1
|
Serrano-Martínez I, Pedreño M, Castillo-González J, Ferraz-de-Paula V, Vargas-Rodríguez P, Forte-Lago I, Caro M, Campos-Salinas J, Villadiego J, Peñalver P, Morales JC, Delgado M, González-Rey E. Cortistatin as a Novel Multimodal Therapy for the Treatment of Parkinson's Disease. Int J Mol Sci 2024; 25:694. [PMID: 38255772 PMCID: PMC10815070 DOI: 10.3390/ijms25020694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/24/2024] Open
Abstract
Parkinson's disease (PD) is a complex disorder characterized by the impairment of the dopaminergic nigrostriatal system. PD has duplicated its global burden in the last few years, becoming the leading neurological disability worldwide. Therefore, there is an urgent need to develop innovative approaches that target multifactorial underlying causes to potentially prevent or limit disease progression. Accumulating evidence suggests that neuroinflammatory responses may play a pivotal role in the neurodegenerative processes that occur during the development of PD. Cortistatin is a neuropeptide that has shown potent anti-inflammatory and immunoregulatory effects in preclinical models of autoimmune and neuroinflammatory disorders. The goal of this study was to explore the therapeutic potential of cortistatin in a well-established preclinical mouse model of PD induced by acute exposure to the neurotoxin 1-methil-4-phenyl1-1,2,3,6-tetrahydropyridine (MPTP). We observed that treatment with cortistatin mitigated the MPTP-induced loss of dopaminergic neurons in the substantia nigra and their connections to the striatum. Consequently, cortistatin administration improved the locomotor activity of animals intoxicated with MPTP. In addition, cortistatin diminished the presence and activation of glial cells in the affected brain regions of MPTP-treated mice, reduced the production of immune mediators, and promoted the expression of neurotrophic factors in the striatum. In an in vitro model of PD, treatment with cortistatin also demonstrated a reduction in the cell death of dopaminergic neurons that were exposed to the neurotoxin. Taken together, these findings suggest that cortistatin could emerge as a promising new therapeutic agent that combines anti-inflammatory and neuroprotective properties to regulate the progression of PD at multiple levels.
Collapse
Affiliation(s)
- Ignacio Serrano-Martínez
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Marta Pedreño
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Julia Castillo-González
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Viviane Ferraz-de-Paula
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Pablo Vargas-Rodríguez
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Irene Forte-Lago
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Marta Caro
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Jenny Campos-Salinas
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Javier Villadiego
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, 41013 Sevilla, Spain;
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Pablo Peñalver
- Department of Biochemistry and Molecular Pharmacology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.P.); (J.C.M.)
| | - Juan Carlos Morales
- Department of Biochemistry and Molecular Pharmacology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.P.); (J.C.M.)
| | - Mario Delgado
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Elena González-Rey
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| |
Collapse
|
2
|
Targeting Oxidative Stress Mechanisms to Treat Alzheimer’s and Parkinson’s Disease: A Critical Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7934442. [PMID: 35958022 PMCID: PMC9357807 DOI: 10.1155/2022/7934442] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/12/2022] [Indexed: 02/05/2023]
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD) are becoming more frequent as the age increases. Contemporary therapies provide symptom resolution instead of targeting underlying pathological pathways. Consequently, there is considerable heterogeneity in response to treatment. Research has elucidated multiple potential of pathophysiological mechanisms contributing to neurodegenerative conditions, among which oxidative stress pathways appear to be suitable drug targets. The oxidative stress pathway has given rise to numerous novel pharmacological therapies that may provide a new avenue for neurodegenerative diseases. For example, SKQ (plastoquinone), MitoVitE, vitamin E, SOD mimic, MitoTEMPO (SOD mimetic), and bioactive molecules like curcumin and vitamin C have indeed been examined. To better understand how oxidative stress contributes to neurodegenerative diseases (such as Alzheimer's and Parkinson's), we analyzed the medicinal qualities of medicines that target markers in the cellular oxidative pathways. The specific pathway by which mitochondrial dysfunction causes neurodegeneration will require more investigation. An animal study should be carried out on medications that tackle cellular redox mechanisms but are not currently licensed for use in the management of neurodegenerative conditions.
