1
|
Rashid F, Khan KM, Saiprakash S, Ahmed G, Sultana R, Parvez F, Islam Z, Rahaman MS. Epidemiological Evidence on the Associations of Metal Exposure with Alzheimer's Disease and Related Dementias Among Elderly Women. J Clin Med 2025; 14:3776. [PMID: 40507537 PMCID: PMC12156812 DOI: 10.3390/jcm14113776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 05/14/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Emerging evidence suggests a potential link between heavy metals and Alzheimer's disease and related dementias (AD/ADRD). This study compiled epidemiological evidence from research published over the past 11 years on the impact of metals on AD/ADRD in women. Women have unique risk factors for late onset of AD/ADRD, in addition to genetic factors, apolipoprotein E allele (APOE4), and longer life expectancy. Furthermore, women are twice likely as men to experience depression, which increases their risk of developing AD/ADRD. Our narrative review underscored the necessity of a sex-specific approach to address women's vulnerability to AD/ADRD. Methods: Electronic databases, including PubMed, Google Scholar, NIOSH Toxline, and Scopus, were thoroughly searched to identify primary epidemiological studies on older women exposed to metals and published between 2014 to 2024. Results: We identified 34 epidemiological studies that met the inclusion criteria. The findings revealed a complex interplay between environmental metals such as lead (Pb), cadmium (Cd), arsenic (As), manganese (Mn), selenium (Se), iron (Fe), zinc (Zn), copper (Cu), magnesium (Mg), and calcium (Ca) and the risk of AD/ADRD in women. Significant adverse effects were reported for Cu, Cd, As, Pb, and Mn while significant protective effects were found between Se, Fe, and Zn in blood and AD/ADRD among older women. However, some studies also reported no correlations. Conclusions: Overall, our review identified contrasting results regarding the effects of metals on AD/ADRD in women. Future studies should collect additional evidence to understanding the effects of heavy metals on AD/ADRD in women for developing preventive measures.
Collapse
Affiliation(s)
- Fahmida Rashid
- Department of Public Health, College of Health Sciences, Sam Houston State University, Texas, TX 77341, USA; (F.R.); (R.S.)
| | - Khalid M. Khan
- Department of Public Health, College of Health Sciences, Sam Houston State University, Texas, TX 77341, USA; (F.R.); (R.S.)
| | - Samyukthaa Saiprakash
- College of Osteopathic Medicine, Sam Houston State University, Texas, TX 77304, USA;
| | - Giasuddin Ahmed
- Department of Biology and Chemistry, Texas A & M International University, Laredo, TX 78041, USA;
| | - Rasheda Sultana
- Department of Public Health, College of Health Sciences, Sam Houston State University, Texas, TX 77341, USA; (F.R.); (R.S.)
| | - Faruque Parvez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA;
| | - Zhahirul Islam
- Gut-Brain Axis Laboratory, Infectious Diseases Division (IDD), Bangladesh (icddr,b), Dhaka 1212, Bangladesh;
| | - Md. Shiblur Rahaman
- Department of Public Health, College of Health Sciences, Sam Houston State University, Texas, TX 77341, USA; (F.R.); (R.S.)
- Department of Environmental Science and Disaster Management, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| |
Collapse
|
2
|
Digman A, Pajarillo E, Kim S, Ajayi I, Son DS, Aschner M, Lee E. Tamoxifen induces protection against manganese toxicity by REST upregulation via the ER-α/Wnt/β-catenin pathway in neuronal cells. J Biol Chem 2025; 301:108529. [PMID: 40280417 DOI: 10.1016/j.jbc.2025.108529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 03/17/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic exposure to elevated levels of manganese (Mn) causes a neurological disorder referred to as manganism, with symptoms resembling Parkinson's disease (PD). The repressor element-1 silencing transcription factor (REST) has been shown to be neuroprotective in several neurological disorders, including PD, suggesting that identifying REST upregulation mechanisms is an important avenue for the development of novel therapeutics. 17β-estradiol (E2) activates the Wnt/β-catenin signaling, which is known to increase REST transcription. E2 and tamoxifen (TX), a selective estrogen receptor modulator, exerted protection against Mn toxicity. In this study, we tested if TX upregulates REST potentially via Wnt/β-catenin signaling in Cath.a-differentiated (CAD) neuronal cells using luciferase assay, qPCR, Western blot analysis, immunocytochemistry, mutagenesis, chromatin immunoprecipitation, and electrophoretic mobility shift assay. TX (1 μM) increased REST promoter activities and mRNA/protein levels and attenuated Mn (250 μM)-decreased REST transcription in parallel with TX's protective effects against Mn-induced toxicity, potentially via Wnt. TX activated Wnt/β-catenin signaling by preventing β-catenin degradation via inactivation of glycogen synthase kinase-3 beta, leading to increased β-catenin levels and its nuclear translocation and binding to T-cell factor/lymphoid enhancer binding factor sites on Wnt-responsive elements (WRE) of the REST promoter. Mutation of WRE abolished TX-induced REST promoter activity. TX-induced Wnt signaling activation was primarily via the estrogen receptor (ER)-α, although ER-β and G protein-coupled estrogen receptor 1 also mediated TX's action on REST transcription. These findings underscore the critical role of Wnt/β-catenin signaling in TX-induced REST transcription, affording protection mechanisms against Mn toxicity and neurological disorders associated with REST dysfunction.
Collapse
Affiliation(s)
- Alexis Digman
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Itunu Ajayi
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA.
| |
Collapse
|
3
|
Nyarko-Danquah I, Pajarillo E, Kim S, Digman A, Multani HK, Ajayi I, Son DS, Aschner M, Lee E. Microglial Sp1 induced LRRK2 upregulation in response to manganese exposure, and 17β-estradiol afforded protection against this manganese toxicity. Neurotoxicology 2024; 103:105-114. [PMID: 38857675 PMCID: PMC11972059 DOI: 10.1016/j.neuro.2024.05.007] [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: 02/12/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024]
Abstract
Chronic exposure to elevated levels of manganese (Mn) causes a neurological disorder referred to as manganism, presenting symptoms similar to those of Parkinson's disease (PD), yet the mechanisms by which Mn induces its neurotoxicity are not completely understood. 17β-estradiol (E2) affords neuroprotection against Mn toxicity in various neural cell types including microglia. Our previous studies have shown that leucine-rich repeat kinase 2 (LRRK2) mediates Mn-induced inflammatory toxicity in microglia. The LRRK2 promoter sequences contain three putative binding sites of the transcription factor (TF), specificity protein 1 (Sp1), which increases LRRK2 promoter activity. In the present study, we tested if the Sp1-LRRK2 pathway plays a role in both Mn toxicity and the protection afforded by E2 against Mn toxicity in BV2 microglial cells. The results showed that Mn induced cytotoxicity, oxidative stress, and tumor necrosis factor-α production, which were attenuated by an LRRK2 inhibitor, GSK2578215A. The overexpression of Sp1 increased LRRK2 promoter activity, mRNA and protein levels, while inhibition of Sp1 with its pharmacological inhibitor, mithramycin A, attenuated the Mn-induced increases in LRRK2 expression. Furthermore, E2 attenuated the Mn-induced Sp1 expression by decreasing the expression of Sp1 via the promotion of the ubiquitin-dependent degradation pathway, which was accompanied by increased protein levels of RING finger protein 4, the E3-ligase of Sp1, Sp1 ubiquitination, and SUMOylation. Taken together, our novel findings suggest that Sp1 serves as a critical TF in Mn-induced LRRK2 expression as well as in the protection afforded by E2 against Mn toxicity through reduction of LRRK2 expression in microglia.
Collapse
Affiliation(s)
- Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Harpreet Kaur Multani
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Itunu Ajayi
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA.
| |
Collapse
|
4
|
Yang J, Wang Y, Xia Y, Ren Y, Wang Z, Meng X, Li S, Liu X, Shao J. PFOS Elicits Cytotoxicity in Neuron Through Astrocyte-Derived CaMKII-DLG1 Signaling In Vitro Rat Hippocampal Model. Neurochem Res 2024; 49:1226-1238. [PMID: 38393622 DOI: 10.1007/s11064-024-04109-9] [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: 08/15/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024]
Abstract
Both epidemiological investigation and animal experiments demonstrated that pre-/postnatal exposure to perfluorooctane sulfonic acid (PFOS) could induce neurodevelopmental disorders. Previous studies showed that astrocyte was involved in PFOS-induced neurotoxicity, while little information is available. In the present study, the role of astrocyte-derived calmodulin-dependent protein kinase II (CaMKII)-phosphorylated discs large homolog 1 (DLG1) signaling in PFOS eliciting cytotoxicity in neuron was explored with primary cultured hippocampal astrocyte and neuron. The application of PFOS showed a decreased cell viability, synapse length and glutamate transporter 1 (GLT-1) expression, but an increased CaMKII, DLG1 and cyclic AMP response element binding protein (CREB) expression in primary cultured astrocyte. With 2-(2-hydroxyethylamino)-6-aminohexylcarbamic acid tert-butyl ester-9-isopropylpurine (CK59), the CaMKII inhibitor, the disturbed cell viability and molecules induced by PFOS could be alleviated (CREB expression was excluded) in astrocytes. The cytotoxic effect of neuron exposed to astrocyte conditional medium collected from PFOS (PFOS-ACM) pretreated with CK59 was also decreased. These results indicated that PFOS mediated GLT-1 expression through astrocyte-derived CaMKII-DLG signaling, which might be associated with injuries on neurons. The present study gave an insight in further exploration of mechanism in PFOS-induced neurotoxicity.
Collapse
Affiliation(s)
- Jiawei Yang
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Ying Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yuyan Xia
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Yajie Ren
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Zhi Wang
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Xin Meng
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Shuangyue Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Xiaohui Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China.
| | - Jing Shao
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
5
|
Pajarillo E, Nyarko-Danquah I, Digman A, Multani HK, Kim S, Gaspard P, Aschner M, Lee E. Mechanisms of manganese-induced neurotoxicity and the pursuit of neurotherapeutic strategies. Front Pharmacol 2022; 13:1011947. [PMID: 36605395 PMCID: PMC9808094 DOI: 10.3389/fphar.2022.1011947] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/01/2022] [Indexed: 01/07/2023] Open
Abstract
Chronic exposure to elevated levels of manganese via occupational or environmental settings causes a neurological disorder known as manganism, resembling the symptoms of Parkinson's disease, such as motor deficits and cognitive impairment. Numerous studies have been conducted to characterize manganese's neurotoxicity mechanisms in search of effective therapeutics, including natural and synthetic compounds to treat manganese toxicity. Several potential molecular targets of manganese toxicity at the epigenetic and transcriptional levels have been identified recently, which may contribute to develop more precise and effective gene therapies. This review updates findings on manganese-induced neurotoxicity mechanisms on intracellular insults such as oxidative stress, inflammation, excitotoxicity, and mitophagy, as well as transcriptional dysregulations involving Yin Yang 1, RE1-silencing transcription factor, transcription factor EB, and nuclear factor erythroid 2-related factor 2 that could be targets of manganese neurotoxicity therapies. This review also features intracellular proteins such as PTEN-inducible kinase 1, parkin, sirtuins, leucine-rich repeat kinase 2, and α-synuclein, which are associated with manganese-induced dysregulation of autophagy/mitophagy. In addition, newer therapeutic approaches to treat manganese's neurotoxicity including natural and synthetic compounds modulating excitotoxicity, autophagy, and mitophagy, were reviewed. Taken together, in-depth mechanistic knowledge accompanied by advances in gene and drug delivery strategies will make significant progress in the development of reliable therapeutic interventions against manganese-induced neurotoxicity.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Harpreet Kaur Multani
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL, United States
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Patric Gaspard
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| |
Collapse
|
6
|
Adamson A, Buck SA, Freyberg Z, De Miranda BR. Sex Differences in Dopaminergic Vulnerability to Environmental Toxicants - Implications for Parkinson's Disease. Curr Environ Health Rep 2022; 9:563-573. [PMID: 36201109 PMCID: PMC10201647 DOI: 10.1007/s40572-022-00380-6] [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] [Accepted: 08/24/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Sex dimorphism in Parkinson's disease (PD) is an ostensible feature of the neurological disorder, particularly as men are 1.5-2 times more likely to develop PD than women. Clinical features of the disease, such as presentation at onset, most prevalent symptoms, and response to treatment, are also affected by sex. Despite these well-known sex differences in PD risk and phenotype, the mechanisms that impart sex dimorphisms in PD remain poorly understood. RECENT FINDINGS As PD incidence is influenced by environmental factors, an intriguing pattern has recently emerged in research studies suggesting a male-specific vulnerability to dopaminergic neurodegeneration caused by neurotoxicant exposure, with relative protection in females. These new experimental data have uncovered potential mechanisms that provide clues to the source of sex differences in dopaminergic neurodegeneration and other PD pathology such as alpha-synuclein toxicity. In this review, we discuss the emerging evidence of increased male sensitivity to neurodegeneration from environmental exposures. We examine mechanisms underlying dopaminergic neurodegeneration and PD-related pathologies with evidence supporting the roles of estrogen, SRY expression, the vesicular glutamate transporter VGLUT2, and the microbiome as prospective catalysts for male vulnerability. We also highlight the importance of including sex as a biological variable, particularly when evaluating dopaminergic neurotoxicity in the context of PD.
