1
|
Neha V, Parithathvi A, Dsouza HS. Ameliorative role of bioactive compounds against lead-induced neurotoxicity. Neuroscience 2025; 568:46-56. [PMID: 39805419 DOI: 10.1016/j.neuroscience.2025.01.018] [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: 09/25/2024] [Revised: 12/04/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Lead (Pb) is an environmental toxin ubiquitously present in the human environment due to anthropogenic activities and industrialization. Lead can enter the human body through various sources and pathways, such as inhalation, ingestion and dermal contact, leading to detrimental health effects. The majority of lead that enters the body is removed by urine or feces; however, under chronic exposure conditions, lead is not efficient, as lead is absorbed and transferred to numerous organs, such as the brain, liver, kidney, muscles, and heart, and it is ultimately stored in mineralizing tissues such as bones and teeth. The central nervous system is the most affected among all the organs and systems affected, as lead is a known neurotoxin. Lead absorption is elevated in the fasting state than in the fed state. Chelation therapy, which is used to treat lead poisoning, has various adverse effects, making this treatment detrimental because it disrupts the levels of other essential elements and redistributes lead to various tissues. One of the main mechanisms by which lead induces toxicity is through the generation of reactive oxygen species. Hence, bioactive compounds that are the source of antioxidants if consumed along with ongoing lead exposure can ameliorate the toxic effects of lead.
Collapse
Affiliation(s)
- Venkatesan Neha
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Aluru Parithathvi
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences,Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - Herman Sunil Dsouza
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
2
|
Chen C, Wang X, Jin Y, Yao Y, Li S, Liao Z, Zhang X, Yan X. Simultaneous exposure to microplastics and heavy metal lead induces oxidative stress, histopathological damage, and immune dysfunction in marine mussel Mytilus coruscus. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117493. [PMID: 39644573 DOI: 10.1016/j.ecoenv.2024.117493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The increasing deposition of microplastics (MPs) in aquatic ecosystems is a worldwide concern. MPs can interact with other environmental pollutants, such as heavy metals, and change their toxicity. In this study, we focused on the effects of MPs and lead (Pb), as a toxic heavy metal, on marine mussel Mytilus coruscus under separate and co-exposure situations at environmentally relevant concentrations: MPs (1 mg/L) and Pb (50 μg/L). We found that MPs alone or in combination with Pb significantly decreased the respiration and filtration rates of the mussels (p < 0.05). Histological observations revealed varying extents of damage to the gill and digestive gland caused by a single exposure to MPs, which was aggravated by co-exposure to Pb. In addition, co-exposure induced a higher level of oxidative stress, which was reflected by an increase in hydrogen peroxide and malondialdehyde content, and a decrease in antioxidant enzyme activity. Meanwhile, co-exposure poses a significant threat to the immune function of the mussels, as evidenced by induction of hemocytes to produce excess reactive oxygen species (ROS), significantly reducing lysosome activity (p < 0.05), inhibiting the expression of autophagy-related genes, and inducing the expression of apoptosis-related genes, resulting in hemocyte apoptosis. Furthermore, the TLR/MyD88/NFκB signaling pathway is involved in the immune response of mussels to environmental stress. This study provides novel perspectives on the toxicity of MPs combined with Pb in marine animals, as well as the molecular mechanisms underlying their ecotoxicological effects.
Collapse
Affiliation(s)
- Chuanyue Chen
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China; School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Xueer Wang
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Yingrong Jin
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Yinchang Yao
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Suchang Li
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Zhi Liao
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Xiaolin Zhang
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Xiaojun Yan
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China; School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
3
|
Cervetto C, Pistollato F, Amato S, Mendoza-de Gyves E, Bal-Price A, Maura G, Marcoli M. Assessment of neurotransmitter release in human iPSC-derived neuronal/glial cells: a missing in vitro assay for regulatory developmental neurotoxicity testing. Reprod Toxicol 2023; 117:108358. [PMID: 36863571 PMCID: PMC10112275 DOI: 10.1016/j.reprotox.2023.108358] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived neural stem cells (NSCs) and their differentiated neuronal/glial derivatives have been recently considered suitable to assess in vitro developmental neurotoxicity (DNT) triggered by exposure to environmental chemicals. The use of human-relevant test systems combined with in vitro assays specific for different neurodevelopmental events, enables a mechanistic understanding of the possible impact of environmental chemicals on the developing brain, avoiding extrapolation uncertainties associated with in vivo studies. Currently proposed in vitro battery for regulatory DNT testing accounts for several assays suitable to study key neurodevelopmental processes, including NSC proliferation and apoptosis, differentiation into neurons and glia, neuronal migration, synaptogenesis, and neuronal network formation. However, assays suitable to measure interference of compounds with neurotransmitter release or clearance are at present not included, which represents a clear gap of the biological applicability domain of such a testing battery. Here we applied a HPLC-based methodology to measure the release of neurotransmitters in a previously characterized hiPSC-derived NSC model undergoing differentiation towards neurons and glia. Glutamate release was assessed in control cultures and upon depolarization, as well as in cultures repeatedly exposed to some known neurotoxicants (BDE47 and lead) and chemical mixtures. Obtained data indicate that these cells have the ability to release glutamate in a vesicular manner, and that both glutamate clearance and vesicular release concur in the maintenance of extracellular glutamate levels. In conclusion, analysis of neurotransmitter release is a sensitive readout that should be included in the envisioned battery of in vitro assays for DNT testing.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Centro 3R, Pisa, Italy.
| | | | - Sarah Amato
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Italy
| | | | - Anna Bal-Price
- European Commission, Joint Research Centre, JRC, Ispra, Italy.
| | - Guido Maura
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Italy
| | - Manuela Marcoli
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Centro 3R, Pisa, Italy.
| |
Collapse
|
4
|
Mukherjee S, Chatterjee N, Sircar A, Maikap S, Singh A, Acharyya S, Paul S. A Comparative Analysis of Heavy Metal Effects on Medicinal Plants. Appl Biochem Biotechnol 2022; 195:2483-2518. [PMID: 35488955 DOI: 10.1007/s12010-022-03938-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 11/02/2022]
Abstract
Popularity of herbal drugs has always been in high demand, but recently it has been increasing all over the world, especially in India, because of the lower range of adverse health effects as compared to synthetic or man-made drugs. Not only this but their cost-effectiveness and easy availability to the poor people and the masses, particularly in developing countries, are major causes for their demand. But there lies a huge problem during the process of plant collection that affects their medicinal properties to certain degrees. This is caused by heavy metal toxicity in soil in different locations of the Indian subcontinent. This was correlated with their potential to cause health damage. Exposure of humans to heavy metals includes diverse pathways from food to water to consumption and inhalation of polluted air to permanent damage to exposed skin and even by occupational exposure at workplaces. As we can understand, the main mechanisms of heavy metal toxicity include the production of free radicals to affect the host by oxidative stress, damaging biological molecules such as enzymes, proteins, lipids, and even nucleic acids and finally damaging DNA which is the fastest way to carcinogenesis and in addition, neurotoxicity. Therefore, in this paper, we have researched how the plants/herbs are affected due to heavy metal deposition in their habitat and how it can lead to serious clinical complications.
