1
|
Abbott PW, Hardie JB, Walsh KP, Nessler AJ, Farley SJ, Freeman JH, Wemmie JA, Wendt L, Kim YC, Sowers LP, Parker KL. Knockdown of the Non-canonical Wnt Gene Prickle2 Leads to Cerebellar Purkinje Cell Abnormalities While Cerebellar-Mediated Behaviors Remain Intact. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1741-1753. [PMID: 38165577 PMCID: PMC11217148 DOI: 10.1007/s12311-023-01648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/08/2023] [Indexed: 01/04/2024]
Abstract
Autism spectrum disorders (ASD) involve brain wide abnormalities that contribute to a constellation of symptoms including behavioral inflexibility, cognitive dysfunction, learning impairments, altered social interactions, and perceptive time difficulties. Although a single genetic variation does not cause ASD, genetic variations such as one involving a non-canonical Wnt signaling gene, Prickle2, has been found in individuals with ASD. Previous work looking into phenotypes of Prickle2 knock-out (Prickle2-/-) and heterozygous mice (Prickle2-/+) suggest patterns of behavior similar to individuals with ASD including altered social interaction and behavioral inflexibility. Growing evidence implicates the cerebellum in ASD. As Prickle2 is expressed in the cerebellum, this animal model presents a unique opportunity to investigate the cerebellar contribution to autism-like phenotypes. Here, we explore cerebellar structural and physiological abnormalities in animals with Prickle2 knockdown using immunohistochemistry, whole-cell patch clamp electrophysiology, and several cerebellar-associated motor and timing tasks, including interval timing and eyeblink conditioning. Histologically, Prickle2-/- mice have significantly more empty spaces or gaps between Purkinje cells in the posterior lobules and a decreased propensity for Purkinje cells to fire action potentials. These structural cerebellar abnormalities did not impair cerebellar-associated behaviors as eyeblink conditioning and interval timing remained intact. Therefore, although Prickle-/- mice show classic phenotypes of ASD, they do not recapitulate the involvement of the adult cerebellum and may not represent the pathophysiological heterogeneity of the disorder.
Collapse
Affiliation(s)
- Parker W Abbott
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Jason B Hardie
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Kyle P Walsh
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Aaron J Nessler
- Department of Biochemistry, The University of Iowa, Iowa City, IA, 52245, USA
| | | | - John H Freeman
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - John A Wemmie
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Linder Wendt
- Department of Biostatistics, The University of Iowa, Iowa City, IA, 52245, USA
| | - Young-Cho Kim
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
- Department of Neurology, The University of Iowa, Iowa City, IA, 52245, USA
| | - Levi P Sowers
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
- Department of Pediatrics, The University of Iowa, Iowa City, IA, 52245, USA
| | - Krystal L Parker
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA.
| |
Collapse
|
2
|
Salazar‐Villacorta A, Spaull R, Chowdhury S, Mukhtyar B, Chitre M, Armstrong R, Sa M, Chandratre S, Kini U, Chinthapalli R, Mankad K, Sudhakar S, Pope S, Heales S, Kurian MA. Avoiding Premature Diagnostic Closure: Lessons from Two Children with Neurotransmitter Disorders Associated with Dual Pathology. Mov Disord Clin Pract 2024; 11:1149-1152. [PMID: 39082248 PMCID: PMC11452789 DOI: 10.1002/mdc3.14164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/24/2024] [Indexed: 10/06/2024] Open
Affiliation(s)
| | - Robert Spaull
- Developmental NeurosciencesZayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child HealthLondonUK
- Department of NeurologyGreat Ormond Street HospitalLondonUK
| | - Samyami Chowdhury
- Division of NeurologyThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Bina Mukhtyar
- Department of PaediatricsNorfolk and Norwich University HospitalNorwichUK
| | - Manali Chitre
- Department of Paediatric NeurologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Ruth Armstrong
- Department of Clinical GeneticsCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Mario Sa
- Department of Paediatric NeurologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Saleel Chandratre
- Department of Paediatric NeurologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Usha Kini
- Oxford Centre for Genomic MedicineOxford University Hospitals NHS Foundation TrustOxfordUK
| | | | - Kshitij Mankad
- Department of RadiologyGreat Ormond Street HospitalLondonUK
| | - Sniya Sudhakar
- Department of RadiologyGreat Ormond Street HospitalLondonUK
| | - Simon Pope
- Neurometabolic UnitThe National Hospital for Neurology and NeurosurgeryLondonUK
| | - Simon Heales
- Neurometabolic UnitThe National Hospital for Neurology and NeurosurgeryLondonUK
| | - Manju A. Kurian
- Developmental NeurosciencesZayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child HealthLondonUK