Collapse
Affiliation(s)
- Ashley Adamson
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA
| | - Silas A Buck
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA.
| |
Collapse
|
7
|
Soto-Verdugo J, Siva-Parra J, Hernández-Kelly LC, Ortega A. Acute Manganese Exposure Modifies the Translation Machinery via PI3K/Akt Signaling in Glial Cells. ASN Neuro 2022; 14:17590914221131452. [PMID: 36203371 PMCID: PMC9551334 DOI: 10.1177/17590914221131452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SUMMARY STATEMENT We demonstrate herein that short-term exposure of radial glia cells to Manganese, a neurotoxic metal, induces an effect on protein synthesis, altering the protein repertoire of these cells.
Collapse
Affiliation(s)
| | | | | | - Arturo Ortega
- Arturo Ortega, Departamento de Toxicología,
Centro de Investigación y de Estudios Avanzados del Instituto Politécnico
Nacional, México City, México, 07360.
| |
Collapse
|
8
|
Ijomone OM, Iroegbu JD, Morcillo P, Ayodele AJ, Ijomone OK, Bornhorst J, Schwerdtle T, Aschner M. Sex-dependent metal accumulation and immunoexpression of Hsp70 and Nrf2 in rats' brain following manganese exposure. ENVIRONMENTAL TOXICOLOGY 2022; 37:2167-2177. [PMID: 35596948 PMCID: PMC9357062 DOI: 10.1002/tox.23583] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/21/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
Manganese (Mn), although important for multiple cellular processes, has posed environmental health concerns due to its neurotoxic effects. In recent years, there have been extensive studies on the mechanism of Mn-induced neuropathology, as well as the sex-dependent vulnerability to its neurotoxic effects. Nonetheless, cellular mechanisms influenced by sex differences in susceptibility to Mn have yet to be adequately characterized. Since oxidative stress is a key mechanism of Mn neurotoxicity, here, we have probed Hsp70 and Nrf2 proteins to investigate the sex-dependent changes following exposure to Mn. Male and female rats were administered intraperitoneal injections of MnCl2 (10 mg/kg and 25 mg/kg) 48 hourly for a total of eight injections (15 days). We evaluated changes in body weight, as well as Mn accumulation, Nrf2 and Hsp70 expression across four brain regions; striatum, cortex, hippocampus and cerebellum in both sexes. Our results showed sex-specific changes in body-weight, specifically in males but not in females. Additionally, we noted sex-dependent accumulation of Mn in the brain, as well as in expression levels of Nrf2 and Hsp70 proteins. These findings revealed sex-dependent susceptibility to Mn-induced neurotoxicity corresponding to differential Mn accumulation, and expression of Hsp70 and Nrf2 across several brain regions.
Collapse
Affiliation(s)
- Omamuyovwi M. Ijomone
- Departments of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- The Neuro- Lab, Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Joy D. Iroegbu
- The Neuro- Lab, Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Patricia Morcillo
- Departments of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Akinyemi J. Ayodele
- Departments of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Olayemi K. Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Nigeria
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
- TraceAge – DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Tanja Schwerdtle
- TraceAge – DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Michael Aschner
- Departments of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
9
|
Rimmele TS, Li S, Andersen JV, Westi EW, Rotenberg A, Wang J, Aldana BI, Selkoe DJ, Aoki CJ, Dulla CG, Rosenberg PA. Neuronal Loss of the Glutamate Transporter GLT-1 Promotes Excitotoxic Injury in the Hippocampus. Front Cell Neurosci 2022; 15:788262. [PMID: 35035352 PMCID: PMC8752461 DOI: 10.3389/fncel.2021.788262] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
GLT-1, the major glutamate transporter in the mammalian central nervous system, is expressed in presynaptic terminals that use glutamate as a neurotransmitter, in addition to astrocytes. It is widely assumed that glutamate homeostasis is regulated primarily by glutamate transporters expressed in astrocytes, leaving the function of GLT-1 in neurons relatively unexplored. We generated conditional GLT-1 knockout (KO) mouse lines to understand the cell-specific functions of GLT-1. We found that stimulus-evoked field extracellular postsynaptic potentials (fEPSPs) recorded in the CA1 region of the hippocampus were normal in the astrocytic GLT-1 KO but were reduced and often absent in the neuronal GLT-1 KO at 40 weeks. The failure of fEPSP generation in the neuronal GLT-1 KO was also observed in slices from 20 weeks old mice but not consistently from 10 weeks old mice. Using an extracellular FRET-based glutamate sensor, we found no difference in stimulus-evoked glutamate accumulation in the neuronal GLT-1 KO, suggesting a postsynaptic cause of the transmission failure. We hypothesized that excitotoxicity underlies the failure of functional recovery of slices from the neuronal GLT-1 KO. Consistent with this hypothesis, the non-competitive NMDA receptor antagonist MK801, when present in the ACSF during the recovery period following cutting of slices, promoted full restoration of fEPSP generation. The inclusion of an enzymatic glutamate scavenging system in the ACSF conferred partial protection. Excitotoxicity might be due to excess release or accumulation of excitatory amino acids, or to metabolic perturbation resulting in increased vulnerability to NMDA receptor activation. Previous studies have demonstrated a defect in the utilization of glutamate by synaptic mitochondria and aspartate production in the synGLT-1 KO in vivo, and we found evidence for similar metabolic perturbations in the slice preparation. In addition, mitochondrial cristae density was higher in synaptic mitochondria in the CA1 region in 20–25 weeks old synGLT-1 KO mice in the CA1 region, suggesting compensation for loss of axon terminal GLT-1 by increased mitochondrial efficiency. These data suggest that GLT-1 expressed in presynaptic terminals serves an important role in the regulation of vulnerability to excitotoxicity, and this regulation may be related to the metabolic role of GLT-1 expressed in glutamatergic axon terminals.
Collapse
Affiliation(s)
- Theresa S Rimmele
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jens Velde Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Rotenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| | - Jianlin Wang
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Blanca Irene Aldana
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Chiye J Aoki
- Center for Neural Science, New York University, NY, United States.,Neuroscience Institute NYU Langone Medical Center, NY, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Paul Allen Rosenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Critical Involvement of Glial Cells in Manganese Neurotoxicity. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1596185. [PMID: 34660781 PMCID: PMC8514895 DOI: 10.1155/2021/1596185] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
Over the years, most of the research concerning manganese exposure was restricted to the toxicity of neuronal cells. Manganese is an essential trace element that in high doses exerts neurotoxic effects. However, in the last two decades, efforts have shifted toward a more comprehensive approach that takes into account the involvement of glial cells in the development of neurotoxicity as a brain insult. Glial cells provide structural, trophic, and metabolic support to neurons. Nevertheless, these cells play an active role in adult neurogenesis, regulation of synaptogenesis, and synaptic plasticity. Disturbances in glial cell function can lead to neurological disorders, including neurodegenerative diseases. This review highlights the pivotal role that glial cells have in manganese-induced neurotoxicity as well as the most sounding mechanisms involved in the development of this phenomenon.
Collapse
|
11
|
Effect of 17β-Estradiol, Progesterone, and Tamoxifen on Neurons Infected with Toxoplasma gondii In Vitro. Microorganisms 2021; 9:microorganisms9102174. [PMID: 34683495 PMCID: PMC8541540 DOI: 10.3390/microorganisms9102174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 11/16/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is the causal agent of toxoplasmosis, which produces damage in the central nervous system (CNS). Toxoplasma-CNS interaction is critical for the development of disease symptoms. T. gondii can form cysts in the CNS; however, neurons are more resistant to this infection than astrocytes. The probable mechanism for neuron resistance is a permanent state of neurons in the interface, avoiding the replication of intracellular parasites. Steroids regulate the formation of Toxoplasma cysts in mice brains. 17β-estradiol and progesterone also participate in the control of Toxoplasma infection in glial cells in vitro. The aim of this study was to evaluate the effects of 17β-estradiol, progesterone, and their specific agonists-antagonists on Toxoplasma infection in neurons in vitro. Neurons cultured were pretreated for 48 h with 17β-estradiol or progesterone at 10, 20, 40, 80, or 160 nM/mL or tamoxifen 1 μM/mL plus 17β-estradiol at 10, 20, 40, 80, and 160 nM/mL. In other conditions, the neurons were pretreated during 48 h with 4,4',4″-(4-propyl-[1H] pyrozole-1,3,5-triyl) trisphenol or 23-bis(4-hydroxyphenyl) propionitrile at 1 nM/mL, and mifepristone 1 µM/mL plus progesterone at 10, 20, 40, 80, and 160 nM/mL. Neurons were infected with 5000 tachyzoites of the T. gondii strain RH. The effect of 17β estradiol, progesterone, their agonists, or antagonists on Toxoplasma infection in neurons was evaluated at 24 and 48 h by immunocytochemistry. T. gondii replication was measured with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction assay. 17β-Estradiol alone or plus tamoxifen reduced infected neurons (50%) compared to the control at 48 h. Progesterone plus estradiol decreased the number of intracellular parasites at 48 h of treatment compared to the control (p < 0.001). 4,4',4″-(4-propyl-[1H] pyrozole-1,3,5-triyl) trisphenol and 23-bis(4-hydroxyphenyl) propionitrile reduced infected neurons at 48 h of treatment significantly compared to the control (p < 0.05 and p < 0.001, respectively). The Toxoplasma infection process was decreased by the effect of 17β-estradiol alone or combined with tamoxifen or progesterone in neurons in vitro. These results suggest the essential participation of progesterone and estradiol and their classical receptors in the regulation of T. gondii neuron infection.