Collapse
Affiliation(s)
- Susmita Mukherjee
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Nivedita Chatterjee
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Asmeeta Sircar
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Shimantika Maikap
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Abhilasha Singh
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Sudeshna Acharyya
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Sonali Paul
- Department of Biotechnology, University of Engineering and Management, Kolkata, India.
| |
Collapse
|
5
|
Kannappan S, Ramisetty BCM. Engineered Whole-Cell-Based Biosensors: Sensing Environmental Heavy Metal Pollutants in Water-a Review. Appl Biochem Biotechnol 2021; 194:1814-1840. [PMID: 34783990 DOI: 10.1007/s12010-021-03734-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/21/2021] [Indexed: 11/27/2022]
Abstract
The frequent exposure and accumulation of heavy metals in organisms cause serious health issues affecting a range of organs such as the brain, liver, and reproductive organs in adults, infants, and children. Several parts of the world have high levels of heavy metals affecting millions of people, costing millions of dollars for improving the potability of water and medical treatment of the affected. Hence, water quality assessment is required to monitor the degree of heavy metal contamination in potable water. In nature, organisms respond to various environmental pollutants such as heavy metals, allowing their survival in a diverse environmental niche. With the advent of recombinant DNA technology, it is now possible to manipulate these natural bioreporters into controlled systems which either turn on or off gene expression or activity of enzymes in the presence of specific heavy metals (compound-specific biosensors) otherwise termed as whole-cell biosensors (WCBs). WCBs provide an upper hand compared to other immunosensors, enzyme-based sensors, and DNA-based sensors since microbes can be relatively easily manipulated, scaled up with relative ease, and can detect only the bioavailable heavy metals. In this review, we summarize the current knowledge of the various mechanisms of toxicity elicited by various heavy metals, thence emphasizing the need to develop heavy metal sensing platforms. Following this, the biosensor-based platforms including WCBs for detecting heavy metals developed thus far have been briefly elaborated upon, emphasizing the challenges and solutions associated with WCBs.
Collapse
Affiliation(s)
- Shrute Kannappan
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea
- Research Center for Advanced Materials Technology, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | | |
Collapse
|
6
|
Carmona A, Roudeau S, Ortega R. Molecular Mechanisms of Environmental Metal Neurotoxicity: A Focus on the Interactions of Metals with Synapse Structure and Function. TOXICS 2021; 9:toxics9090198. [PMID: 34564349 PMCID: PMC8471991 DOI: 10.3390/toxics9090198] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023]
Abstract
Environmental exposure to neurotoxic metals and metalloids such as arsenic, cadmium, lead, mercury, or manganese is a global health concern affecting millions of people worldwide. Depending on the period of exposure over a lifetime, environmental metals can alter neurodevelopment, neurobehavior, and cognition and cause neurodegeneration. There is increasing evidence linking environmental exposure to metal contaminants to the etiology of neurological diseases in early life (e.g., autism spectrum disorder) or late life (e.g., Alzheimer’s disease). The known main molecular mechanisms of metal-induced toxicity in cells are the generation of reactive oxygen species, the interaction with sulfhydryl chemical groups in proteins (e.g., cysteine), and the competition of toxic metals with binding sites of essential metals (e.g., Fe, Cu, Zn). In neurons, these molecular interactions can alter the functions of neurotransmitter receptors, the cytoskeleton and scaffolding synaptic proteins, thereby disrupting synaptic structure and function. Loss of synaptic connectivity may precede more drastic alterations such as neurodegeneration. In this article, we will review the molecular mechanisms of metal-induced synaptic neurotoxicity.
Collapse
|
7
|
Monchanin C, Blanc-Brude A, Drujont E, Negahi MM, Pasquaretta C, Silvestre J, Baqué D, Elger A, Barron AB, Devaud JM, Lihoreau M. Chronic exposure to trace lead impairs honey bee learning. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 212:112008. [PMID: 33578129 DOI: 10.1016/j.ecoenv.2021.112008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
Pollutants can have severe detrimental effects on insects, even at sublethal doses, damaging developmental and cognitive processes involved in crucial behaviours. Agrochemicals have been identified as important causes of pollinator declines, but the impacts of other anthropogenic compounds, such as metallic trace elements in soils and waters, have received considerably less attention. Here, we exposed colonies of the European honey bee Apis mellifera to chronic field-realistic concentrations of lead in food and demonstrated that consumption of this trace element impaired bee cognition and morphological development. Honey bees exposed to the highest of these low concentrations had reduced olfactory learning performances. These honey bees also developed smaller heads, which may have constrained their cognitive functions as we show a general relationship between head size and learning performance. Our results demonstrate that lead pollutants, even at trace levels, can have dramatic effects on honey bee cognitive abilities, potentially altering key colony functions and the pollination service.
Collapse
Affiliation(s)
- Coline Monchanin
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI); CNRS, University Paul Sabatier - Toulouse III, France; Department of Biological Sciences, Macquarie University, NSW, Australia.
| | - Amaury Blanc-Brude
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI); CNRS, University Paul Sabatier - Toulouse III, France
| | - Erwann Drujont
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI); CNRS, University Paul Sabatier - Toulouse III, France
| | - Mohammed Mustafa Negahi
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI); CNRS, University Paul Sabatier - Toulouse III, France
| | - Cristian Pasquaretta
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI); CNRS, University Paul Sabatier - Toulouse III, France
| | - Jérôme Silvestre
- EcoLab, Université de Toulouse, CNRS, INPT, UPS, Toulouse, France
| | - David Baqué
- EcoLab, Université de Toulouse, CNRS, INPT, UPS, Toulouse, France
| | - Arnaud Elger
- EcoLab, Université de Toulouse, CNRS, INPT, UPS, Toulouse, France
| | - Andrew B Barron
- Department of Biological Sciences, Macquarie University, NSW, Australia
| | - Jean-Marc Devaud
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI); CNRS, University Paul Sabatier - Toulouse III, France
| | - Mathieu Lihoreau
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI); CNRS, University Paul Sabatier - Toulouse III, France.
| |
Collapse
|
8
|
Cognitive Impairment Induced by Lead Exposure during Lifespan: Mechanisms of Lead Neurotoxicity. TOXICS 2021; 9:toxics9020023. [PMID: 33525464 PMCID: PMC7912619 DOI: 10.3390/toxics9020023] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/25/2022]
Abstract
Lead (Pb) is considered a strong environmental toxin with human health repercussions. Due to its widespread use and the number of people potentially exposed to different sources of this heavy metal, Pb intoxication is recognized as a public health problem in many countries. Exposure to Pb can occur through ingestion, inhalation, dermal, and transplacental routes. The magnitude of its effects depends on several toxicity conditions: lead speciation, doses, time, and age of exposure, among others. It has been demonstrated that Pb exposure induces stronger effects during early life. The central nervous system is especially vulnerable to Pb toxicity; Pb exposure is linked to cognitive impairment, executive function alterations, abnormal social behavior, and fine motor control perturbations. This review aims to provide a general view of the cognitive consequences associated with Pb exposure during early life as well as during adulthood. Additionally, it describes the neurotoxic mechanisms associated with cognitive impairment induced by Pb, which include neurochemical, molecular, and morphological changes that jointly could have a synergic effect on the cognitive performance.