Collapse
|
3
|
Bhuiyan P, Chen Y, Karim M, Dong H, Qian Y. Bidirectional communication between mast cells and the gut-brain axis in neurodegenerative diseases: Avenues for therapeutic intervention. Brain Res Bull 2021; 172:61-78. [PMID: 33892083 DOI: 10.1016/j.brainresbull.2021.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 03/02/2021] [Accepted: 04/17/2021] [Indexed: 12/12/2022]
Abstract
Although the global incidence of neurodegenerative diseases has been steadily increasing, especially in adults, there are no effective therapeutic interventions. Neurodegeneration is a heterogeneous group of disorders that is characterized by the activation of immune cells in the central nervous system (CNS) (e.g., mast cells and microglia) and subsequent neuroinflammation. Mast cells are found in the brain and the gastrointestinal tract and play a role in "tuning" neuroimmune responses. The complex bidirectional communication between mast cells and gut microbiota coordinates various dynamic neuro-cellular responses, which propagates neuronal impulses from the gastrointestinal tract into the CNS. Numerous inflammatory mediators from degranulated mast cells alter intestinal gut permeability and disrupt blood-brain barrier, which results in the promotion of neuroinflammatory processes leading to neurological disorders, thereby offsetting the balance in immune-surveillance. Emerging evidence supports the hypothesis that gut-microbiota exert a pivotal role in inflammatory signaling through the activation of immune and inflammatory cells. Communication between inflammatory cytokines and neurocircuits via the gut-brain axis (GBA) affects behavioral responses, activates mast cells and microglia that causes neuroinflammation, which is associated with neurological diseases. In this comprehensive review, we focus on what is currently known about mast cells and the gut-brain axis relationship, and how this relationship is connected to neurodegenerative diseases. We hope that further elucidating the bidirectional communication between mast cells and the GBA will not only stimulate future research on neurodegenerative diseases but will also identify new opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China
| | - Yinan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China
| | - Mazharul Karim
- College of Pharmacy, Western University of Health Science, 309 East 2nd Street, Pomona, CA, 91766, USA
| | - Hongquan Dong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China.
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China.
| |
Collapse
|
4
|
Thadathil N, Delotterie DF, Xiao J, Hori R, McDonald MP, Khan MM. DNA Double-Strand Break Accumulation in Alzheimer's Disease: Evidence from Experimental Models and Postmortem Human Brains. Mol Neurobiol 2021; 58:118-131. [PMID: 32895786 DOI: 10.1007/s12035-020-02109-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/28/2020] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that accounts for a majority of dementia cases. AD is characterized by progressive neuronal death associated with neuropathological lesions consisting of neurofibrillary tangles and senile plaques. While the pathogenesis of AD has been widely investigated, significant gaps in our knowledge remain about the cellular and molecular mechanisms promoting AD. Recent studies have highlighted the role of DNA damage, particularly DNA double-strand breaks (DSBs), in the progression of neuronal loss in a broad spectrum of neurodegenerative diseases. In the present study, we tested the hypothesis that accumulation of DNA DSB plays an important role in AD pathogenesis. To test our hypothesis, we examined DNA DSB expression and DNA repair function in the hippocampus of human AD and non-AD brains by immunohistochemistry, ELISA, and RT-qPCR. We observed increased DNA DSB accumulation and reduced DNA repair function in the hippocampus of AD brains compared to the non-AD control brains. Next, we found significantly increased levels of DNA DSB and altered levels of DNA repair proteins in the hippocampus of 5xFAD mice compared to non-transgenic mice. Interestingly, increased accumulation of DNA DSBs and altered DNA repair proteins were also observed in cellular models of AD. These findings provided compelling evidence that AD is associated with accumulation of DNA DSB and/or alteration in DSB repair proteins which may influence an important early part of the pathway toward neural damage and memory loss in AD.
Collapse
Affiliation(s)
- Nidheesh Thadathil
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
| | - David F Delotterie
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
| | - Jianfeng Xiao
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
| | - Roderick Hori
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Michael P McDonald
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
- Department of Anatomy & Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA.