Collapse
|
12
|
Avram CM, Brumbach BH, Hiller AL. A Report of Tamoxifen and Parkinson's Disease in a US Population and a Review of the Literature. Mov Disord 2021; 36:1238-1242. [PMID: 33449420 DOI: 10.1002/mds.28471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/08/2020] [Accepted: 12/09/2020] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Tamoxifen, a selective estrogen receptor modulator, has been shown to variably affect Parkinson's disease (PD) risk. OBJECTIVE The aim of this study was to review epidemiological literature and evaluate the rate of PD in women with breast cancer with tamoxifen exposure in a US population. METHODS A literature search was conducted to identify relevant studies. We performed a retrospective cohort analysis using the Nurses' Health Study Version One to report descriptive statistics. RESULTS Most studies suggest there may be a time-dependent effect of tamoxifen on PD risk, with the risk increasing with time from exposure. However, rates of PD in persons exposed to tamoxifen overall appear to be low. In our cohort, PD was evident in 6.2 per 1,000 of those with tamoxifen use and 3.6 per 1,000 of those without tamoxifen use. Time from breast cancer to PD diagnosis was 9.7 years among women with tamoxifen exposure and 11.7 among women without. CONCLUSIONS Tamoxifen may be associated with an increased risk for PD. Further research is needed to elucidate the role of estrogen and selective estrogen antagonism in PD. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Carmen M Avram
- Duke University Medical Center, Durham, North Carolina, USA
| | | | - Amie L Hiller
- Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
13
|
Bordone MC, Barbosa-Morais NL. Unraveling Targetable Systemic and Cell-Type-Specific Molecular Phenotypes of Alzheimer's and Parkinson's Brains With Digital Cytometry. Front Neurosci 2020; 14:607215. [PMID: 33362460 PMCID: PMC7756021 DOI: 10.3389/fnins.2020.607215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the two most common neurodegenerative disorders worldwide, with age being their major risk factor. The increasing worldwide life expectancy, together with the scarcity of available treatment choices, makes it thus pressing to find the molecular basis of AD and PD so that the causing mechanisms can be targeted. To study these mechanisms, gene expression profiles have been compared between diseased and control brain tissues. However, this approach is limited by mRNA expression profiles derived for brain tissues highly reflecting their degeneration in cellular composition but not necessarily disease-related molecular states. We therefore propose to account for cell type composition when comparing transcriptomes of healthy and diseased brain samples, so that the loss of neurons can be decoupled from pathology-associated molecular effects. This approach allowed us to identify genes and pathways putatively altered systemically and in a cell-type-dependent manner in AD and PD brains. Moreover, using chemical perturbagen data, we computationally identified candidate small molecules for specifically targeting the profiled AD/PD-associated molecular alterations. Our approach therefore not only brings new insights into the disease-specific and common molecular etiologies of AD and PD but also, in these realms, foster the discovery of more specific targets for functional and therapeutic exploration.
Collapse
Affiliation(s)
- Marie C Bordone
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno L Barbosa-Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
14
|
Zhou L, Wong KY, Cao S, Poon CCW, Yu W, Dong X, Tsim KWK, Wong MS. A standardized extract of Danggui Buxue Tang decoction selectively exerts estrogenic activities distinctly from tamoxifen. Phytother Res 2020; 35:1456-1467. [PMID: 33063371 DOI: 10.1002/ptr.6909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 11/06/2022]
Abstract
More and more menopausal women use Danggui Buxue Tang (DBT) for relieving their symptoms. Concerns for its safety have been raised as it contains phytoestrogen and acts via estrogen receptors (ERs). Our study aimed to determine whether DBT could selectively exert estrogenic activities and interact with tamoxifen in bone, brain, uterus, and breast by using ovariectomized (OVX) rats and ER-positive cells. In OVX rats, DBT induced a 31.4% increase in bone mineral density and restored the mRNA expression of dopamine biomarker in striatum, 3.32-fold for tyrosine hydrolase (p < .001) and 0.21-fold for dopamine transporter (p < .001), which was similar to tamoxifen; tamoxifen, but not DBT, increased uterus weight and Complement component 3 expression by more than twofold (p < .001); unlike tamoxifen, DBT induced mild proliferation in mammary gland. Two-way ANOVA indicated the interactions between them in OVX rats (p < .05) but DBT did not alter the responses to tamoxifen. DBT stimulated proliferation or differentiation and estrogen response element in MCF-7, MG-63, Ishikawa, and SHSY5Y cells and altered the effects of tamoxifen. In summary, DBT exerted estrogenic effects in tissue-selective manner, which was different from tamoxifen. DBT interacted with tamoxifen but did not significantly alter its effects in OVX rats.
Collapse
Affiliation(s)
- Liping Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Ka-Ying Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Sisi Cao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Christina Chui-Wa Poon
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Wenxuan Yu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Xiaoli Dong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Karl Wah-Keung Tsim
- Division of Life Science and Center for Chinese Medicine R&D, Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Man-Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China.,State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
15
|
Vahidinia Z, Karimian M, Joghataei MT. Neurosteroids and their receptors in ischemic stroke: From molecular mechanisms to therapeutic opportunities. Pharmacol Res 2020; 160:105163. [DOI: 10.1016/j.phrs.2020.105163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023]
|
16
|
Pajarillo E, Rizor A, Son DS, Aschner M, Lee E. The transcription factor REST up-regulates tyrosine hydroxylase and antiapoptotic genes and protects dopaminergic neurons against manganese toxicity. J Biol Chem 2020; 295:3040-3054. [PMID: 32001620 PMCID: PMC7062174 DOI: 10.1074/jbc.ra119.011446] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Dopaminergic functions are important for various biological activities, and their impairment leads to neurodegeneration, a hallmark of Parkinson's disease (PD). Chronic manganese (Mn) exposure causes the neurological disorder manganism, presenting symptoms similar to those of PD. Emerging evidence has linked the transcription factor RE1-silencing transcription factor (REST) to PD and also Alzheimer's disease. But REST's role in dopaminergic neurons is unclear. Here, we investigated whether REST protects dopaminergic neurons against Mn-induced toxicity and enhances expression of the dopamine-synthesizing enzyme tyrosine hydroxylase (TH). We report that REST binds to RE1 consensus sites in the TH gene promoter, stimulates TH transcription, and increases TH mRNA and protein levels in dopaminergic cells. REST binding to the TH promoter recruited the epigenetic modifier cAMP-response element-binding protein-binding protein/p300 and thereby up-regulated TH expression. REST relieved Mn-induced repression of TH promoter activity, mRNA, and protein levels and also reduced Mn-induced oxidative stress, inflammation, and apoptosis in dopaminergic neurons. REST reduced Mn-induced proinflammatory cytokines, including tumor necrosis factor α, interleukin 1β (IL-1β), IL-6, and interferon γ. Moreover, REST inhibited the Mn-induced proapoptotic proteins Bcl-2-associated X protein (Bax) and death-associated protein 6 (Daxx) and attenuated an Mn-induced decrease in the antiapoptotic proteins Bcl-2 and Bcl-xL. REST also enhanced the expression of antioxidant proteins, including catalase, NF-E2-related factor 2 (Nrf2), and heme oxygenase 1 (HO-1). Our findings indicate that REST activates TH expression and thereby protects neurons against Mn-induced toxicity and neurological disorders associated with dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32301
| | - Asha Rizor
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32301
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, Tennessee 37208
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, New York 10461
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32301.
| |
Collapse
|
17
|
Xiao Z, Tang Z, Wu L, Feng X, Sun X, Tang W, Wang J, Jin L, Wang R. Manganese-enhanced magnetic resonance imaging in the whole visual pathway: chemical identification and neurotoxic changes. Acta Radiol 2019; 60:1653-1662. [PMID: 30922072 DOI: 10.1177/0284185119840227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Zebin Xiao
- Department of Radiology, Eye & ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, PR China
| | - Zuohua Tang
- Department of Radiology, Eye & ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, PR China
| | - Lingjie Wu
- Department of Otolaryngology, Eye & ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, PR China
| | - Xiaoyuan Feng
- Department of Radiology, Huashan Hospital of Shanghai Medical School, Fudan University, Shanghai, PR China
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital of Shanghai Medical School, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Key Laboratory of Myopia, NHFPC (Fudan University), and Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, PR China
| | - Weijun Tang
- Department of Radiology, Huashan Hospital of Shanghai Medical School, Fudan University, Shanghai, PR China
| | - Jie Wang
- Department of Radiotherapy, Eye & ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, PR China
| | - Lixin Jin
- Siemens Ltd., Healthcare Sector, Shanghai, PR China
| | - Rong Wang
- Department of Radiology, Eye & ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, PR China
| |
Collapse
|
18
|
Zhao CC, Jiang MY, Zhang LY, Hu YY, Hu ZJ, Zhang MY, Qi J, Su AC, Lou N, Xian XH, Zhang JG, Li WB, Zhang M. Peroxisome proliferator-activated receptor gamma participates in the acquisition of brain ischemic tolerance induced by ischemic preconditioning via glial glutamate transporter 1 in vivo and in vitro. J Neurochem 2019; 151:608-625. [PMID: 31314916 DOI: 10.1111/jnc.14824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/08/2019] [Accepted: 07/11/2019] [Indexed: 01/29/2023]
Abstract
Glial glutamate transporter 1 (GLT-1) plays a vital role in the induction of brain ischemic tolerance (BIT) by ischemic preconditioning (IPC). However, the mechanism still needs to be further explained. The aim of this study was to investigate whether peroxisome proliferator-activated receptor gamma (PPARγ) participates in regulating GLT-1 during the acquisition of BIT induced by IPC. Initially, cerebral IPC induced BIT and enhanced PPARγ and GLT-1 expression in the CA1 hippocampus in rats. The ratio of nuclear/cytoplasmic PPARγ was also increased. At the same time, the up-regulation of PPARγ expression in astrocytes in the CA1 hippocampus was revealed by double immunofluorescence for PPARγ and glial fibrillary acidic protein. Then, the mechanism by which PPARγ regulates GLT-1 was studied in rat cortical astrocyte-neuron cocultures. We found that IPC [45 min of oxygen glucose deprivation (OGD)] protected neuronal survival after lethal OGD (4 h of OGD), which usually leads to neuronal death. The activation of PPARγ occurred earlier than the up-regulation of GLT-1 in astrocytes after IPC, as determined by western blot and immunofluorescence. Moreover, the preadministration of the PPARγ antagonist T0070907 or PPARγ siRNA significantly attenuated GLT-1 up-regulation and the neuroprotective effects induced by IPC in vitro. Finally, the effect of the PPARγ antagonist on GLT-1 expression and BIT was verified in vivo. We observed that the preadministration of T0070907 by intracerebroventricular injection dose-dependently attenuated the up-regulation of GLT-1 and BIT induced by cerebral IPC in rats. In conclusion, PPARγ participates in regulating GLT-1 during the acquisition of BIT induced by IPC. Cover Image for this issue: doi: 10.1111/jnc.14532. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Cong-Cong Zhao
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Meng-Yang Jiang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Zhen-Jie Hu
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Meng-Yue Zhang
- Clinical Medicine, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jie Qi
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - A-Chou Su
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Nan Lou
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, People's Republic of China
| |
Collapse
|
19
|
Escalante M, Soto-Verdugo J, Hernández-Kelly LC, Hernández-Melchor D, López-Bayghen E, Olivares-Bañuelos TN, Ortega A. GLAST Activity is Modified by Acute Manganese Exposure in Bergmann Glial Cells. Neurochem Res 2019; 45:1365-1374. [PMID: 31363896 DOI: 10.1007/s11064-019-02848-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022]
Abstract
Glutamate is the major excitatory amino acid neurotransmitter in the vertebrate brain. It exerts its actions through the activation of specific plasma membrane receptors expressed in neurons and glial cells. Overactivation of glutamate receptors results in neuronal death, known as excitotoxicity. A family of sodium-dependent glutamate transporters enriched in glial cells are responsible of the vast majority of the removal of this amino acid form the synaptic cleft. Therefore, a precise and exquisite regulation of these proteins is required not only for a proper glutamatergic transmission but also for the prevention of an excitotoxic insult. Manganese is a trace element essential as a cofactor for several enzymatic systems, although in high concentrations is involved in the disruption of brain glutamate homeostasis. The molecular mechanisms associated to manganese neurotoxicity have been focused on mitochondrial function, although energy depletion severely compromises the glutamate uptake process. In this context, in this contribution we analyze the effect of manganese exposure in glial glutamate transporters function. To this end, we used the well-established model of chick cerebellar Bergmann glia cultures. A time and dose dependent modulation of [3H]-D-aspartate uptake was found. An increase in the transporter catalytic efficiency, most probably linked to a discrete increase in the affinity of the transporter was detected upon manganese exposure. Interestingly, glucose uptake was reduced by this metal. These results favor the notion of a direct effect of manganese on glial cells, this in turn alters their coupling with neurons and might lead to changes in glutamatergic transmission.