Collapse
|
9
|
Paithankar JG, Saini S, Dwivedi S, Sharma A, Chowdhuri DK. Heavy metal associated health hazards: An interplay of oxidative stress and signal transduction. CHEMOSPHERE 2021; 262:128350. [PMID: 33182141 DOI: 10.1016/j.chemosphere.2020.128350] [Citation(s) in RCA: 308] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 05/20/2023]
Abstract
Heavy metal-induced cellular and organismal toxicity have become a major health concern in biomedical science. Indiscriminate use of heavy metals in different sectors, such as, industrial-, agricultural-, healthcare-, cosmetics-, and domestic-sectors has contaminated environment matrices and poses a severe health concern. Xenobiotics mediated effect is a ubiquitous cellular response. Oxidative stress is one such prime cellular response, which is the result of an imbalance in the redox system. Further, oxidative stress is associated with macromolecular damages and activation of several cell survival and cell death pathways. Epidemiological as well as laboratory data suggest that oxidative stress-induced cellular response following heavy metal exposure is linked with an increased risk of neoplasm, neurological disorders, diabetes, infertility, developmental disorders, renal failure, and cardiovascular disease. During the recent past, a relation among heavy metal exposure, oxidative stress, and signaling pathways have been explored to understand the heavy metal-induced toxicity. Heavy metal-induced oxidative stress and its connection with different signaling pathways are complicated; therefore, the systemic summary is essential. Herein, an effort has been made to decipher the interplay among heavy metals/metalloids (Arsenic, Chromium, Cadmium, and Lead) exposures, oxidative stress, and signal transduction, which are essential to mount the cellular and organismal response. The signaling pathways involved in this interplay include NF-κB, NRF2, JAK-STAT, JNK, FOXO, and HIF.
Collapse
Affiliation(s)
- Jagdish Gopal Paithankar
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Sanjay Saini
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Shiwangi Dwivedi
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Anurag Sharma
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India.
| | - Debapratim Kar Chowdhuri
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.
| |
Collapse
|
10
|
Iqubal A, Ahmed M, Ahmad S, Sahoo CR, Iqubal MK, Haque SE. Environmental neurotoxic pollutants: review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:41175-41198. [PMID: 32820440 DOI: 10.1007/s11356-020-10539-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/16/2020] [Indexed: 05/23/2023]
Abstract
Environmental pollutants are recognized as one of the major concerns for public health and responsible for various forms of neurological disorders. Some of the common sources of environmental pollutants related to neurotoxic manifestations are industrial waste, pesticides, automobile exhaust, laboratory waste, and burning of terrestrial waste. Among various environmental pollutants, particulate matter, ultrafine particulate matter, nanoparticles, and lipophilic vaporized toxicant (acrolein) easily cross the blood-brain barrier, activate innate immune responses in the astrocytes, microglia, and neurons, and exert neurotoxicity. Growing shreds of evidence from human epidemiological studies have correlated the environmental pollutants with neuroinflammation, oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, myelin sheath disruption, and alterations in the blood-brain barrier anatomy leading to cognitive dysfunction and poor quality of life. These environmental pollutants also considerably cause developmental neurotoxicity, exhibit teratogenic effect and mental growth retardance, and reduce IQ level. Until now, the exact mechanism of pollutant-induced neurotoxicity is not known, but studies have shown interference of pollutants with the endogenous antioxidant defense system, inflammatory pathway (Nrf2/NF-kB, MAPKs/PI3K, and Akt/GSK3β), modulation of neurotransmitters, and reduction in long-term potentiation. In the current review, various sources of pollutants and exposure to the human population, developmental neurotoxicity, and molecular mechanism of different pollutants involved in the pathogenesis of different neurological disorders have been discussed.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Musheer Ahmed
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Shahnawaz Ahmad
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Chita Ranjan Sahoo
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
11
|
Zou RX, Gu X, Ding JJ, Wang T, Bi N, Niu K, Ge M, Chen XT, Wang HL. Pb exposure induces an imbalance of excitatory and inhibitory synaptic transmission in cultured rat hippocampal neurons. Toxicol In Vitro 2019; 63:104742. [PMID: 31785328 DOI: 10.1016/j.tiv.2019.104742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/12/2019] [Accepted: 11/24/2019] [Indexed: 12/16/2022]
Abstract
An appropriate balance of excitatory and inhibitory synapse maintains the network stability of the central nervous system. Our recent work showed lead (Pb) exposure can inhibit synaptic transmission in cultured hippocampal neurons. However, it is not clear whether Pb exposure disrupt the balance of excitatory and inhibitory synaptic transmission. Here, primary cultured hippocampal neurons from Sprague-Dawley (SD) rats were exposed to Pb (0.2 μM, 1 μM, 5 μM, respectively) from Days in Vitro (DIV) 7 to DIV 12 for 5 days and the excitatory and inhibitory synaptic transmission was examined. Patch clamp recording results showed that distinct from exposures of 0.2 μM and 5 μM, 1 μM Pb exposure significantly increased the mIPSC frequency and decreased the mEPSC frequency, leading to a uniform inhibitory outcome. Further, the number of inhibitory presynaptic puncta was significantly increased after 1 μM Pb exposure, while the number of excitatory presynaptic terminals was decreased. In addition 1 μM Pb increased the glutamic acid decarboxylase (GAD65) expression and the surface GABAA receptor (GABAAR) clusters. This shift might potentiate the synthesis of GABA and enhance the surface distribution of postsynaptic GABAAR clusters in hippocampus neurons. Together, these data showed that Pb exposure disrupted the balance of excitatory and inhibitory synaptic transmission via abnormal GABAergic neurotransmission.
