1
|
Zakaria D, Yamashita T, Kosugi Y. Multi-Omics Analysis of Hippocampus in Rats Administered Trimethyltin Chloride. Neurotox Res 2025; 43:13. [PMID: 40095120 PMCID: PMC11914309 DOI: 10.1007/s12640-025-00737-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Trimethyltin chloride (TMT) is a neurotoxicant that damages the central nervous system (CNS) and triggers neurodegeneration. This study used multi-omic data, including transcriptomics and proteomics of the rat hippocampus, to identify differentially expressed genes and proteins in TMT-induced neurotoxicity over time, related to neuro-axonal damage marked by plasma Neurofilament Light (NfL) levels. Data were collected at 12, 24, 48, 72, and 168 h post-TMT administration. NfL levels surged at 72 and 168 h, confirming neuro-axonal damage. Transcripts of genes in the chemokine signaling pathway (Cxcl10, Cxcl12, Cxcl14, Cxcl16), apoptosis pathway (Caspase-3, PARP1, CTSD), and TNF signaling pathway (TNFR1, MMP9, ICAM-1, TRAF3) showed significant differential expression starting from 48 h, preceding the NfL increase, suggesting their roles in neuro-axonal damage. Additionally, 11 Alzheimer's disease-related proteins, with significant changes from 72 to 168 h, were detected only in the proteomic dataset, indicating post-translational modifications might be crucial in neurotoxicity. Pathway analysis revealed that neurodegeneration and Alzheimer's disease pathways were among the top 15 affected by TMT-induced gene regulation, aligning with the involvement of TNF signaling, apoptosis, and chemokine signaling in neurodegeneration. This research highlighted the value of longitudinal omics studies, combined with pathway enrichment, gene-disease association, and neuro-axonal damage biomarker analyses, to elucidate neurotoxicant-induced neurodegeneration. Findings from this study could enhance the understanding of TMT-induced neurotoxicity, potentially informing future therapeutic strategies and preventive measures.
Collapse
Affiliation(s)
- Douaa Zakaria
- DMPK&Modeling, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
- The Graduate University for Advanced Studies, SOKENDAI, Miura, Kanagawa, Japan
| | - Tomoki Yamashita
- DMPK&Modeling, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Yohei Kosugi
- DMPK&Modeling, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan.
| |
Collapse
|
2
|
Inoue-Yanagimachi M, Himori N, Uchida K, Tawarayama H, Sato K, Yamamoto M, Namekata K, Harada T, Nakazawa T. Changes in glial cells and neurotrophic factors due to rotenone-induced oxidative stress in Nrf2 knockout mice. Exp Eye Res 2023; 226:109314. [PMID: 36400285 DOI: 10.1016/j.exer.2022.109314] [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: 04/05/2022] [Revised: 10/22/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022]
Abstract
Glaucoma is one of the most common causes of blindness worldwide. It is thought to be a multifactorial disease with underlying mechanisms that include mitochondrial dysfunction and oxidative stress. Here, we used NF-E2 related factor 2 (Nrf2) knockout (KO) mice, which are vulnerable to oxidative stress, to examine a neuroprotective effect against oxidative stress due to rotenone, a mitochondrial complex I inhibitor. Wild-type (WT) and Nrf2 KO mice received an oral solution of rotenone for 30 days. We then extracted the retinas and performed immunohistochemistry and quantitative RT-PCR. We also prepared a primary Müller cell culture of samples from each mouse, added 30 μM rotenone, and then measured cell viability, cytotoxicity and CellRox absorbance. We also examined gene expression. We found a significant increase in the number of 8-OHdG-positive retinal ganglion cells (RGCs) after rotenone administration in both the WT and Nrf2 KO mice. There was no difference in the number of RNA-binding protein with multiple splicing (RBPMS)-positive RGCs in the WT and Nrf2 KO mice, but Nrf2 KO mice that were given rotenone had significantly less retinal gene expression of RBPMS than Nrf2 KO mice given a control. Moreover, there was significantly higher mRNA gene expression of vimentin and glial fibrillary acidic protein (GFAP) in Nrf2 KO mice that received rotenone than WT mice that received rotenone. A statistical analysis of the in vitro experiment showed that cell viability was lower, cytotoxicity was higher, and oxidative stress was higher in the Müller cells of the Nrf2 KO mice than the WT mice. Finally, brain-derived neurotrophic factor (BDNF) and basic fibroblast growth factor (bFGF) were significantly higher in the Müller cells of the Nrf2 KO mice than the WT mice. These findings suggest that in Nrf2 KO mice under oxidative stress caused by rotenone, temporary neurotrophic factors are secreted from the Müller cells, conferring neuroprotection in these cells.
Collapse
Affiliation(s)
- Maki Inoue-Yanagimachi
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriko Himori
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Aging Vision Healthcare, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Keiko Uchida
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Tawarayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan; Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
3
|
Jiang Y, Guo J, Tang X, Wang X, Hao D, Yang H. The Immunological Roles of Olfactory Ensheathing Cells in the Treatment of Spinal Cord Injury. Front Immunol 2022; 13:881162. [PMID: 35669779 PMCID: PMC9163387 DOI: 10.3389/fimmu.2022.881162] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 01/16/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating type of neurological disorder of the central nervous system (CNS) with high mortality and disability. The pathological processes of SCI can usually be described as two stages, namely, primary and acute secondary injuries. Secondary injury produces more significant exacerbations of the initial injury. Among all the mechanisms of secondary damage, infection and inflammatory responses, as the principle culprits in initiating the second phase of SCI, can greatly contribute to the severity of SCI and numerous sequelae after SCI. Therefore, effectively antagonizing pro-inflammatory responses may be a promising treatment strategy to facilitate functional recovery after SCI. Olfactory ensheathing cells (OECs), a unique type of glial cells, have increasingly become potential candidates for cell-based therapy in the injured CNS. Strikingly, there is growing evidence that the mechanisms underlying the anti-inflammatory role of OECs are associated with the immune properties and secretory functions of these cells responsible for anti-neuroinflammation and immunoregulatory effects, leading to maintenance of the internal microenvironment. Accordingly, a more profound understanding of the mechanism of OEC immunological functions in the treatment of SCI would be beneficial to improve the therapeutic clinical applications of OECs for SCI. In this review, we mainly summarize recent research on the cellular and molecular immune attributes of OECs. The unique biological functions of these cells in promoting neural regeneration are discussed in relation of the development of novel therapies for CNS injury.
Collapse
Affiliation(s)
- Yizhen Jiang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jianbin Guo
- Department of Joint Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiangwen Tang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Xiaohui Wang
- Department of Spine Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Dingjun Hao
- Department of Spine Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
4
|
Denuzière A, Ghersi-Egea JF. Cerebral concentration and toxicity of endocrine disrupting chemicals: The implication of blood-brain interfaces. Neurotoxicology 2022; 91:100-118. [DOI: 10.1016/j.neuro.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 11/28/2022]
|
5
|
Dragić M, Mitrović N, Adžić M, Nedeljković N, Grković I. Microglial- and Astrocyte-Specific Expression of Purinergic Signaling Components and Inflammatory Mediators in the Rat Hippocampus During Trimethyltin-Induced Neurodegeneration. ASN Neuro 2021; 13:17590914211044882. [PMID: 34569324 PMCID: PMC8495514 DOI: 10.1177/17590914211044882] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present study examined the involvement of purinergic signaling components in
the rat model of hippocampal degeneration induced by trimethyltin (TMT)
intoxication (8 mg/kg, single intraperitoneal injection), which results in
behavioral and neurological dysfunction similar to neurodegenerative disorders.
We investigated spatial and temporal patterns of ecto-nucleoside triphosphate
diphosphohydrolase 1 (NTPDase1/CD39) and ecto-5′ nucleotidase (eN/CD73)
activity, their cell-specific localization, and analyzed gene expression pattern
and/or cellular localization of purinoreceptors and proinflammatory mediators
associated with reactive glial cells. Our study demonstrated that all Iba1+
cells at the injured area, irrespective of their morphology, upregulated
NTPDase1/CD39, while induction of eN/CD73 has been observed at amoeboid Iba1+
cells localized within the hippocampal neuronal layers with pronounced cell
death. Marked induction of P2Y12R, P2Y6R, and
P2X4-messenger RNA at the early stage of TMT-induced
neurodegeneration might reflect the functional properties, migration, and
chemotaxis of microglia, while induction of P2X7R at amoeboid cells
probably modulates their phagocytic role. Reactive astrocytes expressed
adenosine A1, A2A, and P2Y1 receptors, revealed
induction of complement component C3, inducible nitric oxide synthase, nuclear
factor-kB, and proinflammatory cytokines at the late stage of TMT-induced
neurodegeneration. An increased set of purinergic system components on activated
microglia (NTPDase1/CD39, eN/CD73, and P2X7) and astrocytes
(A1R, A2AR, and P2Y1), and loss of
homeostatic glial and neuronal purinergic pathways (P2Y12 and
A1R) may shift purinergic signaling balance toward excitotoxicity
and inflammation, thus favoring progression of pathological events. These
findings may contribute to a better understanding of the involvement of
purinergic signaling components in the progression of neurodegenerative
disorders that could be target molecules for the development of novel
therapies.
Collapse
Affiliation(s)
- Milorad Dragić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Nataša Mitrović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences-National Institute of thе Republic of Serbia, 89101University of Belgrade, Belgrade, Serbia
| | - Marija Adžić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia.,Center for Laser Microscopy, 98829Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Nadežda Nedeljković
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Ivana Grković
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences-National Institute of thе Republic of Serbia, 89101University of Belgrade, Belgrade, Serbia
| |
Collapse
|
6
|
Mataram MBA, Hening P, Harjanti FN, Karnati S, Wasityastuti W, Nugrahaningsih DAA, Kusindarta DL, Wihadmadyatami H. The neuroprotective effect of ethanolic extract Ocimum sanctum Linn. in the regulation of neuronal density in hippocampus areas as a central autobiography memory on the rat model of Alzheimer's disease. J Chem Neuroanat 2020; 111:101885. [PMID: 33188864 DOI: 10.1016/j.jchemneu.2020.101885] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 11/24/2022]
Abstract
The aim of this study was to identify the effects of Ocimum sanctum Linn. ethanolic extract (OSE) on the neurons of the CA1, CA3, and DG hippocampal areas with the use of in vivo and in vitro models of Alzheimer's diseases (AD). Twenty-one two-month-old male rats were divided into three groups: untreated (Group A, n = 3), AD rats model pretreated with OSE followed by induction for Trimethyltin (TMT) on day 7 (group B, n = 9), and AD rats model treated with OSE both as pre-TMT introduction for 7 days and post-TMT induction for 21 days (group C, n = 9). AD rats were sacrificed on days 7, 14, and 21, and brain samples were collected and analyzed for neuronal density and neuropeptide Y (NPY) immunoreactivity. To corroborate the in vivo observations, HEK-293 cells were treated with TMT and used as an in vitro model of AD. The results were then analyzed using FITC Annexin V and flow cytometry. Nuclear fragmentation was observed in cells stained with Hoechst 33342 by confocal microscopy. The results showed a significant increase in the number of neurons and NPY expression in the AD rats that were pre- and post-treated with OSE (p < 0.05). Indeed, OSE was able to retain and promote neuronal density in the rat model of AD. Further studies of an in vitro model of neurodegeneration with Ocimum sanctum Linn. ethanolic extract inhibited apoptosis in TMT-induced HEK-293 cells. Moreover, OSE prevented nuclear fragmentation, which was confirmed by staining the nuclei of HEK-293 cells. Taken together, there findings suggest that OSE has the potential as a neuroprotective agent (retaining the autobiographical memory),and the neuroproliferation of neurons in the CA1, CA3, and DG hippocampal areas in the rats¡ model of neurodegeneration was mediated by activation of NPY expression.