Collapse
Affiliation(s)
- Miguel Escalante
- Departamento de Toxicología, Centro de Investigación Y de Estudios Avanzados del IPN, Apartado Postal 14-740, 07360, Ciudad de Mexico, Mexico
| | - Jazmín Soto-Verdugo
- Departamento de Toxicología, Centro de Investigación Y de Estudios Avanzados del IPN, Apartado Postal 14-740, 07360, Ciudad de Mexico, Mexico
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación Y de Estudios Avanzados del IPN, Apartado Postal 14-740, 07360, Ciudad de Mexico, Mexico
| | - Dinorah Hernández-Melchor
- Departamento de Toxicología, Centro de Investigación Y de Estudios Avanzados del IPN, Apartado Postal 14-740, 07360, Ciudad de Mexico, Mexico
| | - Esther López-Bayghen
- Departamento de Toxicología, Centro de Investigación Y de Estudios Avanzados del IPN, Apartado Postal 14-740, 07360, Ciudad de Mexico, Mexico
| | - Tatiana N Olivares-Bañuelos
- Instituto de Investigaciones Oceanológicas, Universidad Autónoma de Baja California, 22860, Ensenada, Baja California, Mexico
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación Y de Estudios Avanzados del IPN, Apartado Postal 14-740, 07360, Ciudad de Mexico, Mexico.
| |
Collapse
|
20
|
Kim J, Pajarillo E, Rizor A, Son DS, Lee J, Aschner M, Lee E. LRRK2 kinase plays a critical role in manganese-induced inflammation and apoptosis in microglia. PLoS One 2019; 14:e0210248. [PMID: 30645642 PMCID: PMC6333340 DOI: 10.1371/journal.pone.0210248] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/19/2018] [Indexed: 12/11/2022] Open
Abstract
Long-term exposure to elevated levels of manganese (Mn) causes manganism, a neurodegenerative disorder with Parkinson's disease (PD)-like symptoms. Increasing evidence suggests that leucine-rich repeat kinase 2 (LRRK2), which is highly expressed in microglia and macrophages, contributes to the inflammation and neurotoxicity seen in autosomal dominant and sporadic PD. As gene-environment interactions have emerged as important modulators of PD-associated toxicity, LRRK2 may also mediate Mn-induced inflammation and pathogenesis. In this study, we investigated the role of LRRK2 in Mn-induced toxicity using human microglial cells (HMC3), LRRK2-wild-type (WT) and LRRK2-knockout (KO) RAW264.7 macrophage cells. Results showed that Mn activated LRRK2 kinase by phosphorylation of its serine residue at the 1292 position (S1292) as a marker of its kinase activity in macrophage and microglia, while inhibition with GSK2578215A (GSK) and MLi-2 abolished Mn-induced LRRK2 activation. LRRK2 deletion and its pharmacological inhibition attenuated Mn-induced apoptosis in macrophages and microglia, along with concomitant decreases in the pro-apoptotic Bcl-2-associated X (Bax) protein. LRRK2 deletion also attenuated Mn-induced production of reactive oxygen species (ROS) and the pro-inflammatory cytokine TNF-α. Mn-induced phosphorylation of mitogen-activated protein kinase (MAPK) p38 and ERK signaling proteins was significantly attenuated in LRRK2 KO cells and GSK-treated cells. Moreover, inhibition of MAPK p38 and ERK as well as LRRK2 attenuated Mn-induced oxidative stress and cytotoxicity. These findings suggest that LRRK2 kinase activity plays a critical role in Mn-induced toxicity via downstream activation of MAPK signaling in macrophage and microglia. Collectively, these results suggest that LRRK2 could be a potential molecular target for developing therapeutics to treat Mn-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Judong Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, Florida, United States of America
| | - Edward Pajarillo
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, Florida, United States of America
| | - Asha Rizor
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, Florida, United States of America
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Jayden Lee
- Department of Speech, Language & Hearing Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, Florida, United States of America
| |
Collapse
|
21
|
Shimamoto A, Rappeneau V, Munjal H, Farris T, Davis C, Wilson A, Edwards M, Moore C, Reynolds C, Meshul CK. Glutamate-Glutamine Transfer and Chronic Stress-Induced Sex Differences in Cocaine Responses. Neuroscience 2018; 391:104-119. [DOI: 10.1016/j.neuroscience.2018.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 01/16/2023]
|
22
|
McDonough PM, Prigozhina NL, Basa RCB, Price JH. Assay of Calcium Transients and Synapses in Rat Hippocampal Neurons by Kinetic Image Cytometry and High-Content Analysis: An In Vitro Model System for Postchemotherapy Cognitive Impairment. Assay Drug Dev Technol 2018; 15:220-236. [PMID: 28723268 DOI: 10.1089/adt.2017.797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Postchemotherapy cognitive impairment (PCCI) is commonly exhibited by cancer patients treated with a variety of chemotherapeutic agents, including the endocrine disruptor tamoxifen (TAM). The etiology of PCCI is poorly understood. Our goal was to develop high-throughput assay methods to test the effects of chemicals on neuronal function applicable to PCCI. Rat hippocampal neurons (RHNs) were plated in 96- or 384-well dishes and exposed to test compounds (forskolin [FSK], 17β-estradiol [ES]), TAM or fulvestrant [FUL], aka ICI 182,780) for 6-14 days. Kinetic Image Cytometry™ (KIC™) methods were developed to quantify spontaneously occurring intracellular calcium transients representing the activity of the neurons, and high-content analysis (HCA) methods were developed to quantify the expression, colocalization, and puncta formed by synaptic proteins (postsynaptic density protein-95 [PSD-95] and presynaptic protein Synapsin-1 [Syn-1]). As quantified by KIC, FSK increased the occurrence and synchronization of the calcium transients indicating stimulatory effects on RHN activity, whereas TAM had inhibitory effects. As quantified by HCA, FSK also increased PSD-95 puncta and PSD-95:Syn-1 colocalization, whereas ES increased the puncta of both PSD-95 and Syn-1 with little effect on colocalization. The estrogen receptor antagonist FUL also increased PSD-95 puncta. In contrast, TAM reduced Syn-1 and PSD-95:Syn-1 colocalization, consistent with its inhibitory effects on the calcium transients. Thus TAM reduced activity and synapse formation by the RHNs, which may relate to the ability of this agent to cause PCCI. The results illustrate that KIC and HCA can be used to quantify neurotoxic and neuroprotective effects of chemicals in RHNs to investigate mechanisms and potential therapeutics for PCCI.
Collapse
Affiliation(s)
| | | | | | - Jeffrey H Price
- 1 Vala Sciences Inc. , San Diego, California.,3 The Scintillon Institute , San Diego, California
| |
Collapse
|
23
|
Hong CT, Chan L, Hu CJ, Lin CM, Hsu CY, Lin MC. Tamoxifen and the Risk of Parkinson's Disease in Female Patients with Breast Cancer in Asian People: A Nationwide Population-Based Study. J Breast Cancer 2017; 20:356-360. [PMID: 29285040 PMCID: PMC5743995 DOI: 10.4048/jbc.2017.20.4.356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/07/2017] [Indexed: 12/25/2022] Open
Abstract
Purpose Whether tamoxifen affects the risk of neurodegenerative disease is controversial. This nationwide population-based study investigated the risk of Parkinson's disease (PD) associated with tamoxifen treatment in female patients with breast cancer using Taiwan's National Health Insurance Research Database. Methods A total of 5,185 and 5,592 female patients with breast cancer who did and did not, respectively, receive tamoxifen treatment between 2000 and 2009 were included in the study. Patients who subsequently developed PD were identified. A Cox proportional hazards model was used to compare the risk of PD between the aforementioned groups. Results Tamoxifen did not significantly increase the crude rate of developing PD in female patients with breast cancer (tamoxifen group, 16/5,169; non-tamoxifen group, 11/5,581; p=0.246). Tamoxifen did not significantly increase the adjusted hazard ratio (aHR) for subsequently developing PD (aHR, 1.310; 95% confidence interval [CI], 0.605-2.837; p=0.494). However, tamoxifen significantly increased the risk of PD among patients followed up for more than 6 years (aHR, 2.435; 95% CI, 1.008-5.882; p=0.048). Conclusion Tamoxifen treatment may increase the risk of PD in Taiwanese female patients with breast cancer more than 6 years after the initiation of treatment.
Collapse
Affiliation(s)
- Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Neurology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Neurology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Neurology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Min Lin
- Department of Neurology, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chien-Yeh Hsu
- Master Program in Global Health and Development, College of Public Health, Taipei Medical University, Taipei, Taiwan.,Department of Information Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Ming-Chin Lin
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
24
|
Bianchi A, Gobbo OL, Dufort S, Sancey L, Lux F, Tillement O, Coll JL, Crémillieux Y. Orotracheal manganese-enhanced MRI (MEMRI): An effective approach for lung tumor detection. NMR IN BIOMEDICINE 2017; 30:e3790. [PMID: 28857310 DOI: 10.1002/nbm.3790] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 07/15/2017] [Accepted: 07/19/2017] [Indexed: 06/07/2023]
Abstract
Lung cancer is a primary cause of cancer deaths worldwide. Timely detection of this pathology is necessary to delay or interrupt lung cancer progression, ultimately resulting in a possible better prognosis for the patient. In this context, magnetic resonance imaging (MRI) is especially promising. Ultra-short echo time (UTE) MRI sequences, in combination with gadolinium-based contrast agents, have indeed shown to be especially adapted to the detection of lung neoplastic lesions at submillimeter precision. Manganese-enhanced MRI (MEMRI) increasingly appears to be a possible effective alternative to gadolinium-enhanced MRI. In this work, we investigated whether low-dose MEMRI can effectively target non-small-cell lung cancer in rodents, whilst minimizing the potential toxic effect of manganese. Both systemic and orotracheal administration modalities allowed the identification of tumors of submillimeter size, as confirmed by bioluminescence imaging and histology. Equivalent tumor signal enhancements and contrast-to-noise ratios were observed with orotracheal administration using 20 times lower doses compared with the more conventional systemic route. This finding is of crucial importance as it supports the observation that higher performances of contrast agents can be obtained using an orotracheal administration route when targeting lung diseases. As a consequence, lower concentrations of contrast media can be employed, reducing the dose and potential safety issues. The non-detectable accumulation of ionic manganese in the brain and liver following orotracheal administration observed in vivo is extremely encouraging with regard to the safety of the orotracheal protocol with low-dose Mn2+ administration. To our knowledge, this is the first time that a study has clearly allowed the high-precision detection of lung tumor and its contours via the synergic employment of a strongly T1 -weighted MRI UTE sequence and ionic manganese, an inexpensive contrast agent. Overall, these results support the growing interest in drug and contrast agent delivery via the airways to target and diagnose several diseases of the lungs.