Collapse
Affiliation(s)
- Rong-Xin Zou
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Xiaozhen Gu
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Jin-Jun Ding
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Tiandong Wang
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Nanxi Bi
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Kang Niu
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Mengmeng Ge
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Xiang-Tao Chen
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230031, PR China.
| | - Hui-Li Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China.
| |
Collapse
|
12
|
Wang T, Zhu H, Hou Y, Gu W, Wu H, Luan Y, Xiao C, Zhou C. Galantamine reversed early postoperative cognitive deficit via alleviating inflammation and enhancing synaptic transmission in mouse hippocampus. Eur J Pharmacol 2018; 846:63-72. [PMID: 30586550 DOI: 10.1016/j.ejphar.2018.12.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 12/20/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is commonly seen in patients undergoing major surgeries and may persist. Although neuroinflammation is one of the important contributors to the development of POCD, the mechanisms underlying POCD remain unclear. We performed stabilized tibial fracture operation in male mice. In comparison with sham mice (anesthesia only), the surgery mice exhibited cognitive deficits in a fear conditioning paradigm at postsurgery day 3-7, and increased numbers of microglia and elevated levels of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) without change of anti-inflammatory cytokines (IL-4 and IL-10) in the hippocampus. Electrophysiological recordings from CA1 hippocampal neurons revealed that POCD mice exhibited impairment in AMPA receptor-mediated evoked excitatory postsynaptic currents (eEPSCs) without alteration in the rectification property of AMPA receptors. Interestingly, daily intraperitoneal administration of galantamine, an inhibitor of acetylcholinesterase, reversed cognitive dysfunction in surgery mice and attenuated accumulation of microglia and protein levels of IL-1β, IL-6 and TNF-α in the hippocampus. Additionally, galantamine potentiated AMPA receptor-mediated eEPSCs in the hippocampus more prominent in surgery mice than in sham mice. Therefore, enhancement of cholinergic tone in the hippocampus might be a therapeutic strategy for early POCD in terms of suppression of inflammation and normalization of excitatory synaptic transmission.
Collapse
Affiliation(s)
- Tianhai Wang
- Department of Anesthesiology, The third hospital, affiliated to the Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hongge Zhu
- Department of Second Pulmonary Medicine, The third hospital, affiliated to the Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yanshen Hou
- Department of Anesthesiology, The third hospital, affiliated to the Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Weixin Gu
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Haichuan Wu
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yiwen Luan
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cheng Xiao
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chunyi Zhou
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
13
|
Sachana M, Rolaki A, Bal-Price A. Development of the Adverse Outcome Pathway (AOP): Chronic binding of antagonist to N-methyl-d-aspartate receptors (NMDARs) during brain development induces impairment of learning and memory abilities of children. Toxicol Appl Pharmacol 2018; 354:153-175. [PMID: 29524501 PMCID: PMC6095943 DOI: 10.1016/j.taap.2018.02.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 01/06/2023]
Abstract
The Adverse Outcome Pathways (AOPs) are designed to provide mechanistic understanding of complex biological systems and pathways of toxicity that result in adverse outcomes (AOs) relevant to regulatory endpoints. AOP concept captures in a structured way the causal relationships resulting from initial chemical interaction with biological target(s) (molecular initiating event) to an AO manifested in individual organisms and/or populations through a sequential series of key events (KEs), which are cellular, anatomical and/or functional changes in biological processes. An AOP provides the mechanistic detail required to support chemical safety assessment, the development of alternative methods and the implementation of an integrated testing strategy. An example of the AOP relevant to developmental neurotoxicity (DNT) is described here following the requirements of information defined by the OECD Users' Handbook Supplement to the Guidance Document for developing and assessing AOPs. In this AOP, the binding of an antagonist to glutamate receptor N-methyl-d-aspartate (NMDAR) receptor is defined as MIE. This MIE triggers a cascade of cellular KEs including reduction of intracellular calcium levels, reduction of brain derived neurotrophic factor release, neuronal cell death, decreased glutamate presynaptic release and aberrant dendritic morphology. At organ level, the above mentioned KEs lead to decreased synaptogenesis and decreased neuronal network formation and function causing learning and memory deficit at organism level, which is defined as the AO. There are in vitro, in vivo and epidemiological data that support the described KEs and their causative relationships rendering this AOP relevant to DNT evaluation in the context of regulatory purposes.
Collapse
Affiliation(s)
| | | | - Anna Bal-Price
- European Commission, Joint Research Centre, Ispra, Italy.
| |
Collapse
|
14
|
Gorkhali R, Huang K, Kirberger M, Yang JJ. Defining potential roles of Pb(2+) in neurotoxicity from a calciomics approach. Metallomics 2017; 8:563-78. [PMID: 27108875 DOI: 10.1039/c6mt00038j] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metal ions play crucial roles in numerous biological processes, facilitating biochemical reactions by binding to various proteins. An increasing body of evidence suggests that neurotoxicity associated with exposure to nonessential metals (e.g., Pb(2+)) involves disruption of synaptic activity, and these observed effects are associated with the ability of Pb(2+) to interfere with Zn(2+) and Ca(2+)-dependent functions. However, the molecular mechanism behind Pb(2+) toxicity remains a topic of debate. In this review, we first discuss potential neuronal Ca(2+) binding protein (CaBP) targets for Pb(2+) such as calmodulin (CaM), synaptotagmin, neuronal calcium sensor-1 (NCS-1), N-methyl-d-aspartate receptor (NMDAR) and family C of G-protein coupled receptors (cGPCRs), and their involvement in Ca(2+)-signalling pathways. We then compare metal binding properties between Ca(2+) and Pb(2+) to understand the structural implications of Pb(2+) binding to CaBPs. Statistical and biophysical studies (e.g., NMR and fluorescence spectroscopy) of Pb(2+) binding are discussed to investigate the molecular mechanism behind Pb(2+) toxicity. These studies identify an opportunistic, allosteric binding of Pb(2+) to CaM, which is distinct from ionic displacement. Together, these data suggest three potential modes of Pb(2+) activity related to molecular and/or neural toxicity: (i) Pb(2+) can occupy Ca(2+)-binding sites, inhibiting the activity of the protein by structural modulation, (ii) Pb(2+) can mimic Ca(2+) in the binding sites, falsely activating the protein and perturbing downstream activities, or (iii) Pb(2+) can bind outside of the Ca(2+)-binding sites, resulting in the allosteric modulation of the protein activity. Moreover, the data further suggest that even low concentrations of Pb(2+) can interfere at multiple points within the neuronal Ca(2+) signalling pathways to cause neurotoxicity.
Collapse
Affiliation(s)
- Rakshya Gorkhali
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| | - Kenneth Huang
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| | - Michael Kirberger
- Department of Chemistry and Physics, Clayton State University, Morrow, GA 30260, USA.
| | - Jenny J Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| |
Collapse
|
15
|
Lead poisoning: acute exposure of the heart to lead ions promotes changes in cardiac function and Cav1.2 ion channels. Biophys Rev 2017; 9:807-825. [PMID: 28836190 DOI: 10.1007/s12551-017-0303-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/28/2017] [Indexed: 01/02/2023] Open
Abstract
Lead ions (Pb2+) possess characteristics similar to Ca2+. Because of this and its redox capabilities, lead causes different toxic effects. The neurotoxic effects have been well documented; however, the toxic effects on cardiac tissues remain allusive. We utilized isolated guinea pig hearts and measured the effects of Pb2+ on their contractility and excitability. Acute exposure to extracellular Pb2+ had a negative inotropic effect and increased diastolic tension. The speed of contraction and relaxation were affected, though the effects were more dramatic on the speed of contraction. Excitability was also altered. Heart beat frequency increased and later diminished after lead ion exposure. Pro-arrhytmic events, such as early after-depolarization and a reduction of the action potential plateau, were also observed. In isolated cardiomyocytes and tsA 201 cells, extracellular lead blocked currents through Cav1.2 channels, diminished their activation, and enhanced their fast inactivation, negatively affecting their gating currents. Thus, Pb2+ was cardiotoxic and reduced cardiac contractility, making the heart prone to arrhythmias. This was due, in part, to Pb2+ effects on the Cav1.2 channels; however, other channels, transporters or pathways may also be involved. Acute cardiotoxic effects were observed at Pb2+ concentrations achievable during acute lead poisoning. The results suggest how Cav1.2 gating can be affected by divalent cations, such as Pb2, and also suggest a more thorough evaluation of heart function in individuals affected by lead poisoning.