Collapse
Affiliation(s)
| | - Puspa Hening
- Integrated Laboratory for Research and Testing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Fitria N Harjanti
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Srikanth Karnati
- Department of Anatomy and Cell Biology, Julius Maxilimilian University Wurzburg, Germany
| | - Widya Wasityastuti
- Department of Physiology, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | - Dwi Liliek Kusindarta
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Hevi Wihadmadyatami
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| |
Collapse
|
7
|
Autophagy in trimethyltin-induced neurodegeneration. J Neural Transm (Vienna) 2020; 127:987-998. [PMID: 32451631 DOI: 10.1007/s00702-020-02210-1] [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: 04/11/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
Autophagy is a degradative process playing an important role in removing misfolded or aggregated proteins, clearing damaged organelles, such as mitochondria and endoplasmic reticulum, as well as eliminating intracellular pathogens. The autophagic process is important for balancing sources of energy at critical developmental stages and in response to nutrient stress. Recently, autophagy has been involved in the pathophysiology of neurodegenerative diseases although its beneficial (pro-survival) or detrimental (pro-death) role remains controversial. In the present review, we discuss the role of autophagy following intoxication with trimethyltin (TMT), an organotin compound that induces severe hippocampal neurodegeneration associated with astrocyte and microglia activation. TMT is considered a useful tool to study the molecular mechanisms occurring in human neurodegenerative diseases such as Alzheimer's disease and temporal lobe epilepsy. This is also relevant in the field of environmental safety, since organotin compounds are used as heat stabilizers in polyvinyl chloride polymers, industrial and agricultural biocides, and as industrial chemical catalysts.
Collapse
|
8
|
O'Brien JJ, O'Callaghan JP, Miller DB, Chalgeri S, Wennogle LP, Davis RE, Snyder GL, Hendrick JP. Inhibition of calcium-calmodulin-dependent phosphodiesterase (PDE1) suppresses inflammatory responses. Mol Cell Neurosci 2019; 102:103449. [PMID: 31770590 PMCID: PMC7783477 DOI: 10.1016/j.mcn.2019.103449] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/01/2019] [Accepted: 11/21/2019] [Indexed: 11/08/2022] Open
Abstract
A novel, potent, and highly specific inhibitor of calcium-calmodulin-dependent phosphodiesterases (PDE) of the PDE1 family, ITI-214, was used to investigate the role of PDE1 in inflammatory responses. ITI-214 dose-dependently suppressed lipopolysaccharide (LPS)-induced gene expression of pro-inflammatory cytokines in an immortalized murine microglial cell line, BV2 cells. RNA profiling (RNA-Seq) was used to analyze the impact of ITI-214 on the BV2 cell transcriptome in the absence and the presence of LPS. ITI-214 was found to regulate classes of genes that are involved in inflammation and cell migration responses to LPS exposure. The gene expression changes seen with ITI-214 treatment were distinct from those elicited by inhibitors of other PDEs with anti-inflammatory activity (e.g., a PDE4 inhibitor), indicating a distinct mechanism of action for PDE1. Functionally, ITI-214 inhibited ADP-induced migration of BV2 cells through a P2Y12-receptor-dependent pathway, possibly due to increases in the extent of cAMP and VASP phosphorylation downstream of receptor activation. Importantly, this effect was recapitulated in P2 rat microglial cells in vitro, indicating that these pathways are active in native microglial cells. These studies are the first to demonstrate that inhibition of PDE1 exerts anti-inflammatory effects through effects on microglia signaling pathways. The ability of PDE1 inhibitors to prevent or dampen excessive inflammatory responses of BV2 cells and microglia provides a basis for exploring their therapeutic utility in the treatment of neurodegenerative diseases associated with increased inflammation and microglia proliferation such as Parkinson's disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Jennifer J O'Brien
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - James P O'Callaghan
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Diane B Miller
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Suman Chalgeri
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Lawrence P Wennogle
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Robert E Davis
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Gretchen L Snyder
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America.
| | - Joseph P Hendrick
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| |
Collapse
|
9
|
Dragić M, Zarić M, Mitrović N, Nedeljković N, Grković I. Two Distinct Hippocampal Astrocyte Morphotypes Reveal Subfield-Different Fate during Neurodegeneration Induced by Trimethyltin Intoxication. Neuroscience 2019; 423:38-54. [PMID: 31682945 DOI: 10.1016/j.neuroscience.2019.10.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 12/19/2022]
Abstract
Astrocytes comprise a heterogenic group of glial cells, which perform homeostatic functions in the central nervous system. These cells react to all kind of insults by changing the morphology and function that result in a transition from the quiescent to a reactive phenotype. Trimethyltin (TMT) intoxication, which reproduces pathological events in the hippocampus similar to those associated with seizures and cognitive decline, has been proven as a useful model for studying responses of the glial cells to neurodegeneration. In the present study, we have explored morphological varieties of astrocytes in the hippocampal subregions of ovariectomized female rats exposed to TMT. We have demonstrated an early loss of neurons in CA1 and DG subfields. Distinct morphotypes of protoplasmic astrocytes observed in CA1/CA3 and the hilus of control animals developed different responses to TMT intoxication, as assessed by GFAP-immunohistochemistry. In CA1 subregion, GFAP+ astrocytes preserved their domain organization and responded with typical hypertrophy, while the hilar GFAP+ astrocytes developed atrophy-like phenotype and increased expression of vimentin and nestin 7 days after the exposure. Both reactive and atrophied-like astrocytes expressed Kir4.1 in CA1/CA3 and the hilus of DG, respectively, indicating that these cells did not change their potential for normal activity at this time point of pathology. Together, the results demonstrate the persistence of two protoplasmic morphotypes of astrocytes, with distinct appearance, function, and fate after TMT-induced neurodegeneration, suggesting their pleiotropic roles in the hippocampal response to neurodegeneration.
Collapse
Affiliation(s)
- Milorad Dragić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Studentski trg 3, 11001 Belgrade, Serbia; Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia.
| | - Marina Zarić
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia
| | - Nataša Mitrović
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia
| | - Nadežda Nedeljković
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Studentski trg 3, 11001 Belgrade, Serbia
| | - Ivana Grković
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia
| |
Collapse
|
10
|
Trettel F, Di Castro MA, Limatola C. Chemokines: Key Molecules that Orchestrate Communication among Neurons, Microglia and Astrocytes to Preserve Brain Function. Neuroscience 2019; 439:230-240. [PMID: 31376422 DOI: 10.1016/j.neuroscience.2019.07.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/01/2019] [Accepted: 07/19/2019] [Indexed: 12/19/2022]
Abstract
In the CNS, chemokines and chemokine receptors are involved in pleiotropic physiological and pathological activities. Several evidences demonstrated that chemokine signaling in the CNS plays key homeostatic roles and, being expressed on neurons, glia and endothelial cells, chemokines mediate the bidirectional cross-talk among parenchymal cells. An efficient communication between neurons and glia is crucial to establish and maintain a healthy brain environment which ensures normal functionality. Glial cells behave as active sensors of environmental changes induced by neuronal activity or detrimental insults, supporting and exerting neuroprotective activities. In this review we summarize the evidence that chemokines (CXCL12, CX3CL1, CXCL16 and CCL2) modulate neuroprotective processes upon different noxious stimuli and participate to orchestrate neurons-microglia-astrocytes action to preserve and limit brain damage. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.
Collapse
Affiliation(s)
- Flavia Trettel
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Maria Amalia Di Castro
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy; IRCCS Neuromed, Via Atinense 19, 86077, Pozzilli, Italy
| |
Collapse
|
11
|
Schvartz D, González-Ruiz V, Walter N, Antinori P, Jeanneret F, Tonoli D, Boccard J, Zurich MG, Rudaz S, Monnet-Tschudi F, Sandström J, Sanchez JC. Protein pathway analysis to study development-dependent effects of acute and repeated trimethyltin (TMT) treatments in 3D rat brain cell cultures. Toxicol In Vitro 2019; 60:281-292. [PMID: 31176792 DOI: 10.1016/j.tiv.2019.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/18/2019] [Accepted: 05/29/2019] [Indexed: 11/25/2022]
Abstract
Trimethyltin is an organometallic compound, described to be neurotoxic and to trigger neuroinflammation and oxidative stress. Previous studies associated TMT with the perturbation of mitochondrial function, or neurotransmission. However, the mechanisms of toxicity may differ depending on the duration of exposure and on the stage of maturation of brain cells. This study aim at elucidating whether the toxicity pathways triggered by a known neurotoxicant (TMT) differs depending on cell maturation stage or duration of exposure. To this end omics profiling of immature and differentiated 3D rat brain cell cultures exposed for 24 h or 10 days (10-d) to 0.5 and 1 μM of TMT was performed to better understand the underlying mechanisms of TMT associated toxicity. Proteomics identified 55 and 17 proteins affected by acute TMT treatment in immature and differentiated cultures respectively, while 10-day treatment altered 96 proteins in immature cultures versus 353 in differentiated. The results suggest different sensitivity to TMT depending on treatment duration and cell maturation. In accordance with known TMT mechanisms oxidative stress and neuroinflammation was observed after 10-d treatment at both maturation stages, whereas the neuroinflammatory process was more prominent in differentiated cultures than in the immature, no development-dependent difference could be detected for oxidative stress or synaptic neurodegeneration. Pathway analysis revealed that both vesicular trafficking and the synaptic machinery were strongly affected by 10-d TMT treatment in both maturation stages, as was GABAergic and glutamatergic neurotransmission. This study shows that omics approaches combined with pathway analysis constitutes an improved tool-set in elucidating toxicity mechanisms.
Collapse
Affiliation(s)
- Domitille Schvartz
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Víctor González-Ruiz
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Nadia Walter
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Paola Antinori
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Neuroproteomics group, Department of Clinical Neurosciences, University of Geneva, Geneva, Switzerland
| | - Fabienne Jeanneret
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - David Tonoli
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Julien Boccard
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Marie-Gabrielle Zurich
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Serge Rudaz
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Florianne Monnet-Tschudi
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Jenny Sandström
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Jean-Charles Sanchez
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland.
| |
Collapse
|
12
|
Hou J, Xue J, Wang Z, Li W. Ginsenoside Rg3 and Rh2 protect trimethyltin-induced neurotoxicity via prevention on neuronal apoptosis and neuroinflammation. Phytother Res 2018; 32:2531-2540. [PMID: 30277284 DOI: 10.1002/ptr.6193] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
Abstract
The acute exposure of trimethyltin (TMT) develops clinical syndrome characterized by amnesia, aggressive behavior, and complex seizures. This neurotoxicant selectively induces hippocampal neuronal injury and glial activation accompanied with resultant neuroinflammation. Here we report two candidates ginsenosides Rg3 and Rh2 as neuroprotection agents using a mouse model of TMT intoxication via a single injection (2 mg/kg) and primary neuronal culture systems. Four-week administration of Rg3 or Rh2 significantly reduced TMT-induced seizures and behavioral changes. Rg3 and Rh2 significantly attenuated the oxidative stress evidenced by improvement on antioxidant enzymes and neuronal loss and astrocytic activation in mouse brain. In primary cultures, TMT induced significant neuronal death after 24-h intoxication and vigorous secretion of inflammatory cytokines (IL-1α/β, IL-6, TNF-α, and MCP-1) in astrocytes. Pretreatment with Rg3 or Rh2 not only reduced cell death but efficiently suppressed above mentioned inflammatory cytokines confirmed by antibody array test. The underlying protective mechanism by Rg3 and Rh2 was delineated through selective upregulation of PI3K/Akt and suppression of ERK activation. Intriguingly, Rg3 and Rh2 protected oligodendrocyte progenitor cells (O-2A) from TMT intoxication via promoting type 2 astrocytic differentiation without further inflammatory activation. Collectively, Rg3 and Rh2 interventions aimed at reducing oxidative stress and neuroinflammation neurotoxicity therefore are of therapeutic benefit in TMT-induced neurodegeneration.