Collapse
Affiliation(s)
- Andrea Bianchi
- Centre de Résonance Magnétique des Systèmes Biologiques,CNRS UMR 5536, Université Bordeaux, Bordeaux, France
| | - Oliviero L Gobbo
- School of Pharmacy and Pharmaceutical Sciences and Institute of Neuroscience, Trinity College Dublin, Ireland
| | - Sandrine Dufort
- Nano-H S.A.S, Saint Quentin-Fallavier, France
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Lucie Sancey
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Institut Lumiére Matiére, Lyon, France
| | - François Lux
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Institut Lumiére Matiére, Lyon, France
| | - Olivier Tillement
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Institut Lumiére Matiére, Lyon, France
| | - Jean-Luc Coll
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Yannick Crémillieux
- Centre de Résonance Magnétique des Systèmes Biologiques,CNRS UMR 5536, Université Bordeaux, Bordeaux, France
| |
Collapse
|
25
|
Familial manganese-induced neurotoxicity due to mutations in SLC30A10 or SLC39A14. Neurotoxicology 2017; 64:278-283. [PMID: 28789954 DOI: 10.1016/j.neuro.2017.07.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 11/21/2022]
Abstract
Over the last few years, two rare, familial diseases that lead to the onset of manganese (Mn)-induced neurotoxicity have been discovered. Loss-of-function mutations in SLC30A10, a Mn efflux transporter, or SLC39A14, a Mn influx transporter, increase Mn levels in blood and brain, and induce severe neurotoxicity. The discoveries of these genetic diseases have transformed our understanding of Mn homeostasis, detoxification, and neurotoxicity. Current knowledge about the mechanisms by which mutations in these transporters alter Mn homeostasis to induce human disease is reviewed here.
Collapse
|
26
|
Li SJ, Qin WX, Peng DJ, Yuan ZX, He SN, Luo YN, Aschner M, Jiang YM, Liang DY, Xie BY, Xu F. Sodium P-aminosalicylic acid inhibits sub-chronic manganese-induced neuroinflammation in rats by modulating MAPK and COX-2. Neurotoxicology 2017; 64:219-229. [PMID: 28651968 DOI: 10.1016/j.neuro.2017.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/18/2017] [Accepted: 06/21/2017] [Indexed: 02/08/2023]
Abstract
Excessive manganese (Mn) accumulation in the brain may induce an extrapyramidal disorder known as manganism. Inflammatory processes play a critical role in neurodegenerative diseases. Therapeutically, non-steroidal anti-inflammatory drugs or analogous anti-inflammatory therapies have neuroprotective effects. As a non-steroidal anti-inflammatory drug, p-aminosalicylic acid (PAS) has anti-inflammatory effects, which are mediated by decreased prostaglandins E2 (PGE2) levels. The aim of the current study was to investigate whether PAS-Na treatment prevents Mn-induced behavioral changes and neuroinflammation in vivo. Male Sprague-Dawley rats were intraperitoneally (i.p.) injected with MnCl2·4H2O (15mg/kg) for 12 weeks, followed by 6 weeks PAS-Na treatment. Sub-chronic Mn exposure increased Mn levels in the whole blood, cortex, hippocampus and thalamus, and induced learning and memory deficits, concomitant with astrocytes activation in the cortex, hippocampus and thalamus. Moreover inflammatory cytokine levels in serum and brain of Mn-treated group were increased, including IL-1β, IL-6, TNF-αand PGE2, especially in the hippocampus and thalamus. Furthermore, sub-chronic Mn exposure also increased inflammatory cytokines and COX-2 in transcription levels concomitant with increased MAPK signaling and COX-2 in the same selected brain regions. PAS-Na treatment at the highest doses also decreased Mn levels in the whole blood and selected brain tissues, and reversed the Mn-induced learning and memory deficits. PAS-Na inhibited astrocyte activation as well as the Mn-induced increase in inflammatory cytokine levels, reducing p38, ERK MAPK pathway and COX-2 activity. In contrast PAS-Na had no effects on the JNK MAPK pathway. These data establish the efficacy of PAS-Na not only as a chelating agent to mobilize whole blood Mn, but also as an anti-inflammatory agent.
Collapse
Affiliation(s)
- Shao-Jun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Wen-Xia Qin
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Dong-Jie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Zong-Xiang Yuan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Sheng-Nan He
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Yi-Ni Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China.
| | - Dian-Yin Liang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Bing-Yan Xie
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Fang Xu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, PR China
| |
Collapse
|
27
|
Hutchens S, Liu C, Jursa T, Shawlot W, Chaffee BK, Yin W, Gore AC, Aschner M, Smith DR, Mukhopadhyay S. Deficiency in the manganese efflux transporter SLC30A10 induces severe hypothyroidism in mice. J Biol Chem 2017; 292:9760-9773. [PMID: 28461334 DOI: 10.1074/jbc.m117.783605] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/27/2017] [Indexed: 12/31/2022] Open
Abstract
Manganese is an essential metal that becomes toxic at elevated levels. Loss-of-function mutations in SLC30A10, a cell-surface-localized manganese efflux transporter, cause a heritable manganese metabolism disorder resulting in elevated manganese levels and parkinsonian-like movement deficits. The underlying disease mechanisms are unclear; therefore, treatment is challenging. To understand the consequences of loss of SLC30A10 function at the organism level, we generated Slc30a10 knock-out mice. During early development, knock-outs were indistinguishable from controls. Surprisingly, however, after weaning and compared with controls, knock-out mice failed to gain weight, were smaller, and died prematurely (by ∼6-8 weeks of age). At 6 weeks, manganese levels in the brain, blood, and liver of the knock-outs were ∼20-60-fold higher than controls. Unexpectedly, histological analyses revealed that the brain and liver of the knock-outs were largely unaffected, but their thyroid exhibited extensive alterations. Because hypothyroidism leads to growth defects and premature death in mice, we assayed for changes in thyroid and pituitary hormones. At 6 weeks and compared with controls, the knock-outs had markedly reduced thyroxine levels (∼50-80%) and profoundly increased thyroid-stimulating hormone levels (∼800-1000-fold), indicating that Slc30a10 knock-out mice develop hypothyroidism. Importantly, a low-manganese diet produced lower tissue manganese levels in the knock-outs and rescued the phenotype, suggesting that manganese toxicity was the underlying cause. Our unanticipated discovery highlights the importance of determining the role of thyroid dysfunction in the onset and progression of manganese-induced disease and identifies Slc30a10 knock-out mice as a new model for studying thyroid biology.
Collapse
Affiliation(s)
- Steven Hutchens
- From the Division of Pharmacology & Toxicology, College of Pharmacy, Institute for Cellular & Molecular Biology, and Institute for Neuroscience and
| | - Chunyi Liu
- From the Division of Pharmacology & Toxicology, College of Pharmacy, Institute for Cellular & Molecular Biology, and Institute for Neuroscience and
| | - Thomas Jursa
- the Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, California 95064
| | - William Shawlot
- the Mouse Genetic Engineering Facility, Institute for Cellular & Molecular Biology, University of Texas at Austin, Austin, Texas 78712
| | - Beth K Chaffee
- the Department of Veterinary Sciences, Michale E. Keeling Center for Comparative Medicine and Research, M. D. Anderson Cancer Center, Bastrop, Texas 78602, and
| | - Weiling Yin
- From the Division of Pharmacology & Toxicology, College of Pharmacy, Institute for Cellular & Molecular Biology, and Institute for Neuroscience and
| | - Andrea C Gore
- From the Division of Pharmacology & Toxicology, College of Pharmacy, Institute for Cellular & Molecular Biology, and Institute for Neuroscience and
| | - Michael Aschner
- the Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Donald R Smith
- the Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Somshuvra Mukhopadhyay
- From the Division of Pharmacology & Toxicology, College of Pharmacy, Institute for Cellular & Molecular Biology, and Institute for Neuroscience and
| |
Collapse
|
28
|
Bora S, Erdogan MA, Armagan G, Sevgili E, Dagcı T. Vinpocetine and Vasoactive Intestinal Peptide Attenuate Manganese-Induced Toxicity in NE-4C Cells. Biol Trace Elem Res 2016; 174:410-418. [PMID: 27206668 DOI: 10.1007/s12011-016-0742-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
Increased concentration of manganese (Mn) in the brain is known to be associated with excitotoxicity and neuroinflammation. Vinpocetine, an alkaloid derived from the plant Vinca minor L., basically shows its effect via phosphodiesterase inhibition and voltage-dependent Na+ channels. Vasoactive intestinal peptide (VIP) has gastrointestinal, vasomotor, muscular, and neuroprotective effects. The aim of this study was to examine the potential protective effects of vinpocetine and VIP against Mn toxicity in NE-4C neural stem cells (NSCs). VIP treatment at 1 μM and vinpocetine treatment at 2 μM concentrations were sufficient to yield maximum protection, and these concentrations were adopted in the following experiments. In this study, Mn treatment significantly increased lactate dehydrogenase (LDH) leakage, reactive oxygen species (ROS) production, and triggered cell death in NE-4C cultures. However, significant reduction in LDH release was observed following vinpocetine or VIP treatments when compared with control. Similar to these findings, vinpocetine or VIP treatments significantly reduced membrane degradation induced by Mn (p < 0.001). Moreover, vinpocetine attenuated Mn-induced decrease of mitochondrial membrane potential. Similarly, proapoptotic protein bax and ROS production significantly decreased in cells after incubation with vinpocetine (p = 0.01) or VIP in the presence of Mn (p < 0.001). Our study provides the evidence that both vinpocetine and VIP may exert protective effects via modulating oxidative stress and apoptosis in Mn-induced neurodegeneration in NE-4C cells.
Collapse
Affiliation(s)
- Saylav Bora
- Department of Physiology, School of Medicine, Faculty of Medicine, Ege University, 35100, Bornova, Izmir, Turkey.
| | - Mumin Alper Erdogan
- Department of Physiology, School of Medicine, Faculty of Medicine, Ege University, 35100, Bornova, Izmir, Turkey
| | - Güliz Armagan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, 35100, Bornova, Izmir, Turkey
| | - Elvin Sevgili
- Department of Biochemistry, Faculty of Pharmacy, Ege University, 35100, Bornova, Izmir, Turkey
| | - Taner Dagcı
- Department of Physiology, School of Medicine, Faculty of Medicine, Ege University, 35100, Bornova, Izmir, Turkey
| |
Collapse
|
29
|
Li Z, Liu Q, Liu C, Li C, Li Y, Li S, Liu X, Shao J. Evaluation of PFOS-mediated neurotoxicity in rat primary neurons and astrocytes cultured separately or in co-culture. Toxicol In Vitro 2016; 38:77-90. [PMID: 27825932 DOI: 10.1016/j.tiv.2016.11.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 01/28/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a potential neurotoxicant reported by epidemiological investigations and experimental studies, while the underlying mechanisms are still unclear. Astrocytes not only support for the construction of neurons, but also conduct neuronal functions through glutamate-glutamine cycle in astrocyte-neuron crosstalk. In the present study, the effect of PFOS exposure on rat primary hippocampal neurons or cortex astrocytes was evaluated. Then the role of the astrocytes in PFOS-induced toxic effect on neurons was explored with astrocyte-neuron co-culture system. Exposure of rat primary hippocampal neurons to PFOS has led to oxidation-antioxidation imbalance, increased apoptosis and abnormal autophagy. The adverse effect of PFOS on rat primary cortex astrocytes manifested in the form of altered extracellular glutamate and glutamine concentrations, decreased glutamine synthase activity, as well as decreased gene expression of glutamine synthase, glutamate transporters and glutamine transporters in the glutamate-glutamine cycle. Especially, the alleviation of PFOS-inhibited neurite outgrowth in neurons could be observed in astrocyte-neuron co-culture system, though the ability of astrocytes in fostering neurite outgrowth was affected by PFOS. These results indicated that both astrocytes and neurons might be the targets of PFOS-induced neurotoxicity, and astrocytes could protect against PFOS-inhibited neurite outgrowth in primary cultured neurons. Our research might render some information in explaining the mechanisms of PFOS-induced neurotoxicity.