Collapse
|
16
|
Benammi H, Erazi H, El Hiba O, Vinay L, Bras H, Viemari JC, Gamrani H. Disturbed sensorimotor and electrophysiological patterns in lead intoxicated rats during development are restored by curcumin I. PLoS One 2017; 12:e0172715. [PMID: 28267745 PMCID: PMC5340392 DOI: 10.1371/journal.pone.0172715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 02/08/2017] [Indexed: 01/01/2023] Open
Abstract
Lead poisoning is one of the most significant health problem of environmental origin. It is known to cause different damages in the central and peripheral nervous system which could be represented by several neurophysiological and behavioral symptoms. In this study we firstly investigated the effect of lead prenatal exposure in rats to (3g/L), from neonatal to young age, on the motor/sensory performances, excitability of the spinal cord and gaits during development. Then we evaluated neuroprotective effects of curcumin I (Cur I) against lead neurotoxicity, by means of grasping and cliff avoidance tests to reveal the impairment of the sensorimotor functions in neonatal rats exposed prenatally to lead. In addition, extracellular recordings of motor output in spinal cord revealed an hyper-excitability of spinal networks in lead treated rats. The frequency of induced fictive locomotion was also increased in treated rats. At the young age, rats exhibited an impaired locomotor gait. All those abnormalities were attenuated by Cur I treatment at a dose of 16g/kg. Based on our finding, Cur I has shown features of a potent chemical compound able to restore the neuronal and the relative locomotor behaviors disturbances induced by lead intoxication. Therefore, this chemical can be recommended as a new therapeutic trial against lead induced neurotoxicity.
Collapse
Affiliation(s)
- Hind Benammi
- Neuroscience, Pharmacology and Environment Team, faculty of Science Semlalia, Cadi Ayyad University, Marrakech, Morocco
| | - Hasna Erazi
- Neuroscience, Pharmacology and Environment Team, faculty of Science Semlalia, Cadi Ayyad University, Marrakech, Morocco
| | - Omar El Hiba
- Neuroscience, Pharmacology and Environment Team, faculty of Science Semlalia, Cadi Ayyad University, Marrakech, Morocco
- Department of Biology, faculty of Sciences, Chouaib Doukkali University, EL Jadida, Morocco
| | - Laurent Vinay
- Institut de Neurosciences de la Timone, Unité Mixte de Recherche 7289, CNRS, Université Aix-Marseille, Marseille, France
| | - Hélène Bras
- Institut de Neurosciences de la Timone, Unité Mixte de Recherche 7289, CNRS, Université Aix-Marseille, Marseille, France
| | - Jean-Charles Viemari
- Institut de Neurosciences de la Timone, Unité Mixte de Recherche 7289, CNRS, Université Aix-Marseille, Marseille, France
- * E-mail: (HG); (JCV)
| | - Halima Gamrani
- Neuroscience, Pharmacology and Environment Team, faculty of Science Semlalia, Cadi Ayyad University, Marrakech, Morocco
- * E-mail: (HG); (JCV)
| |
Collapse
|
17
|
Łukomska A, Baranowska-Bosiacka I, Budkowska M, Pilutin A, Tarnowski M, Dec K, Dołęgowska B, Metryka E, Chlubek D, Gutowska I. The effect of low levels of lead (Pb) in the blood on levels of sphingosine-1-phosphate (S1P) and expression of S1P receptor 1 in the brain of the rat in the perinatal period. CHEMOSPHERE 2017; 166:221-229. [PMID: 27697711 DOI: 10.1016/j.chemosphere.2016.09.067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/31/2016] [Accepted: 09/15/2016] [Indexed: 06/06/2023]
Abstract
Sphingolipids are the main components of the lipid membrane. They also perform structural functions and participate in many signal transmission processes. One of the bioactive sphingolipids is sphingosine-1-phosphate (S1P), a ligand for five G protein-coupled receptors (S1PRs1-5), which can also act as an intracellular second messenger. S1P is responsible for the stimulation of progenitor cells in the brain, but it can also induce apoptosis of mature neurons. This study is aimed at assessing the effect of pre- and neonatal exposure to permissible Pb concentrations on S1P levels and S1PR1 (EDG1) expression in the prefrontal cortex, cerebellum, and hippocampus of rats. The concentrations of S1P were determined by RP-HPLC, S1PR1 expression was determined by RT PCR and Western Blot, and receptor immunolocalization was determined by immunohistochemistry method. Our results showed that even low blood Pb concentrations, i.e. within the acceptable limit of 10 μg/dL caused changes in the concentration of S1P in the cerebellum, prefrontal cortex, and hippocampus. Our data also showed a significant decrease in the level of S1PR1 in all studied part of brain, without significant changes in S1PR1 gene expression. Pre- and neonatal exposure to Pb also resulted in a decrease in the expression of S1PR1 in glial cells in all regions of the Cornu Ammonis (CA1-CA4) and Dentate Gyrus in the hippocampus, as well as in all layers of the cerebellum and prefrontal cortex, compared to the unexposed control group.
Collapse
Affiliation(s)
- A Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland
| | - I Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland.