Collapse
Affiliation(s)
- Jingang Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,Intelligent Synthetic Biology Center, Daejeon, Republic of Korea
| | - Jianjie Xue
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, China.,Qingdao Institute of Preventive Medicine, Qingdao, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| |
Collapse
|
13
|
Duarte-García A, Romero-Díaz J, Juárez S, Cicero-Casarrubias A, Fragoso-Loyo H, Núñez-Alvarez C, Llorente L, Sánchez-Guerrero J. Disease activity, autoantibodies, and inflammatory molecules in serum and cerebrospinal fluid of patients with Systemic Lupus Erythematosus and Cognitive Dysfunction. PLoS One 2018; 13:e0196487. [PMID: 29723220 PMCID: PMC5933704 DOI: 10.1371/journal.pone.0196487] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/13/2018] [Indexed: 12/01/2022] Open
Abstract
Objective To determine if cognitive dysfunction in patients with systemic lupus erythematosus (SLE) derives from an inflammatory process with continuing disease activity, and increased levels of autoantibodies and inflammatory molecules in serum and cerebrospinal fluid (CSF). Methods 100 randomly selected patients participating in an inception SLE cohort were studied. At entry into the cohort, a standardized medical history and extensive laboratory tests profile, including autoantibodies were completed. Follow-up occurred every 3–6 months with assessment of lupus characteristics, comorbidities, and treatment. After a mean follow-up of six-years, cross-sectional evaluation of cognitive function was done with standardized tests, and in a subset of patients an extended profile of autoantibodies, cytokines and chemokines was measured in serum and CSF. Results At enrollment into the cohort, patients were 26.4±8.2 years of age and lupus duration 5.3±3.7 months. Moderate/severe cognitive dysfunction was diagnosed in 16 patients; in comparison to patients with normal cognitive function, they had lower education 9 vs. 12 years (P = 0.006), higher body mass index 26.7 vs. 24.3 (P = 0.03), positive IgG anticardiolipin antibodies 50% vs 18% (P = 0.009), and a higher median number of concomitant NPSLE syndromes 3 vs. 1, (P = 0.04). The prevalence of cardiovascular-risk factors, other auto-antibodies, lupus activity, treatment, and incidence of critical events did not differ. In serum and CSF, the levels of autoantibodies, cytokines and chemokine were similar, only CCL2 was elevated in CSF [886.1 (374.9–1439.7) vs. 515.8 (3.2–1958.2) pg/mL, P = 0.04]. Conclusion Scant evidence of inflammation in SLE patients with cognitive dysfunction was observed. Only a higher prevalence of IgG anticardiolipin antibodies in serum and increased levels of CCL2 in CSF were detected.
Collapse
Affiliation(s)
- Alí Duarte-García
- Department of Medicine, Division of Rheumatology,Mayo Clinic. Rochester, Minnesota, United States of America
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic. Rochester, Minnesota United States of America
| | - Juanita Romero-Díaz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Sandra Juárez
- Department of Neurology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Alba Cicero-Casarrubias
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Hilda Fragoso-Loyo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Carlos Núñez-Alvarez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Luis Llorente
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Jorge Sánchez-Guerrero
- Department of Medicine, Division of Rheumatology. Mount Sinai Hospital and University Health Network. Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
14
|
Imam SZ, He Z, Cuevas E, Rosas-Hernandez H, Lantz SM, Sarkar S, Raymick J, Robinson B, Hanig JP, Herr D, MacMillan D, Smith A, Liachenko S, Ferguson S, O'Callaghan J, Miller D, Somps C, Pardo ID, Slikker W, B Pierson J, Roberts R, Gong B, Tong W, Aschner M, J Kallman M, Calligaro D, Paule MG. Changes in the metabolome and microRNA levels in biological fluids might represent biomarkers of neurotoxicity: A trimethyltin study. Exp Biol Med (Maywood) 2017; 243:228-236. [PMID: 29105512 DOI: 10.1177/1535370217739859] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neurotoxicity has been linked with exposure to a number of common drugs and chemicals, yet efficient, accurate, and minimally invasive methods to detect it are lacking. Fluid-based biomarkers such as those found in serum, plasma, urine, and cerebrospinal fluid have great potential due to the relative ease of sampling but at present, data on their expression and translation are lacking or inconsistent. In this pilot study using a trimethyl tin rat model of central nervous system toxicity, we have applied state-of-the-art assessment techniques to identify potential individual biomarkers and patterns of biomarkers in serum, plasma, urine or cerebral spinal fluid that may be indicative of nerve cell damage and degeneration. Overall changes in metabolites and microRNAs were observed in biological fluids that were associated with neurotoxic damage induced by trimethyl tin. Behavioral changes and magnetic resonance imaging T2 relaxation and ventricle volume changes served to identify animals that responded to the adverse effects of trimethyl tin. Impact statement These data will help design follow-on studies with other known neurotoxicants to be used to assess the broad applicability of the present findings. Together this approach represents an effort to begin to develop and qualify a set of translational biochemical markers of neurotoxicity that will be readily accessible in humans. Such biomarkers could prove invaluable for drug development research ranging from preclinical studies to clinical trials and may prove to assist with monitoring of the severity and life cycle of brain lesions.
Collapse
Affiliation(s)
- Syed Z Imam
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - Zhen He
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - Elvis Cuevas
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | | | - Susan M Lantz
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - Sumit Sarkar
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - James Raymick
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - Bonnie Robinson
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | | | - David Herr
- 3 US EPA, 96653 NHEERL , Research Triangle Park, North Carolina, NC 27711, USA
| | - Denise MacMillan
- 3 US EPA, 96653 NHEERL , Research Triangle Park, North Carolina, NC 27711, USA
| | - Aaron Smith
- 4 Lilly, Lilly Corporate Center, Indianapolis, Indiana, IN 46285, USA
| | - Serguei Liachenko
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - Sherry Ferguson
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | | | | | | | | | - William Slikker
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | | | - Ruth Roberts
- 8 Department of Biosciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Binsheng Gong
- 9 Division of Bioinformatics, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - Weida Tong
- 9 Division of Bioinformatics, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| | - Michael Aschner
- 10 Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Mary J Kallman
- 11 Kallman Preclinical Consulting, Greenfield, IN 46140, USA
| | - David Calligaro
- 3 US EPA, 96653 NHEERL , Research Triangle Park, North Carolina, NC 27711, USA
| | - Merle G Paule
- 1 Division of Neurotoxicology, US FDA, 4136 NCTR , Jefferson, AR 72079, USA
| |
Collapse
|
15
|
Lee S, Yang M, Kim J, Kang S, Kim J, Kim JC, Jung C, Shin T, Kim SH, Moon C. Trimethyltin-induced hippocampal neurodegeneration: A mechanism-based review. Brain Res Bull 2016; 125:187-99. [PMID: 27450702 DOI: 10.1016/j.brainresbull.2016.07.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
Trimethyltin (TMT), a toxic organotin compound, induces neurodegeneration selectively involving the limbic system and especially prominent in the hippocampus. Neurodegeneration-associated behavioral abnormalities, such as hyperactivity, aggression, cognitive deficits, and epileptic seizures, occur in both exposed humans and experimental animal models. Previously, TMT had been used generally in industry and agriculture, but the use of TMT has been limited because of its dangers to people. TMT has also been used to make a promising in vivo rodent model of neurodegeneration because of its region-specific characteristics. Several studies have demonstrated that TMT-treated animal models of epileptic seizures can be used as tools for researching hippocampus-specific neurotoxicity as well as the molecular mechanisms leading to hippocampal neurodegeneration. This review summarizes the in vivo and in vitro underlying mechanisms of TMT-induced hippocampal neurodegeneration (oxidative stress, inflammatory responses, and neuronal death/survival). Thus, the present review may be helpful to provide general insights into TMT-induced neurodegeneration and approaches to therapeutic interventions for neurodegenerative diseases, including temporal lobe epilepsy.
Collapse
Affiliation(s)
- Sueun Lee
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Miyoung Yang
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Jeonbuk 54538, South Korea
| | - Jinwook Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Sohi Kang
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Juhwan Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Jong-Choon Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, South Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju 63243, South Korea
| | - Sung-Ho Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Changjong Moon
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
16
|
Thibert KA, Raymond GV, Tolar J, Miller WP, Orchard PJ, Lund TC. Cerebral Spinal Fluid levels of Cytokines are elevated in Patients with Metachromatic Leukodystrophy. Sci Rep 2016; 6:24579. [PMID: 27079147 PMCID: PMC4832325 DOI: 10.1038/srep24579] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/30/2016] [Indexed: 01/08/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disease resulting from a deficiency of arylsulfatase A causing an accumulation of cerebroside sulfate, a lipid normally abundant in myelin. Sulfatide accumulation is associated with progressive demyelination and a clinical presentation in severe disease forms that is dominated by motor manifestations. Cerebral inflammation may contribute to the pathophysiology of MLD. To date, cytokine levels in the cerebral spinal fluid of MLD patients have not previously been reported. The objective of this study was to evaluate the concentration of inflammatory cytokines in the CSF of patients with MLD and to compare these levels to unaffected controls. Of 22 cytokines evaluated, we documented significant elevations of MCP-1, IL-1Ra, IL-8, MIP-1b and VEGF in the MLD patients compared to unaffected controls. The elevated cytokines identified in this study may play a significant role in the pathophysiology of MLD. Better understanding of the inflammatory and neurodegenerative process of MLD may lead to improved targeted therapies.
Collapse
Affiliation(s)
- Kathryn A Thibert
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Gerald V Raymond
- Division of Pediatric Neurology, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Jakub Tolar
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Weston P Miller
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Troy C Lund
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| |
Collapse
|
17
|
Kawamura Y, Nakayama A, Kato T, Miura H, Ishihara N, Ihira M, Takahashi Y, Matsuda K, Yoshikawa T. Pathogenic Role of Human Herpesvirus 6B Infection in Mesial Temporal Lobe Epilepsy. J Infect Dis 2015; 212:1014-21. [DOI: 10.1093/infdis/jiv160] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/29/2015] [Indexed: 11/13/2022] Open
|
18
|
O'Callaghan JP, Kelly KA, Locker AR, Miller DB, Lasley SM. Corticosterone primes the neuroinflammatory response to DFP in mice: potential animal model of Gulf War Illness. J Neurochem 2015; 133:708-21. [PMID: 25753028 PMCID: PMC4722811 DOI: 10.1111/jnc.13088] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/24/2015] [Accepted: 03/02/2015] [Indexed: 11/30/2022]
Abstract
Gulf War Illness (GWI) is a multi‐symptom disorder with features characteristic of persistent sickness behavior. Among conditions encountered in the Gulf War (GW) theater were physiological stressors (e.g., heat/cold/physical activity/sleep deprivation), prophylactic treatment with the reversible AChE inhibitor, pyridostigmine bromide (PB), the insect repellent, N,N‐diethyl‐meta‐toluamide (DEET), and potentially the nerve agent, sarin. Prior exposure to the anti‐inflammatory glucocorticoid, corticosterone (CORT), at levels associated with high physiological stress, can paradoxically prime the CNS to produce a robust proinflammatory response to neurotoxicants and systemic inflammation; such neuroinflammatory effects can be associated with sickness behavior. Here, we examined whether CORT primed the CNS to mount neuroinflammatory responses to GW exposures as a potential model of GWI. Male C57BL/6 mice were treated with chronic (14 days) PB/ DEET, subchronic (7–14 days) CORT, and acute exposure (day 15) to diisopropyl fluorophosphate (DFP), a sarin surrogate and irreversible AChE inhibitor. DFP alone caused marked brain‐wide neuroinflammation assessed by qPCR of tumor necrosis factor‐α, IL6, chemokine (C‐C motif) ligand 2, IL‐1β, leukemia inhibitory factor, and oncostatin M. Pre‐treatment with high physiological levels of CORT greatly augmented (up to 300‐fold) the neuroinflammatory responses to DFP. Anti‐inflammatory pre‐treatment with minocycline suppressed many proinflammatory responses to CORT+DFP. Our findings are suggestive of a possible critical, yet unrecognized interaction between the stressor/environment of the GW theater and agent exposure(s) unique to this war. Such exposures may in fact prime the CNS to amplify future neuroinflammatory responses to pathogens, injury, or toxicity. Such occurrences could potentially result in the prolonged episodes of sickness behavior observed in GWI.
Gulf War (GW) veterans were exposed to stressors, prophylactic medicines and, potentially, nerve agents in theater. Subsequent development of GW Illness, a persistent multi‐symptom disorder with features characteristic of sickness behavior, may be caused by priming of the CNS resulting in exaggerated neuroinflammatory responses to pathogens/insults. Nerve agent, diisopropyl fluorophosphate (DFP), produced a neuroinflammatory response that was exacerbated by pre‐treatment with levels of corticosterone simulating heightened stressor conditions. While prophylactic treatments reduced DFP‐induced neuroinflammation, this effect was negated when those treatments were combined with corticosterone.
Collapse
Affiliation(s)
- James P O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | | | | | | | | |
Collapse
|
19
|
Kim J, Yang M, Son Y, Jang H, Kim D, Kim JC, Kim SH, Kang MJ, Im HI, Shin T, Moon C. Glial activation with concurrent up-regulation of inflammatory mediators in trimethyltin-induced neurotoxicity in mice. Acta Histochem 2014; 116:1490-500. [PMID: 25265880 DOI: 10.1016/j.acthis.2014.09.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Trimethyltin (TMT), a potent neurotoxic chemical, causes dysfunction and neuroinflammation in the brain, particularly in the hippocampus. The present study assessed TMT-induced glial cell activation and inflammatory cytokine alterations in the mouse hippocampus, BV-2 microglia, and primary cultured astrocytes. In the mouse hippocampus, TMT treatment significantly increased the expression of glial cell markers, including the microglial marker ionized calcium-binding adapter molecule 1 and the astroglial marker glial fibrillary acidic protein. The expression of M1 and M2 microglial markers (inducible nitric oxide synthase [iNOS] and CD206, respectively) and pro-inflammatory cytokines (interleukin [IL]-1β, IL-6 and tumor necrosis factor [TNF]-α) were significantly increased in the mouse hippocampus following TMT treatment. In BV-2 microglia, iNOS, IL-1β, TNF-α, and IL-6 expression increased significantly, whereas arginase-1 and CD206 expression decreased significantly after TMT treatment in a time- and concentration-dependent manner. In primary cultured astrocytes, iNOS, arginase-1, IL-1β, TNF-α, and IL-6 expression increased significantly, whereas IL-10 expression decreased significantly after TMT treatment in a time- and concentration-dependent manner. These results indicate that significant up-regulation of pro-inflammatory signals in TMT-induced neurotoxicity may be associated with pathological processing of TMT-induced neurodegeneration.
Collapse
|
20
|
Ceccariglia S, D’altocolle A, Del Fa’ A, Silvestrini A, Barba M, Pizzolante F, Repele A, Michetti F, Gangitano C. Increased expression of Aquaporin 4 in the rat hippocampus and cortex during trimethyltin-induced neurodegeneration. Neuroscience 2014; 274:273-88. [DOI: 10.1016/j.neuroscience.2014.05.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 12/12/2022]
|
21
|
Early activation of STAT3 regulates reactive astrogliosis induced by diverse forms of neurotoxicity. PLoS One 2014; 9:e102003. [PMID: 25025494 PMCID: PMC4098997 DOI: 10.1371/journal.pone.0102003] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/13/2014] [Indexed: 12/31/2022] Open
Abstract
Astrogliosis, a cellular response characterized by astrocytic hypertrophy and accumulation of GFAP, is a hallmark of all types of central nervous system (CNS) injuries. Potential signaling mechanisms driving the conversion of astrocytes into “reactive” phenotypes differ with respect to the injury models employed and can be complicated by factors such as disruption of the blood-brain barrier (BBB). As denervation tools, neurotoxicants have the advantage of selective targeting of brain regions and cell types, often with sparing of the BBB. Previously, we found that neuroinflammation and activation of the JAK2-STAT3 pathway in astrocytes precedes up regulation of GFAP in the MPTP mouse model of dopaminergic neurotoxicity. Here we show that multiple mechanistically distinct mouse models of neurotoxicity (MPTP, AMP, METH, MDA, MDMA, KA, TMT) engender the same neuroinflammatory and STAT3 activation responses in specific regions of the brain targeted by each neurotoxicant. The STAT3 effects seen for TMT in the mouse could be generalized to the rat, demonstrating cross-species validity for STAT3 activation. Pharmacological antagonists of the neurotoxic effects blocked neuroinflammatory responses, pSTAT3tyr705 and GFAP induction, indicating that damage to neuronal targets instigated astrogliosis. Selective deletion of STAT3 from astrocytes in STAT3 conditional knockout mice markedly attenuated MPTP-induced astrogliosis. Monitoring STAT3 translocation in GFAP-positive cells indicated that effects of MPTP, METH and KA on pSTAT3tyr705 were localized to astrocytes. These findings strongly implicate the STAT3 pathway in astrocytes as a broadly triggered signaling pathway for astrogliosis. We also observed, however, that the acute neuroinflammatory response to the known inflammogen, LPS, can activate STAT3 in CNS tissue without inducing classical signs of astrogliosis. Thus, acute phase neuroinflammatory responses and neurotoxicity-induced astrogliosis both signal through STAT3 but appear to do so through different modules, perhaps localized to different cell types.
Collapse
|
22
|
Sawyer AJ, Tian W, Saucier-Sawyer JK, Rizk PJ, Saltzman WM, Bellamkonda RV, Kyriakides TR. The effect of inflammatory cell-derived MCP-1 loss on neuronal survival during chronic neuroinflammation. Biomaterials 2014; 35:6698-706. [PMID: 24881026 DOI: 10.1016/j.biomaterials.2014.05.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 05/01/2014] [Indexed: 01/22/2023]
Abstract
Intracranial implants elicit neurodegeneration via the foreign body response (FBR) that includes BBB leakage, macrophage/microglia accumulation, and reactive astrogliosis, in addition to neuronal degradation that limit their useful lifespan. Previously, monocyte chemoattractant protein 1 (MCP-1, also CCL2), which plays an important role in monocyte recruitment and propagation of inflammation, was shown to be critical for various aspects of the FBR in a tissue-specific manner. However, participation of MCP-1 in the brain FBR has not been evaluated. Here we examined the FBR to intracortical silicon implants in MCP-1 KO mice at 1, 2, and 8 weeks after implantation. MCP-1 KO mice had a diminished FBR compared to WT mice, characterized by reductions in BBB leakage, macrophage/microglia accumulation, and astrogliosis, and an increased neuronal density. Moreover, pharmacological inhibition of MCP-1 in implant-bearing WT mice maintained the increased neuronal density. To elucidate the relative contribution of microglia and macrophages, bone marrow chimeras were generated between MCP-1 KO and WT mice. Increased neuronal density was observed only in MCP-1 knockout mice transplanted with MCP-1 knockout marrow, which indicates that resident cells in the brain are major contributors. We hypothesized that these improvements are the result of a phenotypic switch of the macrophages/microglia polarization state, which we confirmed using PCR for common activation markers. Our observations suggest that MCP-1 influences neuronal loss, which is integral to the progression of neurological disorders like Alzheimer's and Parkinson disease, via BBB leakage and macrophage polarization.
Collapse
Affiliation(s)
- Andrew J Sawyer
- Department of Pathology, Yale School of Medicine, 310 Cedar Street LH 108, New Haven, CT 06520-8023, USA
| | - Weiming Tian
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, PR China
| | | | - Paul J Rizk
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Themis R Kyriakides
- Department of Pathology, Yale School of Medicine, 310 Cedar Street LH 108, New Haven, CT 06520-8023, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
23
|
Cho H, Sajja V, VandeVord P, Lee Y. Blast induces oxidative stress, inflammation, neuronal loss and subsequent short-term memory impairment in rats. Neuroscience 2013; 253:9-20. [DOI: 10.1016/j.neuroscience.2013.08.037] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 08/06/2013] [Accepted: 08/21/2013] [Indexed: 12/17/2022]
|
24
|
McPherson CA, Merrick BA, Harry GJ. In vivo molecular markers for pro-inflammatory cytokine M1 stage and resident microglia in trimethyltin-induced hippocampal injury. Neurotox Res 2013; 25:45-56. [PMID: 24002884 DOI: 10.1007/s12640-013-9422-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 08/13/2013] [Accepted: 08/20/2013] [Indexed: 12/18/2022]
Abstract
Microglia polarization to the classical M1 activation state is characterized by elevated pro-inflammatory cytokines; however, a full profile has not been generated in the early stages of a sterile inflammatory response recruiting only resident microglia. We characterized the initial M1 state in a hippocampal injury model dependent upon tumor necrosis factor (TNF) receptor signaling for dentate granule cell death. Twenty-one-day-old CD1 male mice were injected with trimethyltin (TMT 2.3 mg/kg, i.p.) and the hippocampus was examined at an early stage (24-h post-dosing) of neuronal death. Glia activation was assessed using a custom quantitative nuclease protection assay. We report elevated mRNA levels for glia response such as ionizing calcium-binding adapter molecule-1 and glial fibrillary acidic protein (Gfap); Fas, hypoxia inducible factor alpha, complement component 1qb, TNF-related genes (Tnf, Tnfaip3, Tnfrsfla); interleukin-1 alpha, Cd44, chemokine (C-C motif) ligand (Ccl)2, Cc14, integrin alpha M, lipocalin (Lcn2), and secreted phosphoprotein 1 (Spp1). These changes occurred in the absence of changes in matrix metalloproteinase 9 and 12, neural cell adhesion molecule, metabotropic glutamate receptor (Grm)3, and Ly6/neurotoxin 1 (Lynx1), as well as, a decrease in neurotrophin 3, glutamate receptor subunit epsilon (Grin)-2b, and neurotrophic tyrosine kinase receptor, type 3. The M2 anti-inflammatory marker, transforming growth factor beta-1 (Tgfb1) was elevated. mRNAs associated with early stage of injury-induced neurogenesis including fibroblast growth factor 21 and Mki67 were elevated. In the "non-injured" temporal cortex receiving projections from the hippocampus, Lynx1, Grm3, and Grin2b were decreased and Gfap increased. Formalin fixed-paraffin-embedded tissue did not generate a comparable profile.