Collapse
Affiliation(s)
- Zhenwei Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Qi Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Chang Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Chunna Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Yachen Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Shuangyue Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Xiaohui Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China.
| | - Jing Shao
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
30
|
Mikelman S, Mardirossian N, Gnegy ME. Tamoxifen and amphetamine abuse: Are there therapeutic possibilities? J Chem Neuroanat 2016; 83-84:50-58. [PMID: 27585851 DOI: 10.1016/j.jchemneu.2016.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/05/2016] [Accepted: 08/14/2016] [Indexed: 12/11/2022]
Abstract
Although best known as a selective estrogen receptor modulator (SERM), tamoxifen is a drug with a wide range of activities. Tamoxifen has demonstrated some efficacy has a therapeutic for bipolar mania and is believed to exert these effects through inhibition of protein kinase C (PKC). As the symptoms of amphetamine treatment in rodents are believed to mimic the symptoms of a manic episode, many of the preclinical studies for this indication have demonstrated that tamoxifen inhibits amphetamine action. The amphetamine-induced increase in extracellular dopamine which gives rise to the 'manic' effects is due to interaction of amphetamine with the dopamine transporter. We and others have demonstrated that PKC reduces amphetamine-induced reverse transport through the dopamine transporter. In this review, we will outline the actions of tamoxifen as a SERM and further detail another known action of tamoxifen-inhibition of PKC. We will summarize the literature showing how tamoxifen affects amphetamine action. Finally, we will present our hypothesis that tamoxifen, or an analog, could be used therapeutically to reduce amphetamine abuse in addition to treating mania.
Collapse
Affiliation(s)
- Sarah Mikelman
- Department of Pharmacology, 2220E MSRB III, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, MI 28109-5632, United States
| | - Natalie Mardirossian
- Department of Pharmacology, 2220E MSRB III, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, MI 28109-5632, United States
| | - Margaret E Gnegy
- Department of Pharmacology, 2220E MSRB III, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, MI 28109-5632, United States.
| |
Collapse
|
31
|
Khan MM, Wakade C, de Sevilla L, Brann DW. Selective estrogen receptor modulators (SERMs) enhance neurogenesis and spine density following focal cerebral ischemia. J Steroid Biochem Mol Biol 2015; 146:38-47. [PMID: 24815952 PMCID: PMC4419701 DOI: 10.1016/j.jsbmb.2014.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/23/2014] [Accepted: 05/02/2014] [Indexed: 12/23/2022]
Abstract
Selective estrogen receptor modulators (SERMs) have been reported to enhance synaptic plasticity and improve cognitive performance in adult rats. SERMs have also been shown to induce neuroprotection against cerebral ischemia and other CNS insults. In this study, we sought to determine whether acute regulation of neurogenesis and spine remodeling could be a novel mechanism associated with neuroprotection induced by SERMs following cerebral ischemia. Toward this end, ovariectomized adult female rats were either implanted with pellets of 17β-estradiol (estrogen) or tamoxifen, or injected with raloxifene. After one week, cerebral ischemia was induced by the transient middle-cerebral artery occlusion (MCAO) method. Bromodeoxyuridine (BrdU) was injected to label dividing cells in brain. We analyzed neurogenesis and spine density at day-1 and day-5 post MCAO. In agreement with earlier findings, we observed a robust induction of neurogenesis in the ipsilateral subventricular zone (SVZ) of both the intact as well as ovariectomized female rats following MCAO. Interestingly, neurogenesis in the ipsilateral SVZ following ischemia was significantly higher in estrogen and raloxifene-treated animals compared to placebo-treated rats. In contrast, this enhancing effect on neurogenesis was not observed in tamoxifen-treated rats. Finally, both SERMs, as well as estrogen significantly reversed the spine density loss observed in the ischemic cortex at day-5 post ischemia. Taken, together these results reveal a profound structural remodeling potential of SERMs in the brain following cerebral ischemia. This article is part of a Special Issue entitled "Sex steroids and brain disorders".
Collapse
Affiliation(s)
- Mohammad M Khan
- Department of Biochemistry, Faculty of Medicine, Zaiwa University, AZ-Zawia, Libya
| | - Chandramohan Wakade
- Department of Physical Therapy, Georgia Regents University, Augusta, GA 30912, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Liesl de Sevilla
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
32
|
Melatonin Antagonizes Mn-Induced Oxidative Injury Through the Activation of Keap1–Nrf2–ARE Signaling Pathway in the Striatum of Mice. Neurotox Res 2014; 27:156-71. [DOI: 10.1007/s12640-014-9489-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 07/19/2014] [Accepted: 07/28/2014] [Indexed: 10/24/2022]
|
33
|
Lan YL, Zhao J, Li S. Estrogen receptors' neuroprotective effect against glutamate-induced neurotoxicity. Neurol Sci 2014; 35:1657-62. [PMID: 25228013 DOI: 10.1007/s10072-014-1937-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/29/2014] [Indexed: 01/28/2023]
Abstract
Glutamate is the most abundant excitatory brain neurotransmitter that has important functional significance with respect to neurodegenerative conditions. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer's disease (AD) has been gradually becoming elucidated recently. Excessive release of glutamate induces an increase in intracellular Ca(2+) levels, thus triggers a cascade of cellular responses, ultimately leading to neuronal cell death. This type of neuronal damage induced by over-excitation has been proposed to be involved in a number of neuropathological conditions, ranging from acute insults to chronic neurodegenerative disorders. Estrogen could be effective in modulating glutamate-induced neurotoxicity and the protective responsivenesses are mostly estrogen receptors (ERs)-dependent. However, the mechanism underlying estrogen's neuroprotective effect is not fully clarified and is complicated by the presence of several distinct ER types. So a deeper research into the neuroprotection of ERs might be informative about the positive effect that estrogen might have on ageing-related cognitive changes. Extensive studies have indicated the neuroprotective effects of ERs against glutamate-induced neurotoxicity. The purpose of this review is to elucidate ERs' neuroprotective effects against glutamate-induced cytotoxicity and explore new ways to prevent and cure neurotoxicity-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | | | | |
Collapse
|
34
|
Genetic dys-regulation of astrocytic glutamate transporter EAAT2 and its implications in neurological disorders and manganese toxicity. Neurochem Res 2014; 40:380-8. [PMID: 25064045 DOI: 10.1007/s11064-014-1391-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/09/2014] [Accepted: 07/15/2014] [Indexed: 12/14/2022]
Abstract
Astrocytic glutamate transporters, the excitatory amino acid transporter (EAAT) 2 and EAAT1 (glutamate transporter 1 and glutamate aspartate transporter in rodents, respectively), are the main transporters for maintaining optimal glutamate levels in the synaptic clefts by taking up more than 90% of glutamate from extracellular space thus preventing excitotoxic neuronal death. Reduced expression and function of these transporters, especially EAAT2, has been reported in numerous neurological disorders, including amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, schizophrenia and epilepsy. The mechanism of down-regulation of EAAT2 in these diseases has yet to be fully established. Genetic as well as transcriptional dys-regulation of these transporters by various modes, such as single nucleotide polymorphisms and epigenetics, resulting in impairment of their functions, might play an important role in the etiology of neurological diseases. Consequently, there has been an extensive effort to identify molecular targets for enhancement of EAAT2 expression as a potential therapeutic approach. Several pharmacological agents increase expression of EAAT2 via nuclear factor κB and cAMP response element binding protein at the transcriptional level. However, the negative regulatory mechanisms of EAAT2 have yet to be identified. Recent studies, including those from our laboratory, suggest that the transcriptional factor yin yang 1 plays a critical role in the repressive effects of various neurotoxins, such as manganese (Mn), on EAAT2 expression. In this review, we will focus on transcriptional epigenetics and translational regulation of EAAT2.
Collapse
|
35
|
Karki P, Smith K, Johnson J, Lee E. Astrocyte-derived growth factors and estrogen neuroprotection: role of transforming growth factor-α in estrogen-induced upregulation of glutamate transporters in astrocytes. Mol Cell Endocrinol 2014; 389:58-64. [PMID: 24447465 PMCID: PMC4040305 DOI: 10.1016/j.mce.2014.01.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/09/2014] [Accepted: 01/09/2014] [Indexed: 01/31/2023]
Abstract
Extensive studies from the past decade have completely revolutionized our understanding about the role of astrocytes in the brain from merely supportive cells to an active role in various physiological functions including synaptic transmission via cross-talk with neurons and neuroprotection via releasing neurotrophic factors. Particularly, numerous studies have reported that astrocytes mediate the neuroprotective effects of 17β-estradiol (E2) and selective estrogen receptor modulators (SERMs) in various clinical and experimental models of neuronal injury. Astrocytes contain two main glutamate transporters, glutamate aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1), that play a key role in preventing excitotoxic neuronal death, a process associated with most neurodegenerative diseases. E2 has been shown to increase expression of both GLAST and GLT-1 mRNA and protein and glutamate uptake in astrocytes. Growth factors such as transforming growth factor-α (TGF-α) appear to mediate E2-induced enhancement of these transporters. These findings suggest that E2 exerts neuroprotection against excitotoxic neuronal injuries, at least in part, by enhancing astrocytic glutamate transporter levels and function. Therefore, the present review will discuss proposed mechanisms involved in astrocyte-mediated E2 neuroprotection, with a focus on glutamate transporters.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Keisha Smith
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - James Johnson
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Eunsook Lee
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA.
| |
Collapse
|
36
|
Karki P, Webb A, Zerguine A, Choi J, Son DS, Lee E. Mechanism of raloxifene-induced upregulation of glutamate transporters in rat primary astrocytes. Glia 2014; 62:1270-83. [PMID: 24782323 DOI: 10.1002/glia.22679] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 04/03/2014] [Accepted: 04/07/2014] [Indexed: 12/19/2022]
Abstract
Raloxifene (RX), a selective estrogen receptor modulator (SERM), exerts neuroprotection in multiple clinical and experimental settings. Astrocytic glutamate transporters GLT-1 (EAAT2) and GLAST (EAAT1) are the main glutamate transporters in the central nervous system, taking up most of excess glutamate from the synaptic cleft to prevent excitotoxic neuronal death. Since drugs targeting astrocytic glutamate transporters to enhance their expression and function represent potential therapeutics for neurodegenerative disorders associated with excitotoxicity, we tested if RX modulates the expression and function of GLT-1 and GLAST in rat primary astrocytes. The results showed that RX significantly increased glutamate uptake and expression of GLT-1 mRNA and protein levels. RX enhanced GLT-1 expression by the activation of multiple signaling pathways including ERK, EGFR, and CREB mediated by estrogen receptors (ERs) ER-α, ER-β, and GPR30. At the transcriptional level, NF-κB played a critical role in RX-induced GLT-1 expression as RX increased NF-κB reporter activity and induced binding of NF-κB p65 and p50 to the GLT-1 promoter. RX attenuated the reduction of GLT-1 expression and glutamate uptake induced by manganese (Mn) whose chronic high levels of exposure cause manganism. RX also upregulated GLAST by increasing its promoter activity and protein levels via the NF-κB pathway and ERs. Our findings provide new insight into the mechanism of RX-induced enhancement of GLT-1 and GLAST expression, as well as the attenuation of Mn-reduced expression of these transporters. These findings will be highly valuable for developing therapeutics of neurodegenerative diseases associated with impaired astrocytic glutamate transporters.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Physiology, Meharry Medical College, Nashville, Tennessee
| | | | | | | | | | | |
Collapse
|
37
|
Martinez-Finley EJ, Gavin CE, Aschner M, Gunter TE. Manganese neurotoxicity and the role of reactive oxygen species. Free Radic Biol Med 2013; 62:65-75. [PMID: 23395780 PMCID: PMC3713115 DOI: 10.1016/j.freeradbiomed.2013.01.032] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is an essential dietary nutrient, but an excess or accumulation can be toxic. Disease states, such as manganism, are associated with overexposure or accumulation of Mn and are due to the production of reactive oxygen species, free radicals, and toxic metabolites; alteration of mitochondrial function and ATP production; and depletion of cellular antioxidant defense mechanisms. This review focuses on all of the preceding mechanisms and the scientific studies that support them as well as providing an overview of the absorption, distribution, and excretion of Mn and the stability and transport of Mn compounds in the body.