| | - M Budkowska
- Department of Microbiology and Immunology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - A Pilutin
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - M Tarnowski
- Department of Physiology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - K Dec
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland
| | - B Dołęgowska
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - E Metryka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - D Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - I Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland
| |
Collapse
|
18
|
Gąssowska M, Baranowska-Bosiacka I, Moczydłowska J, Frontczak-Baniewicz M, Gewartowska M, Strużyńska L, Gutowska I, Chlubek D, Adamczyk A. Perinatal exposure to lead (Pb) induces ultrastructural and molecular alterations in synapses of rat offspring. Toxicology 2016; 373:13-29. [DOI: 10.1016/j.tox.2016.10.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/22/2016] [Accepted: 10/27/2016] [Indexed: 12/26/2022]
|
19
|
Guariglia SR, Stansfield KH, McGlothan J, Guilarte TR. Chronic early life lead (Pb 2+) exposure alters presynaptic vesicle pools in hippocampal synapses. BMC Pharmacol Toxicol 2016; 17:56. [PMID: 27802838 PMCID: PMC5090882 DOI: 10.1186/s40360-016-0098-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/14/2016] [Indexed: 12/23/2022] Open
Abstract
Background Lead (Pb2+) exposure has been shown to impair presynaptic neurotransmitter release in both in vivo and in vitro model systems. The mechanism by which Pb2+ impairs neurotransmitter release has not been fully elucidated. In previous work, we have shown that Pb2+ exposure inhibits vesicular release and reduces the number of fast-releasing sites in cultured hippocampal neurons. We have also shown that Pb2+ exposure inhibits vesicular release and alters the distribution of presynaptic vesicles in Shaffer Collateral – CA1 synapses of rodents chronically exposed to Pb2+ during development. Methods In the present study, we used transmission electron microscopy to examine presynaptic vesicle pools in Mossy Fiber-CA3 synapses and in Perforant Path-Dentate Gyrus synapses of rats to determine if in vivo Pb2+ exposure altered presynaptic vesicle distribution in these hippocampal regions. Data were analyzed using T-test for each experimental endpoint. Results We found that Pb2+ exposure significantly reduced the number of vesicles in the readily releasable pool and recycling pool in Mossy Fiber-CA3 terminals. In both Mossy Fiber-CA3 terminals and in Perforant Path-Dentate Gyrus terminals, Pb2+ exposure significantly increased vesicle nearest neighbor distance in all vesicular pools (Rapidly Releasable, Recycling and Resting). We also found a reduction in the size of the postsynaptic densities of CA3 dendrites in the Pb2+ exposed group. Conclusions In our previous work, we have demonstrated that Pb2+ exposure impairs vesicular release in Shaffer Collateral - CA1 terminals of the hippocampus and that the number of docked vesicles in the presynaptic active zone was reduced. Our current data shows that Pb2+ exposure reduces the number of vesicles that are in proximity to release sites in Mossy Fiber- CA3 terminals. Furthermore, Pb2+ exposure causes presynaptic vesicles to be further from one another, in both Mossy Fiber- CA3 terminals and in Perforant Pathway – Dentate Gyrus terminals, which may interfere with vesicle movement and release. Our findings provide a novel in vivo mechanism by which Pb2+ exposure impairs vesicle dynamics and release in the hippocampus.
Collapse
Affiliation(s)
- Sara Rose Guariglia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, NY, 10032, USA
| | - Kirstie H Stansfield
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, NY, 10032, USA
| | - Jennifer McGlothan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, NY, 10032, USA
| | - Tomas R Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
20
|
Sanders T, Liu YM, Tchounwou PB. Cytotoxic, genotoxic, and neurotoxic effects of Mg, Pb, and Fe on pheochromocytoma (PC-12) cells. ENVIRONMENTAL TOXICOLOGY 2015; 30:1445-58. [PMID: 24942330 PMCID: PMC4270943 DOI: 10.1002/tox.22014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/30/2014] [Accepted: 05/31/2014] [Indexed: 05/07/2023]
Abstract
Metals such as lead (Pb), magnesium (Mg), and iron (Fe) are ubiquitous in the environment as a result of natural occurrence and anthropogenic activities. Although Mg, Fe, and others are considered essential elements, high level of exposure has been associated with severe adverse health effects including cardiovascular, hematological, nephrotoxic, hepatotoxic, and neurologic abnormalities in humans. In the present study we hypothesized that Mg, Pb, and Fe are cytotoxic, genotoxic and neurotoxic, and their toxicity is mediated through oxidative stress and alteration in protein expression. To test the hypothesis, we used the pheochromocytoma (PC-12) cell line as a neuro cell model and performed the LDH assay for cell viability, Comet assay for DNA damage, Western blot for oxidative stress, and HPLC-MS to assess the concentration levels of neurological biomarkers such as glutamate, dopamine (DA), and 3-methoxytyramine (3-MT). The results of this study clearly show that Mg, Pb, and Fe, respectively in the form of MgSO4 , Pb(NO3 )2 , FeCl2 , and FeCl3 induce cytotoxicity, oxidative stress, and genotoxicity in PC-12 cells. In addition, exposure to these metallic compounds caused significant changes in the concentration levels of glutamate, dopamine, and 3-MT in PC-12 cells. Taken together the findings suggest that MgSO4 , Pb(NO3 )2 , FeCl2 , and FeCl3 have the potential to induce substantial toxicity to PC-12 cells.
Collapse
Affiliation(s)
- Talia Sanders
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, 39217, USA
| | - Yi-Ming Liu
- Bioanalytical Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, 39217, USA
| | - Paul B Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, 39217, USA
| |
Collapse
|
21
|
Zhang XL, Guariglia SR, McGlothan JL, Stansfield KH, Stanton PK, Guilarte TR. Presynaptic mechanisms of lead neurotoxicity: effects on vesicular release, vesicle clustering and mitochondria number. PLoS One 2015; 10:e0127461. [PMID: 26011056 PMCID: PMC4444102 DOI: 10.1371/journal.pone.0127461] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/15/2015] [Indexed: 01/12/2023] Open
Abstract
Childhood lead (Pb2+) intoxication is a global public health problem and accounts for 0.6% of the global burden of disease associated with intellectual disabilities. Despite the recognition that childhood Pb2+ intoxication contributes significantly to intellectual disabilities, there is a fundamental lack of knowledge on presynaptic mechanisms by which Pb2+ disrupts synaptic function. In this study, using a well-characterized rodent model of developmental Pb2+ neurotoxicity, we show that Pb2+ exposure markedly inhibits presynaptic vesicular release in hippocampal Schaffer collateral-CA1 synapses in young adult rats. This effect was associated with ultrastructural changes which revealed a reduction in vesicle number in the readily releasable/docked vesicle pool, disperse vesicle clusters in the resting pool, and a reduced number of presynaptic terminals with multiple mitochondria with no change in presynaptic calcium influx. These studies provide fundamental knowledge on mechanisms by which Pb2+ produces profound inhibition of presynaptic vesicular release that contribute to deficits in synaptic plasticity and intellectual development.
Collapse
Affiliation(s)
- Xiao-lei Zhang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - Sara R. Guariglia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Jennifer L. McGlothan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Kirstie H. Stansfield
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Patric K. Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - Tomás R. Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
22
|
Qu CL, Huo FQ, Huang FS, Tang JS. Activation of mu-opioid receptors in the ventrolateral orbital cortex inhibits the GABAergic miniature inhibitory postsynaptic currents in rats. Neurosci Lett 2015; 592:64-9. [DOI: 10.1016/j.neulet.2015.02.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/09/2015] [Accepted: 02/18/2015] [Indexed: 01/09/2023]
|
23
|
Parkia biglobosa improves mitochondrial functioning and protects against neurotoxic agents in rat brain hippocampal slices. BIOMED RESEARCH INTERNATIONAL 2014; 2014:326290. [PMID: 25177688 PMCID: PMC4142556 DOI: 10.1155/2014/326290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/26/2014] [Accepted: 06/28/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Methanolic leaf extracts of Parkia biglobosa, PBE, and one of its major polyphenolic constituents, catechin, were investigated for their protective effects against neurotoxicity induced by different agents on rat brain hippocampal slices and isolated mitochondria. METHODS Hippocampal slices were preincubated with PBE (25, 50, 100, or 200 µg/mL) or catechin (1, 5, or 10 µg/mL) for 30 min followed by further incubation with 300 µM H2O2, 300 µM SNP, or 200 µM PbCl2 for 1 h. Effects of PBE and catechin on SNP- or CaCl2-induced brain mitochondrial ROS formation and mitochondrial membrane potential (ΔΨm) were also determined. RESULTS PBE and catechin decreased basal ROS generation in slices and blunted the prooxidant effects of neurotoxicants on membrane lipid peroxidation and nonprotein thiol contents. PBE rescued hippocampal cellular viability from SNP damage and caused a significant boost in hippocampus Na(+), K(+)-ATPase activity but with no effect on the acetylcholinesterase activity. Both PBE and catechin also mitigated SNP- or CaCl2-dependent mitochondrial ROS generation. Measurement by safranine fluorescence however showed that the mild depolarization of the ΔΨm by PBE was independent of catechin. CONCLUSION The results suggest that the neuroprotective effect of PBE is dependent on its constituent antioxidants and mild mitochondrial depolarization propensity.