Collapse
Affiliation(s)
- C A McPherson
- Neurotoxicology Group, Division of National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, MD E1-07, Research Triangle Park, NC, 27709, USA
| | | | | |
Collapse
|
25
|
Gene expression profiling as a tool to investigate the molecular machinery activated during hippocampal neurodegeneration induced by trimethyltin (TMT) administration. Int J Mol Sci 2013; 14:16817-35. [PMID: 23955266 PMCID: PMC3759937 DOI: 10.3390/ijms140816817] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 08/06/2013] [Accepted: 08/08/2013] [Indexed: 12/31/2022] Open
Abstract
Trimethyltin (TMT) is an organotin compound exhibiting neurotoxicant effects selectively localized in the limbic system and especially marked in the hippocampus, in both experimental animal models and accidentally exposed humans. TMT administration causes selective neuronal death involving either the granular neurons of the dentate gyrus or the pyramidal cells of the Cornu Ammonis, with a different pattern of localization depending on the different species studied or the dosage schedule. TMT is broadly used to realize experimental models of hippocampal neurodegeneration associated with cognitive impairment and temporal lobe epilepsy, though the molecular mechanisms underlying the associated selective neuronal death are still not conclusively clarified. Experimental evidence indicates that TMT-induced neurodegeneration is a complex event involving different pathogenetic mechanisms, probably acting differently in animal and cell models, which include neuroinflammation, intracellular calcium overload, and oxidative stress. Microarray-based, genome-wide expression analysis has been used to investigate the molecular scenario occurring in the TMT-injured brain in different in vivo and in vitro models, producing an overwhelming amount of data. The aim of this review is to discuss and rationalize the state-of-the-art on TMT-associated genome wide expression profiles in order to identify comparable and reproducible data that may allow focusing on significantly involved pathways.
Collapse
|
26
|
Neuroprotective strategies in hippocampal neurodegeneration induced by the neurotoxicant trimethyltin. Neurochem Res 2012. [PMID: 23179590 DOI: 10.1007/s11064-012-0932-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The selective vulnerability of specific neuronal subpopulations to trimethyltin (TMT), an organotin compound with neurotoxicant effects selectively involving the limbic system and especially marked in the hippocampus, makes it useful to obtain in vivo models of neurodegeneration associated with behavioural alterations, such as hyperactivity and aggression, cognitive impairment as well as temporal lobe epilepsy. TMT has been widely used to study neuronal and glial factors involved in selective neuronal death, as well as the molecular mechanisms leading to hippocampal neurodegeneration (including neuroinflammation, excitotoxicity, intracellular calcium overload, mitochondrial dysfunction and oxidative stress). It also offers a valuable instrument to study the cell-cell interactions and signalling pathways that modulate injury-induced neurogenesis, including the involvement of newly generated neurons in the possible repair processes. Since TMT appears to be a useful tool to damage the brain and study the various responses to damage, this review summarises current data from in vivo and in vitro studies on neuroprotective strategies to counteract TMT-induced neuronal death, that may be useful to elucidate the role of putative candidates for translational medical research on neurodegenerative diseases.
Collapse
|
27
|
Kaufmann W, Bolon B, Bradley A, Butt M, Czasch S, Garman RH, George C, Gröters S, Krinke G, Little P, McKay J, Narama I, Rao D, Shibutani M, Sills R. Proliferative and nonproliferative lesions of the rat and mouse central and peripheral nervous systems. Toxicol Pathol 2012; 40:87S-157S. [PMID: 22637737 DOI: 10.1177/0192623312439125] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Harmonization of diagnostic nomenclature used in the pathology analysis of tissues from rodent toxicity studies will enhance the comparability and consistency of data sets from different laboratories worldwide. The INHAND Project (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) is a joint initiative of four major societies of toxicologic pathology to develop a globally recognized nomenclature for proliferative and nonproliferative lesions in rodents. This article recommends standardized terms for classifying changes observed in tissues of the mouse and rat central (CNS) and peripheral (PNS) nervous systems. Sources of material include academic, government, and industrial histopathology databases from around the world. Covered lesions include frequent, spontaneous, and aging-related changes as well as principal toxicant-induced findings. Common artifacts that might be confused with genuine lesions are also illustrated. The neural nomenclature presented in this document is also available electronically on the Internet at the goRENI website (http://www.goreni.org/).
Collapse
|
28
|
Ehrlich D, Pirchl M, Humpel C. Effects of long-term moderate ethanol and cholesterol on cognition, cholinergic neurons, inflammation, and vascular impairment in rats. Neuroscience 2012; 205:154-66. [PMID: 22244974 PMCID: PMC3314917 DOI: 10.1016/j.neuroscience.2011.12.054] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 12/23/2011] [Accepted: 12/28/2011] [Indexed: 12/20/2022]
Abstract
There is strong evidence that vascular risk factors play a role in the development of Alzheimer's disease (AD) or vascular dementia (vaD). Ethanol (EtOH) and cholesterol are such vascular risk factors, and we recently showed that hypercholesterolemia causes pathologies similar to AD [Ullrich et al. (2010) Mol Cell Neurosci 45, 408–417]. The aim of this study was to investigate the effects of long-term (12 months) EtOH treatment (20% v/v in drinking water) alone or long-term 5% cholesterol diet alone or a combination (mix) in adult Sprague–Dawley rats. Long-term EtOH treatment (plasma EtOH levels 58±23 mg/dl) caused significant impairment of spatial memory, reduced the number of choline acetyltransferase- and p75 neurotrophin receptor-positive nucleus basalis of Meynert neurons, decreased cortical acetylcholine, elevated cortical monocyte chemoattractant protein-1 and tissue-type plasminogen activator, enhanced microglia, and markedly induced anti-rat immunoglobulin G-positive blood–brain barrier leakage. The effect of long-term hypercholesterolemia was similar. Combined long-term treatment of rats with 20% EtOH and 5% cholesterol (mix) did not potentiate treatment with EtOH alone, but instead counteracted some of the EtOH-associated effects. In conclusion, our data show that vascular risk factors EtOH and cholesterol play a role in cognitive impairment and possibly vaD.
Collapse
Affiliation(s)
- D Ehrlich
- Laboratory for Psychiatry and Exp. Alzheimer's Research, Department of Psychiatry and Psychotherapy, Anichstr. 35, 6020 Innsbruck, Austria
| | | | | |
Collapse
|
29
|
Little AR, Miller DB, Li S, Kashon ML, O'Callaghan JP. Trimethyltin-induced neurotoxicity: gene expression pathway analysis, q-RT-PCR and immunoblotting reveal early effects associated with hippocampal damage and gliosis. Neurotoxicol Teratol 2011; 34:72-82. [PMID: 22108043 DOI: 10.1016/j.ntt.2011.09.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 08/19/2011] [Accepted: 09/14/2011] [Indexed: 12/17/2022]
Abstract
Damage to the CNS results in a complex series of molecular and cellular changes involving the affected targets and the ensuing glial reaction. The initial gene expression events that underlie these cellular responses may serve as early biomarkers of neurotoxicity. Here, we examined gene expression profiles during the initial phase of hippocampal damage resulting from systemic exposure of rats to the organometallic neurotoxicant, trimethyltin (TMT, 8.0 mg/kg, i.p.). Using TMT as a neurodegeneration tool confers several advantages for evaluating molecular events associated with neural damage: 1) regional and cellular targets and time course of damage are known, 2) the blood-brain barrier is not compromised, which limits the contribution of blood-borne factors, e.g. immune, to neural injury responses and 3) known protein and mRNA signatures of TMT-induced neurotoxicity can be used as positive controls to validate novel expression events associated with exposure to this neurotoxicant. Using Affymetrix Gene Chip® to assess gene expression after TMT, combined with Ingenuity Pathway Analysis®, we observed changes consistent for genes known to be affected in hippocampus, while corresponding changes were not detected in cerebellum, a non-target region. In agreement with previous observations, limited changes in expression of inflammation-related genes were observed. Correlated expression profiles were found after exposure to TMT, including changes in gene ontologies associated with neurological disease, cellular assembly and maintenance, as well as signaling pathways associated with cellular stress, energy metabolism and glial activation. Selected gene changes were confirmed from each category by q-RT-PCR and immunoblot analysis. The canonical relationships identified implicate molecular pathways and processes relevant to detection of early stages of hippocampal damage in the TMT model. These observations provide new insight into early events associated with neuronal degeneration and associated glial activation that may serve as the basis for discovery and development of biomarkers of neurotoxicity.
Collapse
Affiliation(s)
- A R Little
- Molecular Neurotoxicology Laboratory, Health Effects Research Laboratory, Centers for Disease Control and Prevention-NIOSH, 1095 Willowdale Road, Morgantown, WV 26505, USA.
| | | | | | | | | |
Collapse
|
30
|
Geloso MC, Corvino V, Michetti F. Trimethyltin-induced hippocampal degeneration as a tool to investigate neurodegenerative processes. Neurochem Int 2011; 58:729-38. [DOI: 10.1016/j.neuint.2011.03.009] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/02/2011] [Accepted: 03/08/2011] [Indexed: 12/29/2022]
|
31
|
Nelson TE, Hao C, Manos J, Ransohoff R, Gruol DL. Altered hippocampal synaptic transmission in transgenic mice with astrocyte-targeted enhanced CCL2 expression. Brain Behav Immun 2011; 25 Suppl 1:S106-19. [PMID: 21356306 PMCID: PMC4467826 DOI: 10.1016/j.bbi.2011.02.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 02/18/2011] [Accepted: 02/18/2011] [Indexed: 01/17/2023] Open
Abstract
Elevated expression of neuroinflammatory factors in the central nervous system (CNS) contributes to the cognitive impairment in CNS disorders such as injury, disease and neurodegenerative disorders. However, information on the role of specific neuroimmune factors in normal and abnormal CNS function is limited. In this study, we investigated the effects of chronic exposure to the chemokine CCL2 on hippocampal synaptic function at the Schaffer collateral-CA1 synapse, a synapse that is known to play an important role in cognitive functions such as memory and learning. Synaptic function was measured in vitro using hippocampal slices obtained from transgenic mice that express elevated levels of CCL2 in the CNS through astrocyte expression and their non-transgenic littermate controls. Extracellular field potential electrophysiological recordings showed a significant reduction in the magnitude of synaptic responses in hippocampal slices from the CCL2 transgenic mice compared with slices from non-transgenic littermate controls. Two forms of short-term synaptic plasticity (post-tetanic potentiation and short-term potentiation) thought to be important cellular mechanisms of short-term memory were enhanced in hippocampal slices from CCL2 transgenic mice compared to non-transgenic hippocampal slices, whereas long-term synaptic plasticity (LTP), which is critical to long-term memory formation, was not altered. Western blot analysis of hippocampus from the CCL2 transgenic mice and non-transgenic mice showed no change in level of neuronal specific enolase, a neuronal specific protein, GFAP, an astrocyte specific protein, and several synaptic proteins compared with non-transgenic littermate controls. These results show that CCL2, which is known to be chronically produced at elevated levels within the CNS in a number of CNS disorders, can significantly alter hippocampal function and implicate a role for CCL2 in the cognitive dysfunction associated with these CNS disorders.
Collapse
Affiliation(s)
- Thomas E. Nelson
- Molecular and Integrative Neurosciences Dept., The Scripps Research Institute, La Jolla, CA, USA
| | - Christine Hao
- Molecular and Integrative Neurosciences Dept., The Scripps Research Institute, La Jolla, CA, USA
| | - Jessica Manos
- Molecular and Integrative Neurosciences Dept., The Scripps Research Institute, La Jolla, CA, USA
| | - R.M. Ransohoff
- Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Donna L. Gruol
- Molecular and Integrative Neurosciences Dept., The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
32
|
Kozuki M, Kurata T, Miyazaki K, Morimoto N, Ohta Y, Ikeda Y, Abe K. Atorvastatin and pitavastatin protect cerebellar Purkinje cells in AD model mice and preserve the cytokines MCP-1 and TNF-α. Brain Res 2011; 1388:32-8. [DOI: 10.1016/j.brainres.2011.03.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 03/04/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
|
33
|
Choi J, Lee HW, Suk K. Increased plasma levels of lipocalin 2 in mild cognitive impairment. J Neurol Sci 2011; 305:28-33. [PMID: 21463871 DOI: 10.1016/j.jns.2011.03.023] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 02/24/2011] [Accepted: 03/10/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by an irreversible cognitive decline and neuronal loss associated with neurofibrillary tangles and senile plaques. Mild cognitive impairment (MCI) is a prodromal stage of AD and is associated with memory loss and a high risk of developing AD. Lipocalin 2 (LCN2) is an acute phase protein. Our previous studies have shown that exposure to inflammatory stimuli resulted in elevated LCN2 levels in brain microglia and astrocytes implicating LCN2 in brain inflammation. Therefore, we hypothesize that there may be a significant change in the plasma LCN2 levels in patients with MCI and AD when compared to healthy control subjects. METHODS Forty-one patients with MCI, 62 patients with AD and 38 healthy elderly control subjects were recruited for this study. They were given a comprehensive battery of neuropsychological tests including a mini-mental status examination (MMSE) and clinical dementia rating (CDR). A variety of clinical information was collected from the semi-structured questionnaire administered. The LCN2 levels were measured using a specific enzyme-linked immunosorbent assay in the plasma, which had been collected early in the morning after overnight fasting. RESULTS The LCN2 levels were significantly higher in MCI patients compared to the healthy control subjects and AD patients [control vs. MCI (p=0.005); MCI vs. AD (p=0.009)]. There was a significant negative correlation between the LCN2 levels and CDR scores (r=-0.245, p=0.014), and there was a positive correlation between the LCN2 levels and MMSE scores (r=0.317, p=0.001) among all of the MCI and AD patients. CONCLUSION MCI represents a prodromal stage of AD, and inflammation occurs as one of the earliest pathological events in AD. Thus, increased plasma LCN2 levels during MCI could be helpful in predicting the progression from MCI to AD.
Collapse
Affiliation(s)
- Jihye Choi
- Department of Pharmacology, Brain Science and Engineering Institute, CMRI, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | | | | |
Collapse
|
34
|
Andjus PR, Bataveljić D, Vanhoutte G, Mitrecic D, Pizzolante F, Djogo N, Nicaise C, Gankam Kengne F, Gangitano C, Michetti F, van der Linden A, Pochet R, Bacić G. In vivo morphological changes in animal models of amyotrophic lateral sclerosis and Alzheimer's-like disease: MRI approach. Anat Rec (Hoboken) 2010; 292:1882-92. [PMID: 19943341 DOI: 10.1002/ar.20995] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Magnetic resonance imaging (MRI) is the only noninvasive technique that provides structural information on both cell loss and metabolic changes. After reviewing all the results obtained in clinical studies, reliable biomarkers in neurological diseases are still lacking. Diffusional MRI, MR spectroscopy, and the assessment of regional atrophy are promising approaches, but they cannot be simultaneously used on a single patient. Thus, for further research progress, reliable animal models are needed. To this aim, we have used the clinical MRI to assess neurodegenerative processes in the hSOD-1(G93A) ALS rat model and in the trimethyltin (TMT)-treated model of Alzheimer's-like disease. T2-weighted (T2W) hyperintensive neurodegenerative foci were found in the brainstem of the ALS rat with apparent lateral ventricle dilation (T1W-hypointensity vs. T2W-hyperintensity). Degenerative processes in these areas were also confirmed by confocal images of GFAP-positive astrogliosis. MRI after i.v.i. of magnetic anti-CD4 antibodies indicated an accumulation of inflammatory cells near dilated ventricles. TMT-treated rats also revealed the dilation of lateral ventricles. Expected deterioration in the hippocampus was not observed by clinical MRI, but immunocytochemistry could reveal significant redistribution of macro- and microglia in this structure. In both models, Gd-DTPA contrast revealed a compromised blood brain barrier that may serve as the passage for inflammatory immune cells in the vicinity of dilated lateral ventricles. Moreover, in both models the midbrain region of the dorsal hippocampus was the target of BBB compromise, thus revealing a potentially vulnerable point that can be the primary target of neurodegeneration in the central nervous system.
Collapse
Affiliation(s)
- Pavle R Andjus
- Department of Physiology and Biochemistry, School of Biology, University of Belgrade, Studentski trg 12, Belgrade, Serbia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kraft AD, McPherson CA, Harry GJ. Heterogeneity of microglia and TNF signaling as determinants for neuronal death or survival. Neurotoxicology 2009; 30:785-93. [PMID: 19596372 PMCID: PMC3329780 DOI: 10.1016/j.neuro.2009.07.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 07/01/2009] [Indexed: 12/30/2022]
Abstract
Microglia do not constitute a single, uniform cell population, but rather comprise cells with varied phenotypes, some which are beneficial and others that may require active regulatory control. Thus, gaining a better understanding of the heterogeneity of resident microglia responses will contribute to any interpretation regarding the impact of any such response in the brain. Microglia are the primary source of the pro-inflammatory cytokine, tumor necrosis factor (TNF) that can initiate various effects through the activation of membrane receptors. The TNF p55 receptor contains a death domain and activation normally leads to cellular apoptosis; however, under specific conditions, receptor activation can also lead to the activation of NF-kappaB and contribute to cell survival. These divergent outcomes have been linked to receptor localization with receptor internalization leading to cell death and membrane localization supporting cell survival. A second TNF receptor, TNF p75 receptor, is normally linked to cell growth and survival, however, it can cooperate with the p55 receptor and contribute to cell death. Thus, while an elevation in TNFalpha in the brain is often considered an indicator of microglia activation and neuroinflammation, a number of factors come into play to determine the final outcome. Data are reviewed demonstrating that heterogeneity in morphological response of microglia and the expression of TNFalpha and TNF receptors are critical in identifying and characterizing neurotoxic events as they relate to neuroinflammation, neuronal damage and in stimulating neuroprotection.
Collapse
Affiliation(s)
- Andrew D. Kraft
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| | - Christopher A McPherson
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| | - G. Jean Harry
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| |
Collapse
|
36
|
The multifaceted profile of activated microglia. Mol Neurobiol 2009; 40:139-56. [PMID: 19629762 DOI: 10.1007/s12035-009-8077-9] [Citation(s) in RCA: 250] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 06/17/2009] [Indexed: 12/17/2022]
Abstract
Although relatively neglected previously, research efforts in the past decade or so have identified a pivotal role for glial cells in regulating neuronal function. Particular emphasis has been placed on increasing our understanding of the function of microglia because a change from the ramified "resting" state of these cells has been associated with the pathogenesis of several neurodegenerative diseases, notably Alzheimer's disease. However, it is not clear whether activation of microglia and the associated inflammatory changes play a part in triggering disease processes or whether cell activation is a response to the early changes associated with the disease. In either case, the possibility exists that modulation of microglial activation may be beneficial in some circumstances, underlying the need to pursue research in this area. The original morphological categorization of microglia by Del Rio Hortega into ameboid, ramified, and intermediate forms, must now be elaborated to encompass a functional description. The evidence which has been generated recently suggests that microglia are probably never in a "resting" state and that several intermediate transitional states, based on function and morphology, probably exist. A more complete understanding of these states and the triggers which lead to a change from one to another state, and the factors which modulate the molecular switch that determines the persistence of the "activated" state remain to be identified.
Collapse
|
37
|
Kurkowska-Jastrzebska I, Zaremba M, Członkowska A, Oderfeld-Nowak B. Down-regulation of microglia and NG2-positive cells reaction in trimethyltin-injured hippocampus of rats treated with myelin basic protein-reactive T cells: possible contribution to the neuroprotective effect of T cells. J Neurosci Res 2009; 88:24-32. [PMID: 19610113 DOI: 10.1002/jnr.22187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In our previous investigations, we demonstrated that CD4(+) antimyelin basic protein (MBP) T cells protect hippocampal neurons against trimethyltin-induced damage. We hypothesized involvement of T cells, interacting with the various glial populations activated during the neurodegeneration process. In this study, we employ immunocytochemical methods to investigate the influence of administration of T cells on the response of microglia and of NG2(+) cells to trimethyltin (TMT)-induced damage. Female Lewis rats were treated with anti-MBP CD4(+) T cells (4 million per animal, i.v) 24 hr after TMT (8 mg/kg, i.p) intoxication. TMT caused degeneration of CA4 hipppocampal neurons and evoked an abundant reaction of microglial and NG2(+) cells in the injured region. The cells changed morphology into the activated state, and the number of OX42(+) and NG2(+) cells increased about 4.5-fold and 3-fold, respectively, relative to controls as assessed on day 21 after TMT treatment. Additionally, the cells of ameboid morphology, which expressed NG2 or microglial antigens, appeared in the zone of neurodegeneration. Furthermore, certain cells of ameboid phenotype shared both antigens. In rats treated with T cells, down-regulation of the activation of both glial classes and reduction of formation of their ameboid forms was observed. The number of the total OX42(+) and NG2(+) cells decreased by 21% and 54%, respectively, and the number of their ameboid forms decreased by 46% and 73%, respectively. Our data suggest that the diminished activation of microglia and NG2(+) cells, particularly the reduced number of their ameboid forms, may contribute to the neuroprotective effect of T cells.
Collapse
|
38
|
Abstract
Neuroinflammation is a hot topic in contemporary neuroscience. A relatively new open-access journal, the Journal of Neuroinflammation, focuses on this field. As another example, abstracts to the 2007 Annual Meeting of the Society for Neuroscience could be submitted in several subcategories of neuroinflammation, a strong signal of growth in this research area. While it is becoming clear that activation of microglia and astroglia and the attendant expression of proinflammatory cytokines and chemokines often are associated with disease-, trauma-, and toxicant-induced damage to the CNS, it is by no means clear that a cause-and-effect relationship exists between the presence of a neuroinflammatory process and neural damage. We have explored this issue with two models of dopaminergic neurotoxicity. We used a single low-dose regimen of MPTP or METH, a paradigm that causes selective degeneration of striatal dopaminergic nerve terminals without affecting the cell body in the substantia nigra. Both compounds increased the expression of the microglia-associated factors, Il-1alpha, Il6, Ccl2, and Tnf-alpha, and also elicited morphologic evidence of microglial activation prior to induction of astrogliosis. Pharmacologic antagonism of MPTP and METH neurotoxicity prevented these proinflammatory responses, findings suggestive of a link between neuroinflammation and the observed neurotoxic outcomes. Nevertheless, when we used minocycline to suppress the expression of all these mediators, with the exception of Tnf-alpha, we failed to see neuroprotection. Likewise, when we examined the effects of MPTP or METH in transgenic mice lacking Il6, Ccl2, or Tnfr1/2 genes, deficiency of either Il6 or Ccl2 did not alter neurotoxicity, whereas deficiency in Tnfr1/2 was neuroprotective. Although these observations pointed to a role of the proinflammatory cytokine, TNF-alpha, in the neurotoxic effects of MPTP and METH, other observations did not support this supposition. For example, activation of NF-kappaB or induction of iNOS, known components of inflammatory responses and free radical formation, were not observed. Moreover, immunosuppressive regimens of glucocorticoids failed to suppress TNF-alpha or attenuate neurotoxicity. Taken together, our observations suggest that MPTP and METH neurotoxicity are associated with the elaboration of a "neuroinflammatory" response, yet this response lacks key features of inflammation and, with the exception of TNF-alpha, neurotoxicity appears to be the cause rather than the consequence of proinflammatory signals.
Collapse
Affiliation(s)
- James P O'Callaghan
- Centers for Disease Control and Prevention-NIOSH Morgantown, West Virginia, USA.
| | | | | |
Collapse
|
39
|
Harry GJ, Kraft AD. Neuroinflammation and microglia: considerations and approaches for neurotoxicity assessment. Expert Opin Drug Metab Toxicol 2008; 4:1265-77. [PMID: 18798697 PMCID: PMC2658618 DOI: 10.1517/17425255.4.10.1265] [Citation(s) in RCA: 197] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The impact of an inflammatory response, as well as interactions between the immune and nervous systems, are rapidly assuming major roles in neurodegenerative disease and injury. However, it is now appreciated that the exact nature of such responses can differ with each type of insult and interaction. More recently, neuroinflammation and the associated cellular response of microglia are being considered for their contribution to neurotoxicity of environmental agents; yet, so far, the inclusion of inflammatory end points into neurotoxicity assessment have relied primarily on relatively limited measures or driven by in vitro models of neurotoxicity. OBJECTIVE To present background information on relevant biological considerations of neuroinflammation and the microglia response demonstrating the complex integrative nature of these biological processes and raising concern with regards to translation of effects demonstrated in vitro to the in vivo situation. Specific points are addressed that would influence the design and interpretation of neuroinflammation with regards to neurotoxicology assessment. CONCLUSION There is a complex and dynamic response in the brain to regulate inflammatory processes and maintain a normal homeostatic level. The classification of such responses as beneficial or detrimental is an oversimplification. Neuroinflammation should be considered as a balanced network of processes in which subtle modifications can shift the cells toward disparate outcomes. The tendency to overinterpret data obtained in an isolated culture system should be discouraged. Rather, the use of cross-disciplinary approaches to evaluate several end points should be incorporated into the assessment of inflammatory contributions to the neurotoxicity of environmental exposures.
Collapse
Affiliation(s)
- Gaylia Jean Harry
- National Institute of Environmental Health Sciences, National Institutes of Health, Neurotoxicology Group, Laboratory of Neurobiology, Department of Health and Human Services, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
40
|
Upregulation of Semaphorin 3A and the associated biochemical and cellular events in a rat model of retinal detachment. Graefes Arch Clin Exp Ophthalmol 2008; 247:73-86. [DOI: 10.1007/s00417-008-0945-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 07/09/2008] [Accepted: 08/29/2008] [Indexed: 12/18/2022] Open
|
41
|
Cho J, Gruol DL. The chemokine CCL2 activates p38 mitogen-activated protein kinase pathway in cultured rat hippocampal cells. J Neuroimmunol 2008; 199:94-103. [PMID: 18584881 PMCID: PMC2583399 DOI: 10.1016/j.jneuroim.2008.05.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 04/25/2008] [Accepted: 05/19/2008] [Indexed: 11/24/2022]
Abstract
Emerging evidence indicates that chemokines can regulate both the physiology and biochemistry of CNS neurons and glia. In the current study, Western blot analysis showed that in rat hippocampal neuronal/glial cultures the signal transduction pathway activated by CCL2, a chemokine expressed in the normal brain and at elevated levels during neuroinflammation, involves a G-protein coupled receptor, p38 MAPK as well as its immediate upstream kinase MKK3/6, and the downstream transcription factor CREB. ERK 1/2 and the transcription factors STAT1 and STAT3 do not play a prominent role. CCL2 also altered Ca(2+) influx and synaptic network activity in the hippocampal neurons. These results suggest an important role for p38 MAPK and CREB in hippocampal actions of CCL2.
Collapse
Affiliation(s)
- Jungsook Cho
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, United States.
| | | |
Collapse
|
42
|
He J, Crews FT. Increased MCP-1 and microglia in various regions of the human alcoholic brain. Exp Neurol 2008; 210:349-58. [PMID: 18190912 PMCID: PMC2346541 DOI: 10.1016/j.expneurol.2007.11.017] [Citation(s) in RCA: 405] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 11/02/2007] [Accepted: 11/13/2007] [Indexed: 01/12/2023]
Abstract
Cytokines and microglia have been implicated in anxiety, depression, neurodegeneration as well as the regulation of alcohol drinking and other consumatory behaviors, all of which are associated with alcoholism. Studies using animal models of alcoholism suggest that microglia and proinflammatory cytokines contribute to alcoholic pathologies [Crews, F.T., Bechara, R., Brown, L.A., Guidot, D.M., Mandrekar, P., Oak, S., Qin, L., Szabo, G., Wheeler, M., Zou, J., (2006) Cytokines and alcohol. Alcohol., Clin. Exp. Res. 30:720-730]. In the current study, human postmortem brains from moderate drinking controls and alcoholics obtained from the New South Wales Tissue Resource Center were used to study the cytokine, monocyte chemoattractant protein 1 (MCP-1,CCL2) and microglia markers in various brain regions. Since MCP-1 is a key proinflammatory cytokine induced by chronic alcohol treatment of mice, and known to regulate drinking behavior in mice, MCP-1 protein levels from human brain homogenate were measured using ELISA, and indicated increased MCP-1 concentration in ventral tegmental area (VTA), substantia nigra (SN), hippocampus and amygdala of alcoholic brains as compared with controls. Immunohistochemistry was further performed to visualize human microglia using ionized calcium binding adaptor protein-1 (Iba-1), and Glucose transporter-5 (GluT5). Alcoholics were found to have brain region-specific increases in microglial markers. In cingulate cortex, both Iba-1 and GluT5 were increased in alcoholic brains relative to controls. Alternatively, no detectable change was found in amygdala nuclei. In VTA and midbrain, only GluT5, but not Iba-1 was increased in alcoholic brains. These data suggest that the enhanced expression of MCP-1 and microglia activities in alcoholic brains could contribute to ethanol-induced pathogenesis.
Collapse
Affiliation(s)
- Jun He
- Bowles Center for Alcohol Studies, Department of Pharmacology and Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA.
| | | |
Collapse
|
43
|
Surendran G, . HAEF. Neuroimmune Responses to Toxic Agents: Comparison of Organometal Electrophiles Using Detection of Antibodies to Neural Cytoskeleton and Myelin as Biomarkers. ACTA ACUST UNITED AC 2008. [DOI: 10.3923/jpt.2008.173.189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Fiedorowicz A, Figiel I, Zaremba M, Dzwonek K, Oderfeld-Nowak B. The ameboid phenotype of NG2 (+) cells in the region of apoptotic dentate granule neurons in trimethyltin intoxicated mice shares antigen properties with microglia/macrophages. Glia 2008; 56:209-22. [PMID: 18023017 DOI: 10.1002/glia.20605] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
NG2+, stellate cells present in the adult central nervous system (CNS) have been recently recognized as a distinct glial class, identified as multipotent progenitor cells. Antigenically, they are indistinguishable from oligodendroglia progenitor cells. In response to a variety of CNS insults, these cells become rapidly activated and undergo morphological changes accompanied by increased cellular proliferation. The role they play with respect to injured neurons is not clear. In our studies, we performed immunocytochemical investigations and identified a response of NG2-expressing cells in the model of selective neurodegeneration of murine dentate gyrus granule cells induced by systemic administration of trimethyltin. Dying neurons exhibited features of apoptotic cells. Around the region of neurodegeneration, we observed activation of NG2+ stellate cells and microglia. During the peak of apoptosis, we detected the appearance of NG2+ cells of the ameboid phenotype, intermingled with granule neurons. These cells also expressed markers of microglia/macrophages, OX42- and ED1-recognized antigens, an antigen recognized by O4 antibody-a marker of more differentiated cells of the oligodendroglia lineage and, in some cases, also a protein of mature oligodendroglia adenomatus polyposis coli. They also expressed nestin. Our results suggest that the injury induces a parallel transformation of both the activated glial classes: NG2+ stellate cells and resident microglia, into ameboid cells, sharing properties of both oligodendrocyte and monocyte lineages. These cells may play a role in the phagocytosis. If this assumption is verified by electron microscopy, it would indicate a novel function of NG2 transformed cells under CNS injury conditions.
Collapse
Affiliation(s)
- Anna Fiedorowicz
- Department of Molecular and Cellular Neurobiology, Laboratory of Mechanisms of Neurodegeneration and Neuroprotection, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland
| | | | | | | | | |
Collapse
|
45
|
Morita M, Imai H, Liu Y, Xu X, Sadamatsu M, Nakagami R, Shirakawa T, Nakano K, Kita Y, Yoshida K, Tsunashima K, Kato N. FK506-protective effects against trimethyltin neurotoxicity in rats: hippocampal expression analyses reveal the involvement of periarterial osteopontin. Neuroscience 2008; 153:1135-45. [PMID: 18440706 DOI: 10.1016/j.neuroscience.2008.01.078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Revised: 01/22/2008] [Accepted: 01/29/2008] [Indexed: 12/26/2022]
Abstract
There is little information on the molecular mechanisms in FK506-mediated neuroprotection. In the present study, we investigated the protective effect of FK506, an immunosuppressant and neuroprotectant, on trimethyltin (TMT)-induced neurotoxicity in the rat hippocampus. Histologically, TMT-induced neuronal damage was partially prevented by FK506 in the hippocampal CA1 region, but not in CA3. FK506 treatment significantly reduced the number of apoptotic cells in CA1, but not in CA3, and also prevented induction of cognitive deficits by TMT. Microarray analysis of the rat hippocampus detected 14 genes with TMT-induced alteration of mRNA expression that was rescued by FK506 treatment. Subsequent quantitative RT-PCR analysis confirmed elevated mRNA levels for four inflammatory genes, glutathione S-transferase, lysozyme, matrix Gla protein, and osteopontin after TMT treatment. Upregulation of these genes was reversed by FK506 treatment at 5 days postgavage. Immunohistochemistry revealed that FK506 reduced osteopontin (OPN) induction by TMT in the periarterial area at 5 days postgavage. Our data suggest that inflammatory gene expression is involved in TMT-induced damage to the hippocampal CA1 region, resulting in apoptosis, and that this process is initiated by periarterial OPN activation, and can be alleviated by FK506.
Collapse
Affiliation(s)
- M Morita
- Department of Neuroscience, Astellas Pharmacology Inc., Miyukigaoka Research Center, 21 Miyukigaoka, Tsukuba, Ibaraki, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abou-Donia MB, Goldstein LB, Bullman S, Tu T, Khan WA, Dechkovskaia AM, Abdel-Rahman AA. Imidacloprid induces neurobehavioral deficits and increases expression of glial fibrillary acidic protein in the motor cortex and hippocampus in offspring rats following in utero exposure. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2008; 71:119-30. [PMID: 18080902 DOI: 10.1080/15287390701613140] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Imidacloprid, a neonicotinoid, is one of the fastest growing insecticides in use worldwide because of its selectivity for insects. The potential for neurotoxicity following in utero exposure to imidacloprid is not known. Timed pregnant Sprague-Dawley rats (300-350 g) on d 9 of gestation were treated with a single intraperitoneal injection (i.p.) of imidacloprid (337 mg/kg, 0.75 x LD50, in corn oil). Control rats were treated with corn oil. On postnatal day (PND) 30, all male and female offspring were evaluated for (a) acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) activity, (b) ligand binding for nicotinic acetylcholine receptors (nAChR) and muscarinic acetylcholine receptors (m2 mAChR), (c) sensorimotor performance (inclined plane, beam-walking, and forepaw grip), and (d) pathological alterations in the brain (using cresyl violet and glial fibrillary acidic protein [GFAP] immunostaining). The offspring of treated mothers exhibited significant sensorimotor impairments at PND 30 during behavioral assessments. These changes were associated with increased AChE activity in the midbrain, cortex and brainstem (125-145% increase) and in plasma (125% increase). Ligand binding densities for [3H]cytosine for alpha4beta2 type nAchR did not show any significant change, whereas [3H]AFDX 384, a ligand for m2mAChR, was significantly increased in the cortex of offspring (120-155% increase) of imidacloprid-treated mothers. Histopathological evaluation using cresyl violet staining did not show any alteration in surviving neurons in various brain regions. On the other hand, there was a rise in GFAP immunostaining in motor cortex layer III, CA1, CA3, and the dentate gyrus subfield of the hippocampus of offspring of imidacloprid-treated mothers. The results indicate that gestational exposure to a single large, nonlethal, dose of imidacloprid produces significant neurobehavioral deficits and an increased expression of GFAP in several brain regions of the offspring on PND 30, corresponding to a human early adolescent age. These changes may have long-term adverse health effects in the offspring.
Collapse
Affiliation(s)
- Mohamed B Abou-Donia
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
El-Fawal HAN, O'Callaghan JP. Autoantibodies to neurotypic and gliotypic proteins as biomarkers of neurotoxicity: assessment of trimethyltin (TMT). Neurotoxicology 2007; 29:109-15. [PMID: 18001836 DOI: 10.1016/j.neuro.2007.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Revised: 08/30/2007] [Accepted: 09/23/2007] [Indexed: 12/24/2022]
Abstract
Developing accessible biomarkers of neurotoxic effects which are readily applicable to human populations poses a challenge for neurotoxicology. In the past, the neurotoxic organometal trimethyltin (TMT) has been used as a denervation tool to validate the enhanced expression of GFAP as a biomarker of astrogliosis and neurotoxicity resulting from chemical exposures. In the present study, TMT was used to assess the detection of serum autoantibodies as biomarkers of neurotoxicity. Previous studies in both human and animals have demonstrated the presence of serum autoantibodies to neurotypic [e.g., neurofilament triplet (NF)] and gliotypic proteins [myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP)] as a peripheral marker of neurodegeneration that may be applicable to humans and experimental studies. Male Long-Evans rats (45 days of age) were administered either TMT (8 mg/kg; s) or an equal volume of sterile 0.9% saline. At 1, 2, and 3 weeks post-administration, serum was collected, and rats were sacrificed for the collection of brains. Serum autoantibodies (both IgM and IgG isotypes) to NF68, NF160, NF200, MBP, and GFAP were assayed using an ELISA. Saline only rats did not have detectable levels of autoantibodies. Only sera from TMT-exposed rats had detectable titers of autoantibodies to NFs with IgG predominating starting week 2. Anti-NF68 titers were highest compared to NF160, or NF200. Autoantibodies to MBP and GFAP also were detected; however, there was no significant increase in their titers until week 3. Hippocampal GFAP, detected at these time points, was significantly (p<0.05) higher than in control brains, indicating the induction of astrogliosis as confirmed by immunostaining of brain sections. The detection of anti-NFs, as indicative of neuronal insult, was consistent with loss of hippocampal neurons in CA3 and CA1. Our results suggest that the detection of autoantibodies to neurotypic and gliotypic proteins may be used as peripheral biomarkers to reveal evidence of nervous system neurotoxicity.
Collapse
Affiliation(s)
- Hassan A N El-Fawal
- Neurotoxicology Laboratory, Division of Health Professions and Natural Sciences, Mercy College, 555 Broadway, Dobbs Ferry, NY 10522, USA.
| | | |
Collapse
|
48
|
Liang Y, Bollen AW, Gupta N. CC chemokine receptor-2A is frequently overexpressed in glioblastoma. J Neurooncol 2007; 86:153-63. [PMID: 17703277 DOI: 10.1007/s11060-007-9463-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 07/31/2007] [Indexed: 01/04/2023]
Abstract
Macrophages and monocytes migrate in response to chemotactic cytokines such as monocyte chemoattractant protein 1 (MCP-1/CCL2) in a variety of tissues including the central nervous system. Overexpression of MCP-1 has been reported in glioblastoma (GBM), which correlates to prominent macrophage infiltration characterized by this tumor type, but whether MCP-1 receptor is also expressed by the neoplastic cells remains unclear. Expression of MCP-1 and its receptor, CC chemokine receptor 2 (CCR2), were examined in GBM using cDNA microarrays and validated in two independent microarray datasets. We investigated the expression of the CCR2A isoform in human glioma cell lines and GBM, and found overexpression of CCR2A in most GBM specimens examined when compared to normal brain tissues. CCR2A is mainly localized in the cytoplasm of neoplastic cells, and pronounced neuronal cytoplasmic CCR2A immunoreactivity in tumor-infiltrating area was associated with prior chemo/radiation therapy. Glioma cells ectopically overexpressing CCR2A demonstrated increased migration compared to vector-transfected cells in vitro. Inhibition of MCP-1 synthesis suppressed migration of CCR2A-overexpressed glioma cells. Our data suggest that CCR2A might be associated with the pathobiology of GBM such as host response to treatment and tumor cell migration.
Collapse
Affiliation(s)
- Yu Liang
- Department of Neurological Surgery, Brain Tumor Research Center, University of California-San Francisco, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
49
|
Nakazawa T, Takeda M, Lewis GP, Cho KS, Jiao J, Wilhelmsson U, Fisher SK, Pekny M, Chen DF, Miller JW. Attenuated glial reactions and photoreceptor degeneration after retinal detachment in mice deficient in glial fibrillary acidic protein and vimentin. Invest Ophthalmol Vis Sci 2007; 48:2760-8. [PMID: 17525210 PMCID: PMC2613948 DOI: 10.1167/iovs.06-1398] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE To characterize the reactions of retinal glial cells (astrocytes and Müller cells) to retinal injury in mice that lack glial fibrillary acidic protein (GFAP) and vimentin (GFAP-/-Vim-/-) and to determine the role of glial cells in retinal detachment (RD)-induced photoreceptor degeneration. METHODS RD was induced by subretinal injection of sodium hyaluronate in adult wild-type (WT) and GFAP-/-Vim-/- mice. Astroglial reaction and subsequent monocyte recruitment were quantified by measuring extracellular signal-regulated kinase (Erk) and c-fos activation and the level of expression of chemokine monocyte chemoattractant protein (MCP)-1 and by counting monocytes/microglia in the detached retinas. Immunohistochemistry, immunoblotting, real-time quantitative polymerase chain reaction (PCR), and enzyme-linked immunosorbent assay (ELISA) were used. RD-induced photoreceptor degeneration was assessed by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) and measurement of outer nuclear layer (ONL) thickness. RESULTS RD-induced reactive gliosis, characterized by GFAP and vimentin upregulation, Erk and c-fos activation, MCP-1 induction, and increased monocyte recruitment in WT mice. Absence of GFAP and vimentin effectively attenuated reactive responses of retinal glial cells and monocyte infiltration. As a result, detached retinas of GFAP-/-Vim-/- mice exhibited significantly reduced numbers of TUNEL-positive photoreceptor cells and increased ONL thickness compared with those of WT mice. CONCLUSIONS The absence of GFAP and vimentin attenuates RD-induced reactive gliosis and, subsequently, limits photoreceptor degeneration. Results of this study indicate that reactive retinal glial cells contribute critically to retinal damage induced by RD and provide a new avenue for limiting photoreceptor degeneration associated with RD and other retinal diseases or damage.
Collapse
Affiliation(s)
- Toru Nakazawa
- Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Masumi Takeda
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Asahikawa Medical College, Asahikawa, Japan
| | - Geoffrey P. Lewis
- Neuroscience Research Institute, University of California, Santa Barbara, California
| | - Kin-Sang Cho
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
| | - Jianwei Jiao
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
| | - Ulrika Wilhelmsson
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at Göteborg University, Göteborg, Sweden
| | - Steven K. Fisher
- Neuroscience Research Institute, University of California, Santa Barbara, California
| | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at Göteborg University, Göteborg, Sweden
| | - Dong F. Chen
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
| | - Joan W. Miller
- Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Sriram K, O'Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol 2007; 2:140-53. [PMID: 18040839 DOI: 10.1007/s11481-007-9070-6] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 03/01/2007] [Indexed: 01/05/2023]
Abstract
Proinflammatory cytokines and chemokines have been implicated in the pathogenesis of several neurological and neurodegenerative disorders. Prominent among such factors is the pleiotropic cytokine, tumor necrosis factor (TNF)-alpha. Under normal physiological conditions, TNF-alpha orchestrates a diverse array of functions involved in immune surveillance and defense, cellular homeostasis, and protection against certain neurological insults. However, paradoxical effects of this cytokine have been observed. TNF-alpha is elicited in the brain following injury (ischemia, trauma), infection (HIV, meningitis), neurodegeneration (Alzheimer's, Parkinson's), and chemically induced neurotoxicity. The multifarious identity for this cytokine appears to be influenced by several mechanisms. Among the most prominent are the regulation of TNFalpha-induced NF-kappaB activation by adapter proteins such as TRADD and TRAF, and second, the heterogeneity of microglia and their distribution pattern across brain regions. Here, we review the differential role of TNF-alpha in response to brain injury, with emphasis on neurodegeneration, and discuss the possible mechanisms for such diverse and region-specific effects.
Collapse
Affiliation(s)
- Krishnan Sriram
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, CDC-NIOSH, 1095 Willowdale Road, Morgantown, WV 26505, USA
| | | |
Collapse
|