Collapse
Affiliation(s)
- Ebany J Martinez-Finley
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | | | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN 37240, USA; Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN 37240, USA; The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN 37240, USA.
| | - Thomas E Gunter
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
38
|
Gobbo OL, Zurek M, Tewes F, Ehrhardt C, Crémillieux Y. Manganese: a new contrast agent for lung imaging? CONTRAST MEDIA & MOLECULAR IMAGING 2013; 7:542-6. [PMID: 22991321 DOI: 10.1002/cmmi.1483] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Lung parenchyma remains one of the most difficult tissues to be imaged by means of magnetic resonance imaging (MRI). Several MRI techniques are routinely used for lung imaging. However, manganese-enhancement MRI (MEMRI) technique has not been associated with pulmonary MRI. Here, we evaluated T(1) -enhancement in the rat lung after a manganese instillation, using a 4.7 T magnet with a radial ultrashort echo time sequence. Our data showed that the signal intensity was increased in lungs receiving a manganese solution compared with a control solution to the lungs. MR signal enhancements above 30% were measured in lung parenchyma following 200 µl instillation of a 1 mm manganese chloride solution. MEMRI, therefore, may be a useful novel tool for enhancing signal intensity and image contrast in lung tissue.
Collapse
Affiliation(s)
- Oliviero L Gobbo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | |
Collapse
|
39
|
Karki P, Lee E, Aschner M. Manganese neurotoxicity: a focus on glutamate transporters. Ann Occup Environ Med 2013; 25:4. [PMID: 24472696 PMCID: PMC3886254 DOI: 10.1186/2052-4374-25-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 03/01/2013] [Indexed: 11/15/2022] Open
Abstract
Manganese (Mn) is an essential element that is required in trace amount for normal growth, development as well maintenance of proper function and regulation of numerous cellular and biochemical reactions. Yet, excessive Mn brain accumulation upon chronic exposure to occupational or environmental sources of this metal may lead to a neurodegenerative disorder known as manganism, which shares similar symptoms with idiopathic Parkinson's disease (PD). In recent years, Mn exposure has gained public health interest for two primary reasons: continuous increased usage of Mn in various industries, and experimental findings on its toxicity, linking it to a number of neurological disorders. Since the first report on manganism nearly two centuries ago, there have been substantial advances in the understanding of mechanisms associated with Mn-induced neurotoxicity. This review will briefly highlight various aspects of Mn neurotoxicity with a focus on the role of astrocytic glutamate transporters in triggering its pathophysiology.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Physiology, Meharry Medical College, Nashville, TN, USA
| | - Eunsook Lee
- Department of Physiology, Meharry Medical College, Nashville, TN, USA
| | - Michael Aschner
- Department of Pediatrics, Vanderbilt University Medical Center, 2215-B Garland Avenue, 11415 MRB IV, Nashville, TN, 37232-0414, USA
| |
Collapse
|
40
|
Matamá T, Araújo R, Preto A, Cavaco-Paulo A, Gomes AC. In vitroinduction of melanin synthesis and extrusion by tamoxifen. Int J Cosmet Sci 2013; 35:368-74. [DOI: 10.1111/ics.12052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/06/2013] [Indexed: 01/25/2023]
Affiliation(s)
- T. Matamá
- CBMA (Centre of Molecular and Environmental Biology); Department of Biology; University of Minho; Campus of Gualtar 4710-057 Braga Portugal
- IBB - Institute for Biotechnology and Bioengineering; Centre of Biological Engineering; University of Minho; Campus de Gualtar 4710-057 Braga Portugal
| | - R. Araújo
- CBMA (Centre of Molecular and Environmental Biology); Department of Biology; University of Minho; Campus of Gualtar 4710-057 Braga Portugal
- IBB - Institute for Biotechnology and Bioengineering; Centre of Biological Engineering; University of Minho; Campus de Gualtar 4710-057 Braga Portugal
| | - A. Preto
- CBMA (Centre of Molecular and Environmental Biology); Department of Biology; University of Minho; Campus of Gualtar 4710-057 Braga Portugal
| | - A. Cavaco-Paulo
- IBB - Institute for Biotechnology and Bioengineering; Centre of Biological Engineering; University of Minho; Campus de Gualtar 4710-057 Braga Portugal
| | - A. C. Gomes
- CBMA (Centre of Molecular and Environmental Biology); Department of Biology; University of Minho; Campus of Gualtar 4710-057 Braga Portugal
| |
Collapse
|
41
|
Yamazaki T, Yamamoto M, Ishihara Y, Komatsu S, Munetsuna E, Onizaki M, Ishida A, Kawato S, Mukuda T. De novo synthesized estradiol protects against methylmercury-induced neurotoxicity in cultured rat hippocampal slices. PLoS One 2013; 8:e55559. [PMID: 23405170 PMCID: PMC3566000 DOI: 10.1371/journal.pone.0055559] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/27/2012] [Indexed: 11/24/2022] Open
Abstract
Background Estrogen, a class of female sex steroids, is neuroprotective. Estrogen is synthesized in specific areas of the brain. There is a possibility that the de novo synthesized estrogen exerts protective effect in brain, although direct evidence for the neuroprotective function of brain-synthesized estrogen has not been clearly demonstrated. Methylmercury (MeHg) is a neurotoxin that induces neuronal degeneration in the central nervous system. The neurotoxicity of MeHg is region-specific, and the molecular mechanisms for the selective neurotoxicity are not well defined. In this study, the protective effect of de novo synthesized 17β-estradiol on MeHg-induced neurotoxicity in rat hippocampus was examined. Methodology/Principal Findings Neurotoxic effect of MeHg on hippocampal organotypic slice culture was quantified by propidium iodide fluorescence imaging. Twenty-four-hour treatment of the slices with MeHg caused cell death in a dose-dependent manner. The toxicity of MeHg was attenuated by pre-treatment with exogenously added estradiol. The slices de novo synthesized estradiol. The estradiol synthesis was not affected by treatment with 1 µM MeHg. The toxicity of MeHg was enhanced by inhibition of de novo estradiol synthesis, and the enhancement of toxicity was recovered by the addition of exogenous estradiol. The neuroprotective effect of estradiol was inhibited by an estrogen receptor (ER) antagonist, and mimicked by pre-treatment of the slices with agonists for ERα and ERβ, indicating the neuroprotective effect was mediated by ERs. Conclusions/Significance Hippocampus de novo synthesized estradiol protected hippocampal cells from MeHg-induced neurotoxicity via ERα- and ERβ-mediated pathways. The self-protective function of de novo synthesized estradiol might be one of the possible mechanisms for the selective sensitivity of the brain to MeHg toxicity.
Collapse
Affiliation(s)
- Takeshi Yamazaki
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Buwalda B, Schagen SB. Is basic research providing answers if adjuvant anti-estrogen treatment of breast cancer can induce cognitive impairment? Life Sci 2013; 93:581-8. [PMID: 23353876 DOI: 10.1016/j.lfs.2012.12.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/27/2012] [Accepted: 12/21/2012] [Indexed: 12/23/2022]
Abstract
Adjuvant treatment of cancer by chemotherapy is associated with cognitive impairment in some cancer survivors. Breast cancer patients are frequently also receiving endocrine therapy with selective estrogen receptor modulators (SERMs) and/or aromatase inhibitors (AIs) to suppress the growth of estradiol sensitive breast tumors. Estrogens are well-known, however, to target brain areas involved in the regulation of cognitive behavior. In this review clinical and basic preclinical research is reviewed on the actions of estradiol, SERMs and AIs on brain and cognitive functioning to see if endocrine therapy potentially induces cognitive impairment and in that respect may contribute to the detrimental effects of chemotherapy on cognitive performance in breast cancer patients. Although many clinical studies may be underpowered to detect changes in cognitive function, current basic and clinical reports suggest that there is little evidence that AIs may have a lasting detrimental effect on cognitive performance in breast cancer patients. The clinical data on SERMs are not conclusive, but some studies do suggest that tamoxifen administration may form a risk for cognitive functioning particularly in older women. An explanation may come from basic preclinical research which indicates that tamoxifen often acts agonistic in the absence of estradiol but antagonistic in the presence of endogenous estradiol. It could be hypothesized that the negative effects of tamoxifen in older women is related to the so-called window of opportunity for estrogen. Administration of SERMs beyond this so-called window of opportunity may not be effective or might even have detrimental effects similar to estradiol.
Collapse
Affiliation(s)
- Bauke Buwalda
- Behavioral Physiology, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| | | |
Collapse
|
43
|
Luo L, Xu H, Li Y, Du Z, Sun X, Ma Z, Hu Y. Manganese-enhanced MRI optic nerve tracking: effect of intravitreal manganese dose on retinal toxicity. NMR IN BIOMEDICINE 2012; 25:1360-1368. [PMID: 22573611 DOI: 10.1002/nbm.2808] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 03/20/2012] [Accepted: 03/26/2012] [Indexed: 05/31/2023]
Abstract
The aim of this study was to provide data on the dose dependence of manganese-enhanced MRI (MEMRI) in the visual pathway of experimental rats and to study the toxicity of MnCl₂ to the retina. Sprague-Dawley rats were intravitreally injected with 2 μL of 0, 10, 25, 50, 75, 100, 150 and 300 mM MnCl₂, respectively. The contrast-to-noise ratio (CNR) of MEMRI for optic nerve enhancement was measured at different concentrations of MnCl₂. Simultaneously, the toxicity of manganese was evaluated by counting retinal ganglion cells and by retinal histological examination using light microscopy and transmission electron microscopy. The CNR increased with increasing concentration of MnCl₂ up to 75 mM. Retinal ganglion cell densities were reduced significantly when the concentration of MnCl₂ in the intravitreal injection was equal to or greater than 75 mM. Increasing numbers of ribosomes in retinal ganglion cells were first detected at 25 mM of MnCl₂. The retinal toxicity of MnCl₂ at higher concentration also included mitochondrial pathology and cell disruption of retinal ganglion cells, as well as abnormalities of photoreceptor and retinal pigment epithelium cells. It can be concluded that intravitreal injection of MnCl₂ induces retinal cell damage that appears to start from 25 mM. The concentration of MnCl₂ should not exceed 25 mm through intravitreal injection for visual pathway MEMRI in the rat.