Collapse
|
24
|
Abstract
Human exposure to neurotoxic metals is a global public health problem. Metals which cause neurological toxicity, such as lead (Pb) and manganese (Mn), are of particular concern due to the long-lasting and possibly irreversible nature of their effects. Pb exposure in childhood can result in cognitive and behavioural deficits in children. These effects are long-lasting and persist into adulthood even after Pb exposure has been reduced or eliminated. While Mn is an essential element of the human diet and serves many cellular functions in the human body, elevated Mn levels can result in a Parkinson's disease (PD)-like syndrome and developmental Mn exposure can adversely affect childhood neurological development. Due to the ubiquitous presence of both metals, reducing human exposure to toxic levels of Mn and Pb remains a world-wide public health challenge. In this review we summarize the toxicokinetics of Pb and Mn, describe their neurotoxic mechanisms, and discuss common themes in their neurotoxicity.
Collapse
Affiliation(s)
| | - Tomas R Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
25
|
Luo W, Ruan D, Yan C, Yin S, Chen J. Effects of chronic lead exposure on functions of nervous system in Chinese children and developmental rats. Neurotoxicology 2012; 33:862-71. [DOI: 10.1016/j.neuro.2012.03.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 03/09/2012] [Accepted: 03/20/2012] [Indexed: 01/23/2023]
|
26
|
Smith SM, Chen W, Vyleta NP, Williams C, Lee CH, Phillips C, Andresen MC. Calcium regulation of spontaneous and asynchronous neurotransmitter release. Cell Calcium 2012; 52:226-33. [PMID: 22748761 DOI: 10.1016/j.ceca.2012.06.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 05/23/2012] [Accepted: 06/01/2012] [Indexed: 11/28/2022]
Abstract
The molecular machinery underlying action potential-evoked, synchronous neurotransmitter release, has been intensely studied. It was presumed that two other forms of exocytosis, delayed (asynchronous) and spontaneous transmission, were mediated by the same voltage-activated Ca(2+) channels (VACCs), intracellular Ca(2+) sensors and vesicle pools. However, a recent explosion in the study of spontaneous and asynchronous release has shown these presumptions to be incorrect. Furthermore, the finding that different forms of synaptic transmission may mediate distinct physiological functions emphasizes the importance of identifying the mechanisms by which Ca(2+) regulates spontaneous and asynchronous release. In this article, we will briefly summarize new and published data on the role of Ca(2+) in regulating spontaneous and asynchronous release at a number of different synapses. We will discuss how an increase of extracellular [Ca(2+)] increases spontaneous and asynchronous release, show that VACCs are involved at only some synapses, and identify regulatory roles for other ion channels and G protein-coupled receptors. In particular, we will focus on two novel pathways that play important roles in the regulation of non-synchronous release at two exemplary synapses: one modulated by the Ca(2+)-sensing receptor and the other by transient receptor potential cation channel sub-family V member 1.
Collapse
Affiliation(s)
- Stephen M Smith
- Division of Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, OR 97239, United States.
| | | | | | | | | | | | | |
Collapse
|
27
|
Borisova T, Krisanova N, Sivko R, Kasatkina L, Borysov A, Griffin S, Wireman M. Presynaptic malfunction: the neurotoxic effects of cadmium and lead on the proton gradient of synaptic vesicles and glutamate transport. Neurochem Int 2011; 59:272-9. [PMID: 21672571 DOI: 10.1016/j.neuint.2011.05.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 05/19/2011] [Accepted: 05/26/2011] [Indexed: 11/17/2022]
Abstract
Exposure to Cd(2+) and Pb(2+) has neurotoxic consequences for human health and may cause neurodegeneration. The study focused on the analysis of the presynaptic mechanisms underlying the neurotoxic effects of non-essential heavy metals Cd(2+) and Pb(2+). It was shown that the preincubation of rat brain nerve terminals with Cd(2+) (200 μM) or Pb(2+) (200 μM) resulted in the attenuation of synaptic vesicles acidification, which was assessed by the steady state level of the fluorescence of pH-sensitive dye acridine orange. A decrease in L-[(14)C]glutamate accumulation in digitonin-permeabilized synaptosomes after the addition of the metals, which reflected lowered L-[(14)C]glutamate accumulation by synaptic vesicles inside of synaptosomes, may be considered in the support of the above data. Using isolated rat brain synaptic vesicles, it was found that 50 μM Cd(2+) or Pb(2+) caused dissipation of their proton gradient, whereas the application of essential heavy metal Mn(2+) did not do it within the range of the concentration of 50-500 μM. Thus, synaptic malfunction associated with the influence of Cd(2+) and Pb(2+) may result from partial dissipation of the synaptic vesicle proton gradient that leads to: (1) a decrease in stimulated exocytosis, which is associated not only with the blockage of voltage-gated Ca(2+) channels, but also with incomplete filling of synaptic vesicles; (2) an attenuation of Na(+)-dependent glutamate uptake.
Collapse
Affiliation(s)
- Tatiana Borisova
- The Department of Neurochemistry, Palladin Institute of Biochemistry, NAS of Ukraine, 9 Leontovicha Street, Kiev 01601, Ukraine
| | | | | | | | | | | | | |
Collapse
|
28
|
Neal AP, Guilarte TR. Molecular neurobiology of lead (Pb(2+)): effects on synaptic function. Mol Neurobiol 2010; 42:151-60. [PMID: 21042954 PMCID: PMC3076195 DOI: 10.1007/s12035-010-8146-0] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 10/13/2010] [Indexed: 12/16/2022]
Abstract
Lead (Pb(2+)) is a ubiquitous environmental neurotoxicant that continues to threaten public health on a global scale. Epidemiological studies have demonstrated detrimental effects of Pb(2+) on childhood IQ at very low levels of exposure. Recently, a mechanistic understanding of how Pb(2+) affects brain development has begun to emerge. The cognitive effects of Pb(2+) exposure are believed to be mediated through its selective inhibition of the N-methyl-D: -aspartate receptor (NMDAR). Studies in animal models of developmental Pb(2+) exposure exhibit altered NMDAR subunit ontogeny and disruption of NMDAR-dependent intracellular signaling. Additional studies have reported that Pb(2+) exposure inhibits presynaptic calcium (Ca(2+)) channels and affects presynaptic neurotransmission, but a mechanistic link between presynaptic and postsynaptic effects has been missing. Recent work has suggested that the presynaptic and postsynaptic effects of Pb(2+) exposure are both due to inhibition of the NMDAR by Pb(2+), and that the presynaptic effects of Pb(2+) may be mediated by disruption of NMDAR activity-dependent signaling of brain-derived neurotrophic factor (BDNF). These findings provide the basis for the first working model to describe the effects of Pb(2+) exposure on synaptic function. Here, we review the neurotoxic effects of Pb(2+) exposure and discuss the known effects of Pb(2+) exposure in light of these recent findings.