- Center for Muscle, Metabolism and Neuropathology, Division of Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
5
|
Shaler T, Lin H, Bakke J, Chen S, Grover A, Chang P. Particle radiation-induced dysregulation of protein homeostasis in primary human and mouse neuronal cells. LIFE SCIENCES IN SPACE RESEARCH 2020; 25:9-17. [PMID: 32414496 DOI: 10.1016/j.lssr.2020.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/12/2020] [Accepted: 02/16/2020] [Indexed: 06/11/2023]
Abstract
Space particle radiations may cause significant damage to proteins and oxidative stress in the cells within the central nervous system and pose a potential health hazard to humans in long-term manned space explorations. Dysregulation of the ubiquitin-proteasome system as evidenced by abnormal accumulation of polyubiquitin (pUb) chain linkages has been implicated in several age-related neurodegenerative disorders by mechanisms that may involve the inter-neuronal spread of toxic misfolded proteins, the induction of chronic neuroinflammation, or the inappropriate inhibition or activation of key enzymes, which could lead to dysfunction in, for example, proteolysis, or the accumulation of post-translationally-modified substrates.In this study, we employed a quantitative proteomics method to evaluate the impact of particle-radiation induced alterations in three major pUb-linked chains at lysine residues Lys-48 (K-48), Lys-63 (K-63), and Lys-11 (K-11), and probed for global proteomic changes in mouse and human neural cells that were irradiated with low doses of 250 MeV proton, 260 MeV/u silicon or 1 GeV/u iron ions. We found significant accumulation in K-48 linkage after 1 Gy protons and K-63 linkage after 0.5 Gy iron ions in human neural cells. Cells derived from different regions of the mouse brain (cortex, striatum and mesencephalon) showed differential sensitivity to particle radiation exposure. Although none of the linkages were altered after proton exposure, both K-48 and K-63 linkages in mouse striatal neuronal cells were elevated after 0.5 Gy of silicon or iron ions. Changes were also seen in proteins commonly used as markers of neural progenitor and stem cells, in DNA binding/damage repair and cellular redox pathways. In contrast, no significant changes were observed at the same time point after proton irradiation. These results suggest that the quality of the particle radiation plays a key role in the level, linkage and cell type specificity of protein homeostasis in key populations of neuronal cells.
Collapse
Affiliation(s)
- Tom Shaler
- SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025 United States
| | - Hua Lin
- SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025 United States
| | - James Bakke
- SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025 United States
| | - Sophia Chen
- SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025 United States
| | - Amber Grover
- SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025 United States
| | - Polly Chang
- SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025 United States.
| |
Collapse
|
6
|
Pathophysiology and Therapeutic Perspectives of Oxidative Stress and Neurodegenerative Diseases: A Narrative Review. Adv Ther 2020; 37:113-139. [PMID: 31782132 PMCID: PMC6979458 DOI: 10.1007/s12325-019-01148-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Indexed: 12/21/2022]
Abstract
Introduction Neurodegeneration is the term describing the death of neurons both in the central nervous system and periphery. When affecting the central nervous system, it is responsible for diseases like Alzheimer’s disease, Parkinson’s disease, Huntington’s disorders, amyotrophic lateral sclerosis, and other less frequent pathologies. There are several common pathophysiological elements that are shared in the neurodegenerative diseases. The common denominators are oxidative stress (OS) and inflammatory responses. Unluckily, these conditions are difficult to treat. Because of the burden caused by the progression of these diseases and the simultaneous lack of efficacious treatment, therapeutic approaches that could target the interception of development of the neurodegeneration are being widely investigated. This review aims to highlight the most recent proposed novelties, as most of the previous approaches have failed. Therefore, older approaches may currently be used by healthcare professionals and are not being presented. Methods This review was based on an electronic search of existing literature, using PubMed as primary source for important review articles, and important randomized clinical trials, published in the last 5 years. Reference lists from the most recent reviews, as well as additional sources of primary literature and references cited by relevant articles, were used. Results Eighteen natural pharmaceutical substances and 24 extracted or recombinant products, and artificial agents that can be used against OS, inflammation, and neurodegeneration were identified. After presenting the most common neurodegenerative diseases and mentioning some of the basic mechanisms that lead to neuronal loss, this paper presents up to date information that could encourage the development of better therapeutic strategies. Conclusions This review shares the new potential pharmaceutical and not pharmaceutical options that have been recently introduced regarding OS and inflammatory responses in neurodegenerative diseases.
Collapse
|
7
|
Raikwar SP, Kikkeri NS, Sakuru R, Saeed D, Zahoor H, Premkumar K, Mentor S, Thangavel R, Dubova I, Ahmed ME, Selvakumar GP, Kempuraj D, Zaheer S, Iyer SS, Zaheer A. Next Generation Precision Medicine: CRISPR-mediated Genome Editing for the Treatment of Neurodegenerative Disorders. J Neuroimmune Pharmacol 2019; 14:608-641. [PMID: 31011884 PMCID: PMC8211357 DOI: 10.1007/s11481-019-09849-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022]
Abstract
Despite significant advancements in the field of molecular neurobiology especially neuroinflammation and neurodegeneration, the highly complex molecular mechanisms underlying neurodegenerative diseases remain elusive. As a result, the development of the next generation neurotherapeutics has experienced a considerable lag phase. Recent advancements in the field of genome editing offer a new template for dissecting the precise molecular pathways underlying the complex neurodegenerative disorders. We believe that the innovative genome and transcriptome editing strategies offer an excellent opportunity to decipher novel therapeutic targets, develop novel neurodegenerative disease models, develop neuroimaging modalities, develop next-generation diagnostics as well as develop patient-specific precision-targeted personalized therapies to effectively treat neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, Frontotemporal dementia etc. Here, we review the latest developments in the field of CRISPR-mediated genome editing and provide unbiased futuristic insights regarding its translational potential to improve the treatment outcomes and minimize financial burden. However, despite significant advancements, we would caution the scientific community that since the CRISPR field is still evolving, currently we do not know the full spectrum of CRISPR-mediated side effects. In the wake of the recent news regarding CRISPR-edited human babies being born in China, we urge the scientific community to maintain high scientific and ethical standards and utilize CRISPR for developing in vitro disease in a dish model, in vivo testing in nonhuman primates and lower vertebrates and for the development of neurotherapeutics for the currently incurable neurodegenerative disorders. Graphical Abstract.
Collapse
Affiliation(s)
- Sudhanshu P Raikwar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA
| | - Nidhi S Kikkeri
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Ragha Sakuru
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Daniyal Saeed
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Haris Zahoor
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Keerthivaas Premkumar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shireen Mentor
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Department of Medical Biosciences, University of the Western Cape, Bellville, 7535, Republic of South Africa
| | - Ramasamy Thangavel
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA
| | - Iuliia Dubova
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA
| | - Govindhasamy P Selvakumar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA
| | - Smita Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shankar S Iyer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA
| | - Asgar Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA.
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran's Hospital, Columbia, MO, USA.
| |
Collapse
|
8
|
Michalovicz LT, Kelly KA, Vashishtha S, Ben‐Hamo R, Efroni S, Miller JV, Locker AR, Sullivan K, Broderick G, Miller DB, O’Callaghan JP. Astrocyte-specific transcriptome analysis using the ALDH1L1 bacTRAP mouse reveals novel biomarkers of astrogliosis in response to neurotoxicity. J Neurochem 2019; 150:420-440. [PMID: 31222732 PMCID: PMC6771645 DOI: 10.1111/jnc.14800] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022]
Abstract
Neurotoxicology is hampered by the inability to predict regional and cellular targets of toxicant-induced damage. Evaluating astrogliosis overcomes this problem because reactive astrocytes highlight the location of toxicant-induced damage. While enhanced expression of glial fibrillary acidic protein is a hallmark of astrogliosis, few other biomarkers have been identified. However, bacterial artificial chromosome - translating ribosome affinity purification (bacTRAP) technology allows for characterization of the actively translating transcriptome of a particular cell type; use of this technology in aldehyde dehydrogenase 1 family member L1 (ALDH1L1) bacTRAP mice can identify genes selectively expressed in astrocytes. The aim of this study was to characterize additional biomarkers of neurotoxicity-induced astrogliosis using ALDH1L1 bacTRAP mice. The known dopaminergic neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 12.5 mg/kg s.c.) was used to induce astrogliosis. Striatal tissue was obtained 12, 24, and 48 h following exposure for the isolation of actively translating RNA. Subsequently, MPTP-induced changes in this RNA pool were analyzed by microarray and 184 statistically significant, differentially expressed genes were identified. The dataset was interrogated by gene ontology, pathway, and co-expression network analyses, which identified novel genes, as well as those with known immune and inflammatory functions. Using these analyses, we were directed to several genes associated with reactive astrocytes. Of these, TIMP1 and miR-147 were identified as candidate biomarkers because of their robust increased expression following both MPTP and trimethyl tin exposures. Thus, we have demonstrated that bacTRAP can be used to identify new biomarkers of astrogliosis and aid in the characterization of astrocyte phenotypes induced by toxicant exposures. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14518.
Collapse
Affiliation(s)
- Lindsay T. Michalovicz
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - Kimberly A. Kelly
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - Saurabh Vashishtha
- Center for Clinical Systems BiologyRochester General Hospital Research InstituteRochesterNew YorkUSA
| | - Rotem Ben‐Hamo
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Sol Efroni
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Julie V. Miller
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - Alicia R. Locker
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | | | - Gordon Broderick
- Center for Clinical Systems BiologyRochester General Hospital Research InstituteRochesterNew YorkUSA
| | - Diane B. Miller
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - James P. O’Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| |
Collapse
|
9
|
Cota-Coronado A, Díaz-Martínez NF, Padilla-Camberos E, Díaz-Martínez NE. Editing the Central Nervous System Through CRISPR/Cas9 Systems. Front Mol Neurosci 2019; 12:110. [PMID: 31191241 PMCID: PMC6546027 DOI: 10.3389/fnmol.2019.00110] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/15/2019] [Indexed: 12/26/2022] Open
Abstract
The translational gap to treatments based on gene therapy has been reduced in recent years because of improvements in gene editing tools, such as the CRISPR/Cas9 system and its variations. This has allowed the development of more precise therapies for neurodegenerative diseases, where access is privileged. As a result, engineering of complexes that can access the central nervous system (CNS) with the least potential inconvenience is fundamental. In this review article, we describe current alternatives to generate systems based on CRISPR/Cas9 that can cross the blood-brain barrier (BBB) and may be used further clinically to improve treatment for neurodegeneration in Parkinson's and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Agustin Cota-Coronado
- Biotecnología Médica y Farmacéutica CONACYT, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | | | - Eduardo Padilla-Camberos
- Biotecnología Médica y Farmacéutica CONACYT, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - N Emmanuel Díaz-Martínez
- Biotecnología Médica y Farmacéutica CONACYT, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| |
Collapse
|
10
|
Activation of microglia synergistically enhances neurodegeneration caused by MPP + in human SH-SY5Y cells. Eur J Pharmacol 2019; 850:64-74. [PMID: 30684467 DOI: 10.1016/j.ejphar.2019.01.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 01/21/2023]
Abstract
While MPP+ may not directly activate microglia, the initial neuronal damage inflicted by the toxin may trigger microglia, possibly leading to synergistic pro-apoptotic interaction between neuro-inflammation and toxin-induced neurotoxicity, which may further aggravate neurodegeneration. However, what molecular targets are synergistically up or downregulated during this interaction is not well understood. Here, we addressed this by co-culturing fully differentiated human SH-SY5Y cells treated with parkinsonian toxin 1-Methyl-4-phenylpyridinium (MPP+), with endotoxin-activated microglial cell line EOC 20 to determine how this interaction affects pro-apoptotic (p38, JNK, and bax:bcl2 ratios) and pro-survival (NF-κB, MEK1) signaling at both mRNA and protein levels. Concurrent MPP+ and endotoxin-treatment aggravated a decrease in SH-SY5Y cell viability and caused strong synergistic increases in the bax:bcl2 ratio, but also NF-κB and JNK signaling. These effects were attenuated by microglia inhibitor minocycline. Altogether, these data provide further molecular insights into the important role or even conditional requirement of microglia activation in the progressive neurodegenerative nature of PD.
Collapse
|
11
|
Dincel GC, Kul O. First description of enhanced expression of transforming growth factor-alpha (TGF-α) and glia maturation factor-beta (GMF-β) correlate with severity of neuropathology in border disease virus-infected small ruminants. Microb Pathog 2019; 128:301-310. [PMID: 30654008 DOI: 10.1016/j.micpath.2019.01.015] [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: 10/30/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 11/25/2022]
Abstract
Border disease (BD) is caused by Pestivirus and characterized by severe neuropathology, and histopathologically observed severe hypomyelination. We have previously shown that small ruminants infected with border disease virus (BDV) play an important role for neuropathology and pathogenesis of severe oxidative damage in brain tissue, neuronal mtDNA; in the production of high pathologic levels of nitric oxide; in glial cell activation and stimulation of intrinsic apoptosis pathway. This study aimed to investigate the relationship between glia maturation factor beta (GMF-β) and transforming growth factor alpha (TGF-α) expressions and the causes of BDV-induced neuropathology and to investigate their role in neuropathogenesis in a way that was not presented before. Expression levels of GMF-β and TGF-α were investigated. Results of the study revealed that the levels of GMF-β (P < 0.005) and TGF-α (P < 0.005) expression in the brain tissue markedly increased in the BDV-infected animals compared to the non-infected healthy control group. While TGF-α expressions were predominantly observed in neurons, GMF-β expressions were found in astrocytes, glial cells and neurons. These results were reasonable to suggest that BDV-mediated increased GMF-β might play a pivotal role neuropathogenesis and a different type of role in the mechanism of neurodegeneration/neuropathology in the process of BD. The results also indicated that increased levels of GMF up-regulation in glial cells and neurons causes neuronal destruction, suggesting pathological pathway involving GMF-mediated brain cell cytotoxicity. It is clearly indicated that the cause of astrogliosis is due to severe TGF-a expression. This is the first study to demonstrate the expression of GMF-β and TGF-α in neurons and reactive glial cells and its association with neuropathology in BD.
Collapse
Affiliation(s)
| | - Oguz Kul
- Department of Pathology, University of Kirikkale, Kirikkale, Turkey
| |
Collapse
|
12
|
Raikwar SP, Thangavel R, Dubova I, Selvakumar GP, Ahmed ME, Kempuraj D, Zaheer SA, Iyer SS, Zaheer A. Targeted Gene Editing of Glia Maturation Factor in Microglia: a Novel Alzheimer's Disease Therapeutic Target. Mol Neurobiol 2019; 56:378-393. [PMID: 29704201 PMCID: PMC6344368 DOI: 10.1007/s12035-018-1068-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/08/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a devastating, progressive neurodegenerative disorder that leads to severe cognitive impairment in elderly patients. Chronic neuroinflammation plays an important role in the AD pathogenesis. Glia maturation factor (GMF), a proinflammatory molecule discovered in our laboratory, is significantly upregulated in various regions of AD brains. We have previously reported that GMF is predominantly expressed in the reactive glial cells surrounding the amyloid plaques (APs) in the mouse and human AD brain. Microglia are the major source of proinflammatory cytokines and chemokines including GMF. Recently clustered regularly interspaced short palindromic repeats (CRISPR) based genome editing has been recognized to study the functions of genes that are implicated in various diseases. Here, we investigated if CRISPR-Cas9-mediated GMF gene editing leads to inhibition of GMF expression and suppression of microglial activation. Confocal microscopy of murine BV2 microglial cell line transduced with an adeno-associated virus (AAV) coexpressing Staphylococcus aureus (Sa) Cas9 and a GMF-specific guide RNA (GMF-sgRNA) revealed few cells expressing SaCas9 while lacking GMF expression, thereby confirming successful GMF gene editing. To further improve GMF gene editing efficiency, we developed lentiviral vectors (LVs) expressing either Streptococcus pyogenes (Sp) Cas9 or GMF-sgRNAs. BV2 cells cotransduced with LVs expressing SpCas9 and GMF-sgRNAs revealed reduced GMF expression and the presence of indels in the exons 2 and 3 of the GMF coding sequence. Lipopolysaccharide (LPS) treatment of GMF-edited cells led to reduced microglial activation as shown by reduced p38 MAPK phosphorylation. We believe that targeted in vivo GMF gene editing has a significant potential for developing a unique and novel AD therapy.
Collapse
Affiliation(s)
- Sudhanshu P Raikwar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Ramasamy Thangavel
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Iuliia Dubova
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Smita A Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shankar S Iyer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Asgar Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA.
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA.
| |
Collapse
|
13
|
Xu FF, Zhang ZB, Wang YY, Wang TH. Brain-Derived Glia Maturation Factor β Participates in Lung Injury Induced by Acute Cerebral Ischemia by Increasing ROS in Endothelial Cells. Neurosci Bull 2018; 34:1077-1090. [PMID: 30191459 PMCID: PMC6246848 DOI: 10.1007/s12264-018-0283-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/03/2018] [Indexed: 02/05/2023] Open
Abstract
Brain damage can cause lung injury. To explore the mechanism underlying the lung injury induced by acute cerebral ischemia (ACI), we established a middle cerebral artery occlusion (MCAO) model in male Sprague-Dawley rats. We focused on glia maturation factor β (GMFB) based on quantitative analysis of the global rat serum proteome. Polymerase chain reaction, western blotting, and immunofluorescence revealed that GMFB was over-expressed in astrocytes in the brains of rats subjected to MCAO. We cultured rat primary astrocytes and confirmed that GMFB was also up-regulated in primary astrocytes after oxygen-glucose deprivation (OGD). We subjected the primary astrocytes to Gmfb RNA interference before OGD and collected the conditioned medium (CM) after OGD. We then used the CM to culture pulmonary microvascular endothelial cells (PMVECs) acquired in advance and assessed their status. The viability of the PMVECs improved significantly when Gmfb was blocked. Moreover, ELISA assays revealed an elevation in GMFB concentration in the medium after OGD. Cell cultures containing recombinant GMFB showed increased levels of reactive oxygen species and a deterioration in the state of the cells. In conclusion, GMFB is up-regulated in astrocytes after ACI, and brain-derived GMFB damages PMVECs by increasing reactive oxygen species. GMFB might thus be an initiator of the lung injury induced by ACI.
Collapse
Affiliation(s)
- Fei-Fei Xu
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Bin Zhang
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yang-Yang Wang
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Goode BL, Sweeney MO, Eskin JA. GMF as an Actin Network Remodeling Factor. Trends Cell Biol 2018; 28:749-760. [PMID: 29779865 DOI: 10.1016/j.tcb.2018.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/22/2018] [Accepted: 04/23/2018] [Indexed: 10/24/2022]
Abstract
Glia maturation factor (GMF) has recently been established as a regulator of the actin cytoskeleton with a unique role in remodeling actin network architecture. Conserved from yeast to mammals, GMF is one of five members of the ADF-H family of actin regulatory proteins, which includes ADF/cofilin, Abp1/Drebrin, Twinfilin, and Coactosin. GMF does not bind actin, but instead binds the Arp2/3 complex with high affinity. Through this association, GMF catalyzes the debranching of actin filament networks and inhibits actin nucleation by Arp2/3 complex. Here, we discuss GMF's emerging role in controlling actin filament spatial organization and dynamics underlying cell motility, endocytosis, and other biological processes. Further, we attempt to reconcile these functions with its earlier characterization as a cell differentiation factor.
Collapse
Affiliation(s)
- Bruce L Goode
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, MA 02454 USA.
| | - Meredith O Sweeney
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, MA 02454 USA
| | - Julian A Eskin
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, MA 02454 USA
| |
Collapse
|
15
|
Fan J, Fong T, Chen X, Chen C, Luo P, Xie H. Glia maturation factor-β: a potential therapeutic target in neurodegeneration and neuroinflammation. Neuropsychiatr Dis Treat 2018; 14:495-504. [PMID: 29445286 PMCID: PMC5810533 DOI: 10.2147/ndt.s157099] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glia maturation factor-β (GMFB) is considered to be a growth and differentiation factor for both glia and neurons. GMFB has been found to be upregulated in several neuroinflammation and neurodegeneration conditions. It may function by mediating apoptosis and by modulating the expression of superoxide dismutase, granulocyte-macrophage colony-stimulating factor, and neurotrophin. In this review, we mainly discussed the role of GMFB in several neuroinflammatory and neurodegenerative diseases. On review of the literature, we propose that GMFB may be a promising therapeutic target for neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Junsheng Fan
- Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Tszhei Fong
- Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xinjie Chen
- Second School of Clinic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuyun Chen
- Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Peng Luo
- Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Haiting Xie
- Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Yu S, Wang X, He X, Wang Y, Gao S, Ren L, Shi Y. Curcumin exerts anti-inflammatory and antioxidative properties in 1-methyl-4-phenylpyridinium ion (MPP(+))-stimulated mesencephalic astrocytes by interference with TLR4 and downstream signaling pathway. Cell Stress Chaperones 2016; 21:697-705. [PMID: 27164829 PMCID: PMC4908001 DOI: 10.1007/s12192-016-0695-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/12/2016] [Accepted: 04/23/2016] [Indexed: 01/01/2023] Open
Abstract
Neuroinflammation is closely associated with the pathophysiology of neurodegenerative diseases including Parkinson's disease (PD). Recent evidence indicates that astrocytes also play pro-inflammatory roles in the central nervous system (CNS) by activation with toll-like receptor (TLR) ligands. Therefore, targeting anti-inflammation may provide a promising therapeutic strategy for PD. Curcumin, a polyphenolic compound isolated from Curcuma longa root, has been commonly used for the treatment of neurodegenerative diseases. However, the details of how curcumin exerts neuroprotection remain uncertain. Here, we investigated the protective effect of curcumin on 1-methyl-4-phenylpyridinium ion-(MPP(+)-) stimulated primary astrocytes. Our results showed that MPP(+) stimulation resulted in significant production of tumor necrosis factor (TNF)-α, interleukin (IL-6), and reactive oxygen species (ROS) in primary mesencephalic astrocytes. Curcumin pretreatment decreased the levels of these pro-inflammatory cytokines while increased IL-10 expression in MPP(+)-stimulated astrocytes. In addition, curcumin increased the levels of antioxidant glutathione (GSH) and reduced ROS production. Our results further showed that curcumin decreased the levels of TLR4 and its downstream effectors including NF-κB, IRF3, MyD88, and TIRF that are induced by MPP(+) as well as inhibited the immunoreactivity of TLR4 and morphological activation in MPP(+)-stimulated astrocytes. Together, data suggest that curcumin might exert a beneficial effect on neuroinflammation in the pathophysiology of PD.
Collapse
Affiliation(s)
- Song Yu
- Department of Histology and Embryology, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, People's Republic of China
| | - Xu Wang
- Department of Histology and Embryology, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, People's Republic of China
| | - Xingliang He
- Department of Sports Medicine, Shenyang Sports University, Shenyang, Liaoning, 110102, People's Republic of China
| | - Yue Wang
- Department of Histology and Embryology, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, People's Republic of China
| | - Sujie Gao
- Department of Cardiac Function, The Second Staff Hospital of Liaohe Oilfield Company, Panjin, Liaoning, 124011, People's Republic of China
| | - Lu Ren
- Department of Mental Diseases, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, People's Republic of China
| | - Yan Shi
- Department of Internal Medicine of Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, 79 East Chongshan Road, Shenyang, Liaoning, 110847, People's Republic of China.
| |
Collapse
|
17
|
Kempuraj D, Thangavel R, Fattal R, Pattani S, Yang E, Zaheer S, Santillan DA, Santillan MK, Zaheer A. Mast Cells Release Chemokine CCL2 in Response to Parkinsonian Toxin 1-Methyl-4-Phenyl-Pyridinium (MPP(+)). Neurochem Res 2015; 41:1042-9. [PMID: 26646004 DOI: 10.1007/s11064-015-1790-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/18/2015] [Accepted: 11/24/2015] [Indexed: 01/29/2023]
Abstract
Microglial activation and release of inflammatory cytokines and chemokines are crucial events in neuroinflammation. Microglial cells interact and respond to other inflammatory cells such as T cells and mast cells as well as inflammatory mediators secreted from these cells. Recent studies have shown that neuroinflammation causes and accelerates neurodegenerative disease such as Parkinson's disease (PD) pathogenesis. 1-methyl-4-phenyl-pyridinium ion (MPP(+)), the active metabolite of neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydro pyridine activates glial cells and mediate neurodegeneration through release of inflammatory mediators. We have shown that glia maturation factor (GMF) activates glia and induces neuroinflammation and neurodegeneration and that MPP(+) activates mast cells and release proinflammatory cytokines and chemokines. The chemokine (C-C motif) ligand 2 (CCL2) levels have been shown to be elevated and play a role in PD pathogenesis. In the present study, we analyzed if MPP(+) activates mouse and human mast cells to release chemokine CCL2. Mouse bone marrow-derived mast cells (BMMCs) and human umbilical cord blood-derived cultured mast cells (hCBMCs) were incubated with MPP(+) (10 µM) for 24 h and CCL2 levels were measured in the supernatant media by ELISA. MPP(+)-significantly induced CCL2 release from BMMCs and hCBMCs. Additionally, GMF overexpression in BMMCs obtained from wild-type mice released significantly more CCL2, while BMMCs obtained from GMF-deficient mice showed less CCL2 release. Further, we show that MPP(+)-induced CCL2 release was greater in BMMCs-astrocyte co-culture conditions. Uncoupling protein 4 (UCP4) which is implicated in neurodegenerative diseases including PD was detected in BMMCs by immunocytochemistry. Our results suggest that mast cells may play role in PD pathogenesis.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Veterans Affairs Health Care System, Iowa City, IA, 52242, USA
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Ramasamy Thangavel
- Veterans Affairs Health Care System, Iowa City, IA, 52242, USA
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Ranan Fattal
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Sagar Pattani
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Evert Yang
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Smita Zaheer
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Mark K Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Asgar Zaheer
- Veterans Affairs Health Care System, Iowa City, IA, 52242, USA.
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.
| |
Collapse
|
18
|
Segura-Aguilar J, Kostrzewa RM. Neurotoxin mechanisms and processes relevant to Parkinson's disease: an update. Neurotox Res 2015; 27:328-54. [PMID: 25631236 DOI: 10.1007/s12640-015-9519-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 12/14/2022]
Abstract
The molecular mechanism responsible for degenerative process in the nigrostriatal dopaminergic system in Parkinson's disease (PD) remains unknown. One major advance in this field has been the discovery of several genes associated to familial PD, including alpha synuclein, parkin, LRRK2, etc., thereby providing important insight toward basic research approaches. There is an consensus in neurodegenerative research that mitochon dria dysfunction, protein degradation dysfunction, aggregation of alpha synuclein to neurotoxic oligomers, oxidative and endoplasmic reticulum stress, and neuroinflammation are involved in degeneration of the neuromelanin-containing dopaminergic neurons that are lost in the disease. An update of the mechanisms relating to neurotoxins that are used to produce preclinical models of Parkinson´s disease is presented. 6-Hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, and rotenone have been the most wisely used neurotoxins to delve into mechanisms involved in the loss of dopaminergic neurons containing neuromelanin. Neurotoxins generated from dopamine oxidation during neuromelanin formation are likewise reviewed, as this pathway replicates neurotoxin-induced cellular oxidative stress, inactivation of key proteins related to mitochondria and protein degradation dysfunction, and formation of neurotoxic aggregates of alpha synuclein. This survey of neurotoxin modeling-highlighting newer technologies and implicating a variety of processes and pathways related to mechanisms attending PD-is focused on research studies from 2012 to 2014.
Collapse
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Independencia 1027, Casilla, 70000, Santiago 7, Chile,
| | | |
Collapse
|