Collapse
Affiliation(s)
- Lisha Luo
- Peking University Eye Center, Peking University Third Hospital, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Lee E, Sidoryk-Wegrzynowicz M, Farina M, Rocha JBT, Aschner M. Estrogen attenuates manganese-induced glutamate transporter impairment in rat primary astrocytes. Neurotox Res 2012; 23:124-30. [PMID: 22878846 DOI: 10.1007/s12640-012-9347-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
Abstract
The astrocytic glutamate transporters (GLT-1, GLAST) are critical for removing excess glutamate from synaptic sites, thereby maintaining glutamate homeostasis within the brain. 17β-Estradiol (E2) is one of the most active estrogen hormones possessing neuroprotective effects both in in vivo and in vitro models, and it has been shown to enhance astrocytic glutamate transporter function (Liang et al. in J Neurochem 80:807-814, 2002; Pawlak et al. in Brain Res Mol Brain Res 138:1-7, 2005). However, E2 is not clinically optimal for neuroprotection given its peripheral feminizing and proliferative effects; therefore, brain selective estrogen receptor modulators (neuro SERMs) (Zhao et al. in Neuroscience 132:299-311, 2005) that specifically target estrogenic mechanisms, but lack the systemic estrogen side effects offer more promising therapeutic modality for the treatment of conditions associated with excessive synaptic glutamate levels. This review highlights recent studies from our laboratory showing that E2 and SERMs effectively reverse glutamate transport inhibition in a manganese (Mn)-induced model of glutamatergic deregulation. Specifically, we discuss mechanisms by which E2 restores the expression and activity of glutamate uptake. We advance the hypothesis that E2 and related compounds, such as tamoxifen may offer a potential therapeutic modality in neurodegenerative disorders, which are characterized by altered glutamate homeostasis.
Collapse
Affiliation(s)
- Eunsook Lee
- Department of Physiology, Meharry Medical College, Nashville, TN 37208, USA.
| | | | | | | | | |
Collapse
|
45
|
Lee E, Sidoryk-Wegrzynowicz M, Yin Z, Webb A, Son DS, Aschner M. Transforming growth factor-α mediates estrogen-induced upregulation of glutamate transporter GLT-1 in rat primary astrocytes. Glia 2012; 60:1024-36. [PMID: 22488924 PMCID: PMC3353324 DOI: 10.1002/glia.22329] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 02/28/2012] [Indexed: 01/02/2023]
Abstract
Glutamate transporter-1 (GLT-1) plays a central role in preventing excitotoxicity by removing excess glutamate from the synaptic clefts. 17β-Estradiol (E2) and tamoxifen (TX), a selective estrogen receptor (ER) modulator, afford neuroprotection in a range of experimental models. However, the mechanisms that mediate E2 and TX neuroprotection have yet to be elucidated. We tested the hypothesis that E2 and TX enhance GLT-1 function by increasing transforming growth factor (TGF)-α expression and, thus, attenuate manganese (Mn)-induced impairment in astrocytic GLT-1 expression and glutamate uptake in rat neonatal primary astrocytes. The results showed that E2 (10 nM) and TX (1 μM) increased GLT-1 expression and reversed the Mn-induced reduction in GLT-1, both at the mRNA and protein levels. E2/TX also concomitantly reversed the Mn-induced inhibition of astrocytic glutamate uptake. E2/TX activated the GLT-1 promoter and attenuated the Mn-induced repression of the GLT-1 promoter in astrocytes. TGF-α knockdown (siRNA) abolished the E2/TX effect on GLT-1 expression, and inhibition of epidermal growth factor receptor (TGF-α receptor) suppressed the effect of E2/TX on GLT-1 expression and GLT-1 promoter activity. E2/TX also increased TGF-α mRNA and protein levels with a concomitant increase in astrocytic glutamate uptake. All ERs (ER-α, ER-β, and G protein-coupled receptor 30) were involved in mediating E2 effects on the regulation of TGF-α, GLT-1, and glutamate uptake. These results indicate that E2/TX increases GLT-1 expression in astrocytes via TGF-α signaling, thus offering an important putative target for the development of novel therapeutics for neurological disorders.
Collapse
Affiliation(s)
- Eunsook Lee
- Department of Physiology, Meharry Medical College, Nashville, TN 37208, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Lee E, Sidoryk-Wêgrzynowicz M, Wang N, Webb A, Son DS, Lee K, Aschner M. GPR30 regulates glutamate transporter GLT-1 expression in rat primary astrocytes. J Biol Chem 2012; 287:26817-28. [PMID: 22645130 DOI: 10.1074/jbc.m112.341867] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The G protein-coupled estrogen receptor GPR30 contributes to the neuroprotective effects of 17β-estradiol (E2); however, the mechanisms associated with this protection have yet to be elucidated. Given that E2 increases astrocytic expression of glutamate transporter-1 (GLT-1), which would prevent excitotoxic-induced neuronal death, we proposed that GPR30 mediates E2 action on GLT-1 expression. To investigate this hypothesis, we examined the effects of G1, a selective agonist of GPR30, and GPR30 siRNA on astrocytic GLT-1 expression, as well as glutamate uptake in rat primary astrocytes, and explored potential signaling pathways linking GPR30 to GLT-1. G1 increased GLT-1 protein and mRNA levels, subject to regulation by both MAPK and PI3K signaling. Inhibition of TGF-α receptor suppressed the G1-induced increase in GLT-1 expression. Silencing GPR30 reduced the expression of both GLT-1 and TGF-α and abrogated the G1-induced increase in GLT-1 expression. Moreover, the G1-induced increase in GLT-1 protein expression was abolished by a protein kinase A inhibitor and an NF-κB inhibitor. G1 also enhanced cAMP response element-binding protein (CREB), as well as both NF-κB p50 and NF-κB p65 binding to the GLT-1 promoter. Finally, to model dysfunction of glutamate transporters, manganese was used, and G1 was found to attenuate manganese-induced impairment in GLT-1 protein expression and glutamate uptake. Taken together, the present data demonstrate that activation of GPR30 increases GLT-1 expression via multiple pathways, suggesting that GPR30 is worthwhile as a potential target to be explored for developing therapeutics of excitotoxic neuronal injury.
Collapse
Affiliation(s)
- Eunsook Lee
- Department of Physiology, Meharry Medical College, Nashville, Tennessee 37208, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Bourque M, Dluzen DE, Di Paolo T. Signaling pathways mediating the neuroprotective effects of sex steroids and SERMs in Parkinson's disease. Front Neuroendocrinol 2012; 33:169-78. [PMID: 22387674 DOI: 10.1016/j.yfrne.2012.02.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 02/15/2012] [Accepted: 02/21/2012] [Indexed: 11/27/2022]
Abstract
Studies with the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) animal model of Parkinson's disease have shown the ability of 17β-estradiol to protect the nigrostriatal dopaminergic system. This paper reviews the signaling pathways mediating the neuroprotective effect of 17β-estradiol against MPTP-induced toxicity. The mechanisms of 17β-estradiol action implicate activation of signaling pathways such as the phosphatidylinositol-3 kinase/Akt and the mitogen-activated protein kinase pathways. 17β-estradiol signaling is complex and integrates multiple interactions with signaling molecules that act to potentiate a protective effect. 17β-estradiol signaling is mediated via estrogen receptors, including GPER1, but others receptors, such as the IGF-1 receptor, are implicated in the neuroprotective effect. Glial and neuronal crosstalk is a critical factor in the maintenance of dopamine neuronal survival and in the neuroprotective action of 17β-estradiol. Compounds that stimulate GPER1 such as selective estrogen receptor modulators and phytoestrogens show neuroprotective activity and are alternatives to 17β-estradiol.
Collapse
Affiliation(s)
- Mélanie Bourque
- Molecular Endocrinology and Genomic Research Center, Centre de recherche du CHUQ (CHUL), Quebec City, QC, Canada G1V 4G2
| | | | | |
Collapse
|
49
|
Zhu G, Wang X, Wu S, Li Q. Involvement of activation of PI3K/Akt pathway in the protective effects of puerarin against MPP+-induced human neuroblastoma SH-SY5Y cell death. Neurochem Int 2012; 60:400-8. [PMID: 22265823 DOI: 10.1016/j.neuint.2012.01.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 12/28/2011] [Accepted: 01/04/2012] [Indexed: 01/01/2023]
Abstract
In an attempt to clarify the protective effect of puerarin on toxin-insulted dopaminergic neuronal death, this present study was carried out by using a typical Parkinson's disease (PD) model - 1-methyl-4-phenylpyridinium iodide (MPP(+))-induced dopaminergic SH-SY5Y cellular model. Data are presented, which showed that puerarin up-regulated Akt phosphorylation in both of MPP(+)-treated and non-MPP(+)-treated cells. The presence of PI3K inhibitor LY294002 completely blocked puerarin-induced activation of Akt phosphorylation. Moreover, puerarin decreased MPP(+)-induced cell death, which was blocked by phosphoinositide 3-kinase (PI3K) inhibitor LY294002. We further demonstrated that puerarin protected against MPP(+)-induced p53 nuclear accumulation, Puma (p53-upregulated mediator of apoptosis) and Bax expression and caspase-3-dependent programmed cell death (PCD). This protection was blocked by applying a PI3K/Akt inhibitor. Additionally, it was Pifithrin-α, but not Pifithrin-μ, which blocked MPP(+)-induced Puma and Bax expression, caspase-3 activation and cell death. Collectively, these data suggest that the activation of PI3K/Akt pathway is involved in the protective effect of puerarin against MPP(+)-induced neuroblastoma SH-SY5Y cell death through inhibiting nuclear p53 accumulation and subsequently caspase-3-dependent PCD. Puerarin might be a potential therapeutic agent for PD.
Collapse
Affiliation(s)
- Guoqi Zhu
- Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Traditional Chinese Medicine, Hefei, China
| | | | | | | |
Collapse
|
50
|
Arevalo MA, Diz-Chaves Y, Santos-Galindo M, Bellini MJ, Garcia-Segura LM. Selective oestrogen receptor modulators decrease the inflammatory response of glial cells. J Neuroendocrinol 2012; 24:183-90. [PMID: 21564348 DOI: 10.1111/j.1365-2826.2011.02156.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuroinflammation comprises a feature of many neurological disorders that is accompanied by the activation of glial cells and the release of pro-inflammatory cytokines and chemokines. Such activation is a normal response oriented to protect neural tissue and it is mainly regulated by microglia and astroglia. However, excessive and chronic activation of glia may lead to neurotoxicity and may be harmful for neural tissue. The ovarian hormone oestradiol exerts protective actions in the central nervous system that, at least in part, are mediated by a reduction of reactive gliosis. Several selective oestrogen receptor modulators may also exert neuroprotective effects by controlling glial inflammatory responses. Thus, tamoxifen and raloxifene decrease the inflammatory response caused by lipopolysaccharide, a bacterial endotoxin, in mouse and rat microglia cells in vitro. Tamoxifen and raloxifene are also able to reduce microglia activation in the brain of male and female rats in vivo after the peripheral administration of lipopolysaccharide. In addition, tamoxifen decreases the microglia inflammatory response induced by irradiation. Furthermore, treatment with tamoxifen and raloxifene resulted in a significant reduction of the number of reactive astrocytes in the hippocampus of young, middle-aged and older female rats after a stab wound injury. Tamoxifen, raloxifene and the new selective oestrogen receptor modulators ospemifene and bazedoxifene decrease the expression and release of interleukine-6 and interferon-γ inducible protein-10 in cultured astrocytes exposed to lipopolysaccharide. Ospemifene and bazedoxifene exert anti-inflammatory effects in astrocytes by a mechanism involving classical oestrogen receptors and the inhibition of nuclear factor-kappa B p65 transactivation. These data suggest that oestrogenic compounds are candidates to counteract brain inflammation under neurodegenerative conditions by targeting the production and release of pro-inflammatory molecules by glial cells.
Collapse
|