Collapse
Affiliation(s)
- April P. Neal
- Department of Pharmacology and Toxicology, Michigan State University, B307 Life Sciences Building, East Lansing, MI 48824, USA
| | - Tomás R. Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 600 Haven Avenue, B1-108, New York, NY 10032, USA
| |
Collapse
|
29
|
Neal AP, Stansfield KH, Worley PF, Thompson RE, Guilarte TR. Lead exposure during synaptogenesis alters vesicular proteins and impairs vesicular release: potential role of NMDA receptor-dependent BDNF signaling. Toxicol Sci 2010; 116:249-63. [PMID: 20375082 DOI: 10.1093/toxsci/kfq111] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Lead (Pb(2+)) exposure is known to affect presynaptic neurotransmitter release in both in vivo and cell culture models. However, the precise mechanism by which Pb(2+) impairs neurotransmitter release remains unknown. In the current study, we show that Pb(2+) exposure during synaptogenesis in cultured hippocampal neurons produces the loss of synaptophysin (Syn) and synaptobrevin (Syb), two proteins involved in vesicular release. Pb(2+) exposure also increased the number of presynaptic contact sites. However, many of these putative presynaptic contact sites lack Soluble NSF attachment protein receptor complex proteins involved in vesicular exocytosis. Analysis of vesicular release using FM 1-43 dye confirmed that Pb(2+) exposure impaired vesicular release and reduced the number of fast-releasing sites. Because Pb(2+) is a potent N-methyl-D-aspartate receptor (NMDAR) antagonist, we tested the hypothesis that NMDAR inhibition may be producing the presynaptic effects. We show that NMDAR inhibition by aminophosphonovaleric acid mimics the presynaptic effects of Pb(2+) exposure. NMDAR activity has been linked to the signaling of the transsynaptic neurotrophin brain-derived neurotrophic factor (BDNF), and we observed that both the cellular expression of proBDNF and release of BDNF were decreased during the same period of Pb(2+) exposure. Furthermore, exogenous addition of BDNF rescued the presynaptic effects of Pb(2+). We suggest that the presynaptic deficits resulting from Pb(2+) exposure during synaptogenesis are mediated by disruption of NMDAR-dependent BDNF signaling.
Collapse
Affiliation(s)
- April P Neal
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
30
|
Kwieciński A, Nowak P. Gestational manganese intoxication and anxiolytic-like effects of diazepam and the 5-HT1A receptor agonist 8-OH-DPAT in male Wistar rats. Pharmacol Rep 2009; 61:1061-8. [DOI: 10.1016/s1734-1140(09)70168-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 10/29/2009] [Indexed: 10/25/2022]
|
31
|
Cao XJ, Wang M, Chen WH, Zhu DM, She JQ, Ruan DY. Effects of chronic administration of melatonin on spatial learning ability and long-term potentiation in lead-exposed and control rats. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2009; 22:70-75. [PMID: 19462691 DOI: 10.1016/s0895-3988(09)60025-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
OBJECTIVE To explore the changes in spatial learning performance and long-term potentiation (LTP) which is recognized as a component of the cellular basis of learning and memory in normal and lead-exposed rats after administration of melatonin (MT) for two months. METHODS Experiment was performed in adult male Wistar rats (12 controls, 12 exposed to melatonin treatment, 10 exposed to lead and 10 exposed to lead and melatonin treatment). The lead-exposed rats received 0.2% lead acetate solution from their birth day while the control rats drank tap water. Melatonin (3 mg/kg) or vehicle was administered to the control and lead-exposed rats from the time of their weaning by gastric gavage each day for 60 days, depending on their groups. At the age of 81-90 days, all the animals were subjected to Morris water maze test and then used for extracellular recording of LTP in the dentate gyrus (DG) area of the hippocampus in vivo. RESULTS Low dose of melatonin given from weaning for two months impaired LTP in the DG area of hippocampus and induced learning and memory deficit in the control rats. When melatonin was administered over a prolonged period to the lead-exposed rats, it exacerbated LTP impairment, learning and memory deficit induced by lead. CONCLUSION Melatonin is not suitable for normal and lead-exposed children.
Collapse
Affiliation(s)
- Xiu-Jing Cao
- School of Life Science, University of Science and Technology of China, Hefei 230027, Anhui, China
| | | | | | | | | | | |
Collapse
|
32
|
Fu M, Sun ZH, Zong M, He XP, Zuo HC, Xie ZP. Deoxyschisandrin modulates synchronized Ca2+ oscillations and spontaneous synaptic transmission of cultured hippocampal neurons. Acta Pharmacol Sin 2008; 29:891-8. [PMID: 18664321 DOI: 10.1111/j.1745-7254.2008.00821.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Deoxyschisandrin is one of the most effective composites of Schisandra chinensis, a famous Chinese medicine widely used as an antistress, anti-aging, and neurological performance-improving herb. In this study, we examined its specific mechanisms of action on cultured hippocampal neurons. METHODS Hippocampal neurons, primarily cultured for 9-11 d in vitro, were used for this study. DS were dissolved in DMSO and applied to calcium imaging and whole-cell patch clamp. RESULTS The application of 3 mg/L DS decreased the frequency of spontaneous and synchronous oscillations of intracellular Ca2+ to 72%+/-2% (mean+/-SEM), and the spontaneous inhibitory postsynaptic currents to 60%+/-3% (mean+/-SEM). The inhibitory concentration 50% (IC50) for the effect of DS on calcium oscillations was 3.8 mg/L. DS also depressed the high voltage-gated Ca2+ channel and the voltage-gated Na+ channel currents at the same time point. It had no effect, however, on voltage-gated K+ and spontaneous excitatory postsynaptic currents. CONCLUSION DS inhibited the spontaneous and synchronous oscillations of intracellular Ca2+ through the depression of influx of extracellular calcium and the initiation of action potential. By repressing the spontaneous neurotransmitter release, DS modulated the neuronal network activities.
Collapse
Affiliation(s)
- Min Fu
- Medical College of Tsinghua University, Beijing 100084, China
| | | | | | | | | | | |
Collapse
|