysis product TCP, thus suggesting the fungus may be a promising candidate for bioremediation of chlorpyrifos-contaminated water, soil or crop.
Collapse
Affiliation(s)
- Shaohua Chen
- Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Chenglan Liu
- Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Chuyan Peng
- Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Hongmei Liu
- Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Meiying Hu
- Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Guohua Zhong
- Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou, People’s Republic of China
- * E-mail:
| |
Collapse
|
25
|
Slotkin TA, Seidler FJ. Does mechanism matter? Unrelated neurotoxicants converge on cell cycle and apoptosis during neurodifferentiation. Neurotoxicol Teratol 2012; 34:395-402. [PMID: 22546817 DOI: 10.1016/j.ntt.2012.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 03/09/2012] [Accepted: 04/13/2012] [Indexed: 10/28/2022]
Abstract
Mechanistically unrelated developmental neurotoxicants often produce neural cell loss culminating in similar functional and behavioral outcomes. We compared an organophosphate pesticide (diazinon), an organochlorine pesticide (dieldrin) and a metal (Ni(2+)) for effects on the genes regulating cell cycle and apoptosis in differentiating PC12 cells, an in vitro model of neuronal development. Each agent was introduced at 30μM for 24 or 72h, treatments devoid of cytotoxicity. Using microarrays, we examined the mRNAs encoding nearly 400 genes involved in each of the biological processes. All three agents targeted both the cell cycle and apoptosis pathways, evidenced by significant transcriptional changes in 40-45% of the cell cycle-related genes and 30-40% of the apoptosis-related genes. There was also a high degree of overlap as to which specific genes were affected by the diverse agents, with 80 cell cycle genes and 56 apoptosis genes common to all three. Concordance analysis, which assesses stringent matching of the direction, magnitude and timing of the transcriptional changes, showed highly significant correlations for pairwise comparisons of all the agents, for both cell cycle and apoptosis. Our results show that otherwise disparate developmental neurotoxicants converge on common cellular pathways governing the acquisition and programmed death of neural cells, providing a specific link to cell deficits. Our studies suggest that identifying the initial mechanism of action of a developmental neurotoxicant may be strategically less important than focusing on the pathways that converge on common final outcomes such as cell loss.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, USA.
| | | |
Collapse
|
26
|
Sadiq S, Ghazala Z, Chowdhury A, Büsselberg D. Metal toxicity at the synapse: presynaptic, postsynaptic, and long-term effects. J Toxicol 2012; 2012:132671. [PMID: 22287959 PMCID: PMC3263637 DOI: 10.1155/2012/132671] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 07/05/2011] [Indexed: 12/19/2022] Open
Abstract
Metal neurotoxicity is a global health concern. This paper summarizes the evidence for metal interactions with synaptic transmission and synaptic plasticity. Presynaptically metal ions modulate neurotransmitter release through their interaction with synaptic vesicles, ion channels, and the metabolism of neurotransmitters (NT). Many metals (e.g., Pb(2+), Cd(2+), and Hg(+)) also interact with intracellular signaling pathways. Postsynaptically, processes associated with the binding of NT to their receptors, activation of channels, and degradation of NT are altered by metals. Zn(2+), Pb(2+), Cu(2+), Cd(2+), Ni(2+), Co(2+), Li(3+), Hg(+), and methylmercury modulate NMDA, AMPA/kainate, and/or GABA receptors activity. Al(3+), Pb(2+), Cd(2+), and As(2)O(3) also impair synaptic plasticity by targeting molecules such as CaM, PKC, and NOS as well as the transcription machinery involved in the maintenance of synaptic plasticity. The multiple effects of metals might occur simultaneously and are based on the specific metal species, metal concentrations, and the types of neurons involved.
Collapse
Affiliation(s)
| | | | | | - Dietrich Büsselberg
- Weill Cornell Medical College in Qatar, Qatar Foundation—Education City, P.O. Box 24144, Doha, Qatar
| |
Collapse
|
27
|
Speed HE, Blaiss CA, Kim A, Haws ME, Melvin NR, Jennings M, Eisch AJ, Powell CM. Delayed reduction of hippocampal synaptic transmission and spines following exposure to repeated subclinical doses of organophosphorus pesticide in adult mice. Toxicol Sci 2012; 125:196-208. [PMID: 21948870 PMCID: PMC3247802 DOI: 10.1093/toxsci/kfr253] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/19/2011] [Indexed: 12/12/2022] Open
Abstract
Agricultural and household organophosphorus (OP) pesticides inhibit acetylcholinesterase (AchE), resulting in increased acetylcholine (Ach) in the central nervous system. In adults, acute and prolonged exposure to high doses of AchE inhibitors causes severe, clinically apparent symptoms, followed by lasting memory impairments and cognitive dysfunction. The neurotoxicity of repeated environmental exposure to lower, subclinical doses of OP pesticides in adults is not as well studied. However, repeated exposure to acetylcholinesterase inhibitors, such as chlorpyrifos (CPF), pyridostigmine, and sarin nerve agent, has been epidemiologically linked to delayed onset symptoms in Gulf War Illness and may be relevant to environmental exposure in farm workers among others. We treated adult mice with a subclinical dose (5 mg/kg) of CPF for 5 consecutive days and investigated hippocampal synaptic transmission and spine density early (2-7 days) and late (3 months) after CPF administration. No signs of cholinergic toxicity were observed at any time during or after treatment. At 2-7 days after the last injection, we found increased synaptic transmission in the CA3-CA1 region of the hippocampus of CPF-treated mice compared with controls. In contrast, at 3 months after CPF administration, we observed a 50% reduction in synaptic transmission likely due to a corresponding 50% decrease in CA1 pyramidal neuron synaptic spine density. This study is the first to identify a biphasic progression of synaptic abnormalities following repeated OP exposure and suggests that even in the absence of acute cholinergic toxicity, repeated exposure to CPF causes delayed persistent damage to the adult brain in vivo.
Collapse
MESH Headings
- Acetylcholinesterase/metabolism
- Animals
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/enzymology
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiopathology
- CA3 Region, Hippocampal/drug effects
- CA3 Region, Hippocampal/enzymology
- CA3 Region, Hippocampal/pathology
- CA3 Region, Hippocampal/physiopathology
- Cell Count
- Chlorpyrifos/toxicity
- Dendritic Spines/drug effects
- Dendritic Spines/pathology
- Dose-Response Relationship, Drug
- Hippocampus/drug effects
- Hippocampus/enzymology
- Hippocampus/pathology
- Hippocampus/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Patch-Clamp Techniques
- Pesticides/toxicity
- Pyramidal Cells/drug effects
- Pyramidal Cells/pathology
- Synaptic Transmission/drug effects
- Time Factors
Collapse
Affiliation(s)
| | | | - Ahleum Kim
- Department of Neurology & Neurotherapeutics
| | - Michael E. Haws
- Department of Neurology & Neurotherapeutics
- Neuroscience Graduate Program
| | - Neal R. Melvin
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813
| | | | - Amelia J. Eisch
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813
| | - Craig M. Powell
- Department of Neurology & Neurotherapeutics
- Neuroscience Graduate Program
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813
| |
Collapse
|
28
|
Slotkin TA, Seidler FJ. Developmental neurotoxicity of organophosphates targets cell cycle and apoptosis, revealed by transcriptional profiles in vivo and in vitro. Neurotoxicol Teratol 2011; 34:232-41. [PMID: 22222554 DOI: 10.1016/j.ntt.2011.12.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 12/20/2011] [Accepted: 12/20/2011] [Indexed: 11/25/2022]
Abstract
Developmental organophosphate exposure reduces the numbers of neural cells, contributing to neurobehavioral deficits. We administered chlorpyrifos or diazinon to newborn rats on postnatal days 1-4, in doses straddling the threshold for barely-detectable cholinesterase inhibition, and evaluated gene expression in the cell cycle and apoptosis pathways on postnatal day 5. Both organophosphates evoked transcriptional changes in 20-25% of the genes in each category; chlorpyrifos and diazinon targeted the same genes, with similar magnitudes of change, as evidenced by high concordance. Furthermore, the same effects were obtained with doses above or below the threshold for cholinesterase inhibition, indicating a mechanism unrelated to anticholinesterase actions. We then evaluated the effects of chlorpyrifos in undifferentiated and differentiating PC12 cells and found even greater targeting of cell cycle and apoptosis genes, affecting up to 40% of all genes in the pathways. Notably, the genes affected in undifferentiated cells were not concordant with those in differentiating cells, pointing to dissimilar outcomes dependent on developmental stage. The in vitro model successfully identified 60-70% of the genes affected by chlorpyrifos in vivo, indicating that the effects are exerted directly on developing neural cells. Our results show that organophosphates target the genes regulating the cell cycle and apoptosis in the developing brain and in neuronotypic cells in culture, with the pattern of vulnerability dependent on the specific stage of development. Equally important, these effects do not reflect actions on cholinesterase and operate at exposures below the threshold for any detectable inhibition of this enzyme.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
29
|
Slotkin TA, Seidler FJ. Developmental exposure to organophosphates triggers transcriptional changes in genes associated with Parkinson's disease in vitro and in vivo. Brain Res Bull 2011; 86:340-7. [PMID: 21968025 DOI: 10.1016/j.brainresbull.2011.09.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/19/2011] [Accepted: 09/20/2011] [Indexed: 10/17/2022]
Abstract
Epidemiologic studies support a connection between organophosphate pesticide exposures and subsequent risk of Parkinson's disease (PD). We used differentiating, neuronotypic PC12 cells to compare organophosphates (chlorpyrifos, diazinon), an organochlorine (dieldrin) and a metal (Ni(2+)) for their effects on the transcription of PD-related genes. Both of the organophosphates elicited significant changes in gene expression but with differing patterns: chlorpyrifos evoked both up- and downregulation whereas diazinon elicited overall reductions in expression. Dieldrin was without effect but Ni(2+) produced a pattern resembling that of diazinon. We then exposed neonatal rats to chlorpyrifos or diazinon for the first 4 days after birth and examined the expression of PD-related genes in the brainstem and forebrain. Chlorpyrifos had no significant effect whereas diazinon produced significant increases and decreases in expression of the same PD genes that were targeted in vitro. Our results provide some of the first evidence for a mechanistic relationship between developmental organophosphate exposure and the genes known to confer PD risk in humans; but they also point to disparities between different organophosphates that reinforce the concept that their neurotoxic actions do not rest solely on their shared property as cholinesterase inhibitors. The parallel effects of diazinon and Ni(2+) also show how otherwise unrelated developmental neurotoxicants can nevertheless produce similar outcomes by converging on common molecular pathways, further suggesting a need to examine metals such as Ni(2+) as potential contributors to PD risk.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Box 3813 DUMC, Durham, NC 27710, USA.
| | | |
Collapse
|
30
|
Canesi L, Negri A, Barmo C, Banni M, Gallo G, Viarengo A, Dondero F. The organophosphate Chlorpyrifos interferes with the responses to 17β-estradiol in the digestive gland of the marine mussel Mytilus galloprovincialis. PLoS One 2011; 6:e19803. [PMID: 21625485 PMCID: PMC3098840 DOI: 10.1371/journal.pone.0019803] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 04/06/2011] [Indexed: 01/12/2023] Open
Abstract
Background Many pesticides have been shown to act as endocrine disrupters. Although the
potencies of currently used pesticides as hormone agonists/antagonists are
low compared with those of natural ligands, their ability to act via
multiple mechanisms might enhance the biological effect. The organophosphate
Chlorpyrifos (CHP) has been shown to be weakly estrogenic and cause adverse
neurodevelopmental effects in mammals. However, no information is available
on the endocrine effects of CHP in aquatic organisms. In the digestive gland
of the bivalve Mytilus galloprovincialis, a target tissue
of both estrogens and pesticides, the possible effects of CHP on the
responses to the natural estrogen 17β-estradiol (E2) were
investigated. Methodology/Principal Findings Mussels were exposed to CHP (4.5 mg/l, 72 hrs) and subsequently injected with
E2 (6.75 ng/g dw). Responses were evaluated in CHP,
E2 and CHP/E2 treatment groups at 24 h p.i. by a
biomarker/transcriptomic approach. CHP and E2 induced additive,
synergistic, and antagonistic effects on lysosomal biomarkers (lysosomal
membrane stability, lysosome/cytoplasm volume ratio, lipofuscin and neutral
lipid accumulation). Additive and synergistic effects were also observed on
the expression of estrogen-responsive genes (GSTπ, catalase, 5-HTR)
evaluated by RT-Q-PCR. The use of a 1.7K cDNA Mytilus
microarray showed that CHP, E2 and CHP/E2, induced 81,
44, and 65 Differentially Expressed Genes (DEGs), respectively. 24 genes
were exclusively shared between CHP and CHP/E2, only 2 genes
between E2 and CHP/E2. Moreover, 36 genes were
uniquely modulated by CHP/E2. Gene ontology annotation was used
to elucidate the putative mechanisms involved in the responses elicited by
different treatments. Conclusions The results show complex interactions between CHP and E2 in the
digestive gland, indicating that the combination of certain pesticides and
hormones may give rise to unexpected effects at the molecular/cellular
level. Overall, these data demonstrate that CHP can interfere with the
mussel responses to natural estrogens.
Collapse
Affiliation(s)
- Laura Canesi
- Dipartimento di Biologia, Università di Genova, Genova, Italy.
| | | | | | | | | | | | | |
Collapse
|
31
|
Dondero F, Banni M, Negri A, Boatti L, Dagnino A, Viarengo A. Interactions of a pesticide/heavy metal mixture in marine bivalves: a transcriptomic assessment. BMC Genomics 2011; 12:195. [PMID: 21496282 PMCID: PMC3094310 DOI: 10.1186/1471-2164-12-195] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 04/16/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mixtures of chemicals present in aquatic environments may elicit toxicity due to additive or synergistic effects among the constituents or, vice versa, the adverse outcome may be reduced by antagonistic interactions. Deviations from additivity should be explained either by the perturbations of toxicokinetic parameters and/or chemical toxicodynamics. We addressed this important question in marine mussels exposed subchronically to a binary mixture made of two wide-spread pollutants: the heavy metal nickel and the organic phosphorus pesticide Chlorpyrifos. To this aim, we carried out in tissues of Mytius galloprovincialis (Lam) a systems approach based on the evaluation and integration of different disciplines, i.e. high throughput gene expression profiling, functional genomics, stress biomakers and toxicokinetics. RESULTS Cellular and tissue biomarkers, viz. digestive gland lysosomal membrane stability, lysosomal/cytosol volume ratio, neutral lipid content and gill acetylcholinesterase activity were, in general, altered by either the exposure to nickel and Chlorpyrifos. However, their joint action rendered (i) an overall decrease of the stress syndrome level, as evaluated through an expert system integrating biomarkers and (ii) statistically significant antagonistic deviations from the reference model systems to predict mixture toxicity. While toxicokinetic modeling did not explain mixture interactions, gene expression profiling and further Gene Ontology-based functional genomics analysis provided clues that the decrement of toxicity may arise from the development of specific toxicodynamics. Multivariate statistics of microarray data (238 genes in total, representing about 14% of the whole microarray catalogue) showed two separate patterns for the single chemicals: the one belonging to the heavy metal -135 differentially expressed genes (DEGs) was characterized by the modulation of transcript levels involved in nucleic acid metabolism, cell proliferation and lipid metabolic processes. Chlorpyrifos exposure (43 DEGs) yielded a molecular signature which was biased towards carbohydrate catabolism (indeed, chitin metabolism) and developmental processes. The exposure to the mixture (103 DEGs) elicited a composite complex profile which encompassed the core properties of the pesticide but also a relevant set of unique features. Finally, the relative mRNA abundance of twelve genes was followed by Q-PCR to either confirm or complement microarray data. These results, in general, were compatible with those from arrays and indeed confirmed the association of the relative abundance of two GM-2 ganglioside activator genes in the development of the hyperlipidosis syndrome observed in digestive gland lysosomes of single chemical exposed mussels. CONCLUSION The transcriptomic assessment fitted with biological data to indicate the occurrence of different toxicodynamic events and, in general, a decrease of toxicity, driven by the mitigation or even abolition of lysosomal responses. Furthermore, our results emphasized the importance of the application of mechanistic approaches and the power of systems assessment to study toxicological responses in ecologically relevant organisms.
Collapse
Affiliation(s)
- Francesco Dondero
- Department of Environmental and Life Sciences, Università del Piemonte Orientale Amedeo Avogadro, Alessandria, Italy.
| | | | | | | | | | | |
Collapse
|
32
|
Maiese K, Chong ZZ, Shang YC, Hou J. Novel avenues of drug discovery and biomarkers for diabetes mellitus. J Clin Pharmacol 2011; 51:128-52. [PMID: 20220043 PMCID: PMC3033756 DOI: 10.1177/0091270010362904] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Globally, developed nations spend a significant amount of their resources on health care initiatives that poorly translate into increased population life expectancy. As an example, the United States devotes 16% of its gross domestic product to health care, the highest level in the world, but falls behind other nations that enjoy greater individual life expectancy. These observations point to the need for pioneering avenues of drug discovery to increase life span with controlled costs. In particular, innovative drug development for metabolic disorders such as diabetes mellitus becomes increasingly critical given that the number of diabetic people will increase exponentially over the next 20 years. This article discusses the elucidation and targeting of novel cellular pathways that are intimately tied to oxidative stress in diabetes mellitus for new treatment strategies. Pathways that involve wingless, β-nicotinamide adenine dinucleotide (NAD(+)) precursors, and cytokines govern complex biological pathways that determine both cell survival and longevity during diabetes mellitus and its complications. Furthermore, the role of these entities as biomarkers for disease can further enhance their utility irrespective of their treatment potential. Greater understanding of the intricacies of these unique cellular mechanisms will shape future drug discovery for diabetes mellitus to provide focused clinical care with limited or absent long-term complications.
Collapse
Affiliation(s)
- Kenneth Maiese
- Department of Neurology, 8C-1 UHC, Wayne State University School of Medicine, 4201 St. Antoine, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
33
|
Ray A, Liu J, Ayoubi P, Pope C. Dose-related gene expression changes in forebrain following acute, low-level chlorpyrifos exposure in neonatal rats. Toxicol Appl Pharmacol 2010; 248:144-55. [PMID: 20691718 PMCID: PMC2946483 DOI: 10.1016/j.taap.2010.07.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/16/2010] [Accepted: 07/27/2010] [Indexed: 12/27/2022]
Abstract
Chlorpyrifos (CPF) is a widely used organophosphorus insecticide (OP) and putative developmental neurotoxicant in humans. The acute toxicity of CPF is elicited by acetylcholinesterase (AChE) inhibition. We characterized dose-related (0.1, 0.5, 1 and 2mg/kg) gene expression profiles and changes in cell signaling pathways 24h following acute CPF exposure in 7-day-old rats. Microarray experiments indicated that approximately 9% of the 44,000 genes were differentially expressed following either one of the four CPF dosages studied (546, 505, 522, and 3,066 genes with 0.1, 0.5, 1.0 and 2.0mg/kg CPF). Genes were grouped according to dose-related expression patterns using K-means clustering while gene networks and canonical pathways were evaluated using Ingenuity Pathway Analysis®. Twenty clusters were identified and differential expression of selected genes was verified by RT-PCR. The four largest clusters (each containing from 276 to 905 genes) constituted over 50% of all differentially expressed genes and exhibited up-regulation following exposure to the highest dosage (2mg/kg CPF). The total number of gene networks affected by CPF also rose sharply with the highest dosage of CPF (18, 16, 18 and 50 with 0.1, 0.5, 1 and 2mg/kg CPF). Forebrain cholinesterase (ChE) activity was significantly reduced (26%) only in the highest dosage group. Based on magnitude of dose-related changes in differentially expressed genes, relative numbers of gene clusters and signaling networks affected, and forebrain ChE inhibition only at 2mg/kg CPF, we focused subsequent analyses on this treatment group. Six canonical pathways were identified that were significantly affected by 2mg/kg CPF (MAPK, oxidative stress, NFΚB, mitochondrial dysfunction, arylhydrocarbon receptor and adrenergic receptor signaling). Evaluation of different cellular functions of the differentially expressed genes suggested changes related to olfactory receptors, cell adhesion/migration, synapse/synaptic transmission and transcription/translation. Nine genes were differentially affected in all four CPF dosing groups. We conclude that the most robust, consistent changes in differential gene expression in neonatal forebrain across a range of acute CPF dosages occurred at an exposure level associated with the classical marker of OP toxicity, AChE inhibition. Disruption of multiple cellular pathways, in particular cell adhesion, may contribute to the developmental neurotoxicity potential of this pesticide.
Collapse
Affiliation(s)
- Anamika Ray
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74075, USA
| | | | | | | |
Collapse
|
34
|
Slotkin TA, Seidler FJ. Diverse neurotoxicants converge on gene expression for neuropeptides and their receptors in an in vitro model of neurodifferentiation: effects of chlorpyrifos, diazinon, dieldrin and divalent nickel in PC12 cells. Brain Res 2010; 1353:36-52. [PMID: 20682304 DOI: 10.1016/j.brainres.2010.07.073] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 07/14/2010] [Accepted: 07/21/2010] [Indexed: 11/17/2022]
Abstract
Unrelated developmental neurotoxicants can produce similar neurobehavioral outcomes. We examined whether disparate agents affect neuromodulators that control numerous neurotransmitters and circuits, employing PC12 cells to explore the targeting of neuroactive peptides by organophosphates (chlorpyrifos, diazinon), an organochlorine (dieldrin) and a metal (Ni(2+)); we utilized microarrays to profile gene expression for the peptides and their receptors. Chlorpyrifos evoked robust upregulation of cholecystokinin, corticotropin releasing hormone, galanin, neuropeptide Y, neurotensin, preproenkephalin and tachykinin 1; this involved a critical period at the commencement of neurodifferentiation, since the effects were much less notable in undifferentiated PC12 cells. Diazinon targeted a similar but smaller repertoire of neuropeptide genes and the magnitude of the effects was also generally less. Surprisingly, dieldrin shared many of the same neuropeptide targets as the organophosphates and concordance analysis showed significant overlap among all three pesticides. However, dieldrin had more notable effects on neuropeptide receptors, and overlap between diazinon and dieldrin for the receptors led to a stronger resemblance of these two agents than of chlorpyrifos and dieldrin. Ni(2+) was unique, evoking upregulation of only one of the peptides affected by the other agents, while causing downregulation of several others. Nevertheless, there was still significant concordance between Ni(2+) and either diazinon or dieldrin, reflecting similarities toward the receptors. Our results show that neuropeptides are likely to be a prominent target for the developmental neurotoxicity of organophosphates and other neurotoxicants, and further, that the convergence of disparate agents on the same genes and pathways may contribute to similar neurobehavioral outcomes.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
35
|
Xu SC, He MD, Zhong M, Zhang YW, Wang Y, Yang L, Yang J, Yu ZP, Zhou Z. Melatonin protects against Nickel-induced neurotoxicity in vitro by reducing oxidative stress and maintaining mitochondrial function. J Pineal Res 2010; 49:86-94. [PMID: 20536687 DOI: 10.1111/j.1600-079x.2010.00770.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nickel is a potential neurotoxic pollutant. Oxidative stress is supposed to be involved in the mechanism underlying nickel-induced neurotoxicity. Melatonin has efficient protective effects against various oxidative damages in nervous system. The purpose of this study was to investigate whether melatonin could efficiently protect against neurotoxicity induced by nickel. Here, we exposed primary cultured cortical neurons and mouse neuroblastoma cell lines (neuro2a) to different concentrations of nickel chloride (NiCl(2)) (0.125, 0.25, 0.5, and 1 mm) for 12 hr or 0.5 mm NiCl(2) for various periods (0, 3, 6, 12, and 24 hr). We found that nickel significantly increased reactive oxygen species production and caused the loss of cell viability both in cortical neurons and neuro2a cells. In addition, nickel exposure obviously inhibited the mitochondrial function, disrupted the mitochondrial membrane potential (DeltaPsim), reduced ATP production, and decreased mitochondrial DNA (mtDNA) content. However, each of these oxidative damages was efficiently attenuated by melatonin pretreatment. These protective effects of melatonin may be attributable to its roles in reducing oxidative stress and improving mitochondrial function in nickel-treated nerve cells. Our results suggested that melatonin may have great pharmacological potential in protecting against the adverse effects of nickel in the nervous system.
Collapse
Affiliation(s)
- Shang-Cheng Xu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Slotkin TA, Lobner D, Seidler FJ. Transcriptional profiles for glutamate transporters reveal differences between organophosphates but similarities with unrelated neurotoxicants. Brain Res Bull 2010; 83:76-83. [PMID: 20600679 DOI: 10.1016/j.brainresbull.2010.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 06/10/2010] [Accepted: 06/19/2010] [Indexed: 01/05/2023]
Abstract
The developmental neurotoxicity of organophosphates involves mechanisms other than their shared property as cholinesterase inhibitors, among which are excitotoxicity and oxidative stress. We used PC12 cells as a neurodevelopmental model to compare the effects of chlorpyrifos and diazinon on the expression of genes encoding glutamate transporters. Chlorpyrifos had a greater effect in cells undergoing nerve growth factor-induced neurodifferentiation as compared to undifferentiated PC12 cells, with peak sensitivity at the initiation of differentiation, reflecting a global upregulation of all the glutamate transporter genes expressed in this cell line. In differentiating cells, chlorpyrifos had a significantly greater effect than did diazinon and concordance analysis indicated no resemblance in their expression patterns. At the same time, the smaller effects of diazinon were highly concordant with those of an organochlorine pesticide (dieldrin) and a metal (divalent nickel). We also performed similar evaluations for the cystine/glutamate exchanger, which provides protection against oxidative stress by moving cystine into the cell; again, chlorpyrifos had the greatest effect, in this case reducing expression in undifferentiated and differentiating cells. Our results point to excitotoxicity and oxidative stress as major contributors to the noncholinesterase mechanisms that distinguish the neurodevelopmental outcomes between different organophosphates while providing a means whereby apparently unrelated neurotoxicants may produce similar outcomes.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
37
|
Maiese K, Shang YC, Chong ZZ, Hou J. Diabetes mellitus: channeling care through cellular discovery. Curr Neurovasc Res 2010; 7:59-64. [PMID: 20158461 DOI: 10.2174/156720210790820217] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 12/29/2009] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus (DM) impacts a significant portion of the world's population and care for this disorder places an economic burden on the gross domestic product for any particular country. Furthermore, both Type 1 and Type 2 DM are becoming increasingly prevalent and there is increased incidence of impaired glucose tolerance in the young. The complications of DM are protean and can involve multiple systems throughout the body that are susceptible to the detrimental effects of oxidative stress and apoptotic cell injury. For these reasons, innovative strategies are necessary for the implementation of new treatments for DM that are generated through the further understanding of cellular pathways that govern the pathological consequences of DM. In particular, both the precursor for the coenzyme beta-nicotinamide adenine dinucleotide (NAD(+)), nicotinamide, and the growth factor erythropoietin offer novel platforms for drug discovery that involve cellular metabolic homeostasis and inflammatory cell control. Interestingly, these agents and their tightly associated pathways that consist of cell cycle regulation, protein kinase B, forkhead transcription factors, and Wnt signaling also function in a broader sense as biomarkers for disease onset and progression.
Collapse
Affiliation(s)
- Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| | | | | | | |
Collapse
|
38
|
Kim YH, Lee Y, Kim D, Jung MW, Lee CJ. Scopolamine-induced learning impairment reversed by physostigmine in zebrafish. Neurosci Res 2010; 67:156-61. [PMID: 20298728 DOI: 10.1016/j.neures.2010.03.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 03/04/2010] [Accepted: 03/08/2010] [Indexed: 01/08/2023]
Abstract
In this study, the effects of scopolamine, an acetylcholine muscarinic receptor antagonist, and physostigmine, an acetylcholinesterase inhibitor, on the learning ability and memory of zebrafish were evaluated using a passive avoidance response test. The zebrafish were trained to stay in a dark compartment to avoid a weight dropping into an acryl shuttle chamber with a central sliding door. The crossing time was increased significantly, from 30.7+/-40.8s to 179.3+/-27.3s in the training session and 179.9+/-28.0s in the test session carried out 2h later in the controls. When treatment with 200 microM scopolamine was administered for 1h prior to the training session, the crossing time did not increase. The scopolamine-induced learning deficit was ameliorated by pretreatment with 20 microM physostigmine for 1h prior to scopolamine treatment; the crossing time was similarly increased, as shown with the controls (60.9+/-11.5s, 130.9+/-27.5s, and 183.4+/-26.6s in the training session and 108.1+/-23.9s in the test session). When scopolamine treatment was administered after the training session, the crossing time in the test session was reduced significantly as compared to that noted in the third trial of the training session, which was also ameliorated by physostigmine pretreatment. These results show that scopolamine impairs both the acquisition of passive avoidance response and retention of the learned response, and that physostigmine rescues the amnesic effects of scopolamine in zebrafish.
Collapse
Affiliation(s)
- Yeon-Hwa Kim
- Department of Biological Sciences, Institute of Molecular and Cellular Biology, Inha University, 253 Yong-Hyun Dong, Nam-Gu, Incheon 402-751, Republic of Korea
| | | | | | | | | |
Collapse
|
39
|
Oxidative stress: Biomarkers and novel therapeutic pathways. Exp Gerontol 2010; 45:217-34. [PMID: 20064603 DOI: 10.1016/j.exger.2010.01.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 12/28/2009] [Accepted: 01/07/2010] [Indexed: 01/12/2023]
Abstract
Oxidative stress significantly impacts multiple cellular pathways that can lead to the initiation and progression of varied disorders throughout the body. It therefore becomes imperative to elucidate the components and function of novel therapeutic strategies against oxidative stress to further clinical diagnosis and care. In particular, both the growth factor and cytokine erythropoietin (EPO) and members of the mammalian forkhead transcription factors of the O class (FoxOs) may offer the greatest promise for new treatment regimens since these agents and the cellular pathways they oversee cover a range of critical functions that directly influence progenitor cell development, cell survival and degeneration, metabolism, immune function, and cancer cell invasion. Furthermore, both EPO and FoxOs function not only as therapeutic targets, but also as biomarkers of disease onset and progression, since their cellular pathways are closely linked and overlap with several unique signal transduction pathways. However, biological outcome with EPO and FoxOs may sometimes be both unexpected and undesirable that can raise caution for these agents and warrant further investigations. Here we present the exciting as well as complicated role EPO and FoxOs possess to uncover the benefits as well as the risks of these agents for cell biology and clinical care in processes that range from stem cell development to uncontrolled cellular proliferation.
Collapse
|
40
|
Adigun AA, Seidler FJ, Slotkin TA. Disparate developmental neurotoxicants converge on the cyclic AMP signaling cascade, revealed by transcriptional profiles in vitro and in vivo. Brain Res 2009; 1316:1-16. [PMID: 20026089 DOI: 10.1016/j.brainres.2009.12.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 12/02/2009] [Accepted: 12/09/2009] [Indexed: 12/23/2022]
Abstract
Cell-signaling cascades are convergent targets for developmental neurotoxicity of otherwise unrelated agents. We compared organophosphates (chlorpyrifos, diazinon), an organochlorine (dieldrin) and a metal (Ni(2+)) for their effects on neuronotypic PC12 cells, assessing gene transcription involved in the cyclic AMP pathway. Each agent was introduced during neurodifferentiation at a concentration of 30 microM for 24 or 72 h and we assessed 69 genes encoding adenylyl cyclase isoforms and regulators, G-protein alpha-and beta,gamma-subunits, protein kinase A subtypes and the phosphodiesterase family. We found strong concordance among the four agents across all the gene families, with the strongest relationships for the G-proteins, followed by adenylyl cyclase, and lesser concordance for protein kinase A and phosphodiesterase. Superimposed on this pattern, chlorpyrifos and diazinon were surprisingly the least alike, whereas there was strong concordance of dieldrin and Ni(2+) with each other and with each individual organophosphate. Further, the effects of chlorpyrifos differed substantially depending on whether cells were undifferentiated or differentiating. To resolve the disparities between chlorpyrifos and diazinon, we performed analyses in rat brain regions after in vivo neonatal exposures; unlike the in vitro results, there was strong concordance. Our results show that unrelated developmental neurotoxicants can nevertheless produce similar outcomes by targeting cell signaling pathways involved in neurodifferentiation during a critical developmental period of vulnerability. Nevertheless, a full evaluation of concordance between different toxicants requires evaluations of in vitro systems that detect direct effects, as well as in vivo systems that allow for more complex interactions that converge on the same pathway.
Collapse
Affiliation(s)
- Abayomi A Adigun
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, Box 3813 DUMC, Duke Univ. Med. Ctr., Durham, NC 27710, USA
| | | | | |
Collapse
|
41
|
Slotkin TA, Seidler FJ. Oxidative stress from diverse developmental neurotoxicants: antioxidants protect against lipid peroxidation without preventing cell loss. Neurotoxicol Teratol 2009; 32:124-31. [PMID: 20004241 DOI: 10.1016/j.ntt.2009.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 12/02/2009] [Accepted: 12/02/2009] [Indexed: 12/20/2022]
Abstract
Oxidative stress has been hypothesized to provide a mechanism by which apparently unrelated chemicals can nevertheless produce similar developmental neurotoxic outcomes. We used differentiating PC12 cells to compare the effects of agents from four different classes and then to evaluate antioxidant amelioration: fipronil, perfluorooctanesulfonamide (PFOSA), dieldrin and chlorpyrifos. The rank order for lipid peroxidation corresponded to the ability to evoke cell loss: fipronil>PFOSA>dieldrin>chlorpyrifos. The same sequence was found for an index of cell enlargement (protein/DNA ratio) but the effects on neurite outgrowth (membrane/total protein) diverged, with fipronil producing a decrease and PFOSA an increase. Cotreatment with antioxidants reduced (ascorbate) or eliminated (Vitamin E) lipid peroxidation caused by each of the agents but failed to protect against cell loss, with the sole exception of chlorpyrifos, for which we earlier showed partial protection by Vitamin E; addition of higher NGF concentrations protected neither against oxidative stress nor cell loss. Despite the failure to prevent cell loss, ascorbate protected the cells from the effects of PFOSA on neuritic outgrowth; NGF, and to a lesser extent, ascorbate, offset the effects of fipronil on both cell enlargement and neuritogenesis. At the same time, the ameliorant treatments also worsened some of the other toxicant effects. Our results point out the problems in concluding that, just because a neurotoxicant produces oxidative stress, antioxidant therapy will be effective in preventing damage. Instead, additional mechanisms for each agent may provide alternative routes to neurotoxicity, or may be additive or synergistic with oxidative stress.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology Duke University Medical Center Durham, North Carolina 27710, USA.
| | | |
Collapse
|
42
|
Maiese K, Chong ZZ, Hou J, Shang YC. New strategies for Alzheimer's disease and cognitive impairment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2009; 2:279-89. [PMID: 20716915 PMCID: PMC2835916 DOI: 10.4161/oxim.2.5.9990] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/24/2009] [Accepted: 09/02/2009] [Indexed: 02/06/2023]
Abstract
Approximately five million people suffer with Alzheimer's disease (AD) and more than twenty-four million people are diagnosed with AD, pre-senile dementia, and other disorders of cognitive loss worldwide. Furthermore, the annual cost per patient with AD can approach $200,000 with an annual population aggregate cost of $100 billion. Yet, complete therapeutic prevention or reversal of neurovascular injury during AD and cognitive loss is not achievable despite the current understanding of the cellular pathways that modulate nervous system injury during these disorders. As a result, identification of novel therapeutic targets for the treatment of neurovascular injury would be extremely beneficial to reduce or eliminate disability from diseases that lead to cognitive loss or impairment. Here we describe the capacity of intrinsic cellular mechanisms for the novel pathways of erythropoietin and forkhead transcription factors that may offer not only new strategies for disorders such as AD and cognitive loss, but also function as biomarkers for disease onset and progression.
Collapse
Affiliation(s)
- Kenneth Maiese
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | | | |
Collapse
|
43
|
Maiese K, Hou J, Chong ZZ, Shang YC. Erythropoietin, forkhead proteins, and oxidative injury: biomarkers and biology. ScientificWorldJournal 2009; 9:1072-104. [PMID: 19802503 PMCID: PMC2762199 DOI: 10.1100/tsw.2009.121] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress significantly impacts multiple cellular pathways that can lead to the initiation and progression of varied disorders throughout the body. It therefore becomes imperative to elucidate the components and function of novel therapeutic strategies against oxidative stress to further clinical diagnosis and care. In particular, both the growth factor and cytokine erythropoietin (EPO), and members of the mammalian forkhead transcription factors of the O class (FoxOs), may offer the greatest promise for new treatment regimens, since these agents and the cellular pathways they oversee cover a range of critical functions that directly influence progenitor cell development, cell survival and degeneration, metabolism, immune function, and cancer cell invasion. Furthermore, both EPO and FoxOs function not only as therapeutic targets, but also as biomarkers of disease onset and progression, since their cellular pathways are closely linked and overlap with several unique signal transduction pathways. Yet, EPO and FoxOs may sometimes have unexpected and undesirable effects that can raise caution for these agents and warrant further investigations. Here we present the exciting as well as the complex role that EPO and FoxOs possess to uncover the benefits as well as the risks of these agents for cell biology and clinical care in processes that range from stem cell development to uncontrolled cellular proliferation.
Collapse
Affiliation(s)
- Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | | | |
Collapse
|