- Department of NeurologyGreat Ormond Street HospitalLondonUK
| |
Collapse
|
3
|
Bellver-Sanchis A, Geng Q, Navarro G, Ávila-López PA, Companys-Alemany J, Marsal-García L, Larramona-Arcas R, Miró L, Perez-Bosque A, Ortuño-Sahagún D, Banerjee DR, Choudhary BS, Soriano FX, Poulard C, Pallàs M, Du HN, Griñán-Ferré C. G9a Inhibition Promotes Neuroprotection through GMFB Regulation in Alzheimer's Disease. Aging Dis 2024; 15:311-337. [PMID: 37307824 PMCID: PMC10796087 DOI: 10.14336/ad.2023.0424-2] [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: 01/22/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Epigenetic alterations are a fundamental pathological hallmark of Alzheimer's disease (AD). Herein, we show the upregulation of G9a and H3K9me2 in the brains of AD patients. Interestingly, treatment with a G9a inhibitor (G9ai) in SAMP8 mice reversed the high levels of H3K9me2 and rescued cognitive decline. A transcriptional profile analysis after G9ai treatment revealed increased gene expression of glia maturation factor β (GMFB) in SAMP8 mice. Besides, a H3K9me2 ChIP-seq analysis after G9a inhibition treatment showed the enrichment of gene promoters associated with neural functions. We observed the induction of neuronal plasticity and a reduction of neuroinflammation after G9ai treatment, and more strikingly, these neuroprotective effects were reverted by the pharmacological inhibition of GMFB in mice and cell cultures; this was also validated by the RNAi approach generating the knockdown of GMFB/Y507A.10 in Caenorhabditis elegans. Importantly, we present evidence that GMFB activity is controlled by G9a-mediated lysine methylation as well as we identified that G9a directly bound GMFB and catalyzed the methylation at lysine (K) 20 and K25 in vitro. Furthermore, we found that the neurodegenerative role of G9a as a GMFB suppressor would mainly rely on methylation of the K25 position of GMFB, and thus G9a pharmacological inhibition removes this methylation promoting neuroprotective effects. Then, our findings confirm an undescribed mechanism by which G9a inhibition acts at two levels, increasing GMFB and regulating its function to promote neuroprotective effects in age-related cognitive decline.
Collapse
Affiliation(s)
- Aina Bellver-Sanchis
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Qizhi Geng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Department Biochemistry and Physiology, Faculty of Pharmacy. Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Pedro A. Ávila-López
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | - Júlia Companys-Alemany
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Laura Marsal-García
- Department of Biochemistry, McGill University, Montréal, Québec, Canada.
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada.
| | - Raquel Larramona-Arcas
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Lluisa Miró
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Anna Perez-Bosque
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunología Molecular, Instituto de Investigación de Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, Jalisco 44340, México.
| | | | - Bhanwar Singh Choudhary
- Department of Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India.
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, Gujarat, India.
| | - Francesc X Soriano
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Coralie Poulard
- Cancer Research Cancer Lyon, Université de Lyon, F-69000 Lyon, France.
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.
- CNRS UMR5286, Centre de Recherche en Cancérlogie de Lyon, F-69000 Lyon, France.
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Hai-Ning Du
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Li H, Luo Q, Cai S, Tie R, Meng Y, Shan W, Xu Y, Zeng X, Qian P, Huang H. Glia maturation factor-γ is required for initiation and maintenance of hematopoietic stem and progenitor cells. Stem Cell Res Ther 2023; 14:117. [PMID: 37122014 PMCID: PMC10150485 DOI: 10.1186/s13287-023-03328-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND In vertebrates, hematopoietic stem and progenitor cells (HSPCs) emerge from hemogenic endothelium in the floor of the dorsal aorta and subsequently migrate to secondary niches where they expand and differentiate into committed lineages. Glia maturation factor γ (gmfg) is a key regulator of actin dynamics that was shown to be highly expressed in hematopoietic tissue. Our goal is to investigate the role and mechanism of gmfg in embryonic HSPC development. METHODS In-depth bioinformatics analysis of our published RNA-seq data identified gmfg as a cogent candidate gene implicated in HSPC development. Loss and gain-of-function strategies were applied to study the biological function of gmfg. Whole-mount in situ hybridization, confocal microscopy, flow cytometry, and western blotting were used to evaluate changes in the number of various hematopoietic cells and expression levels of cell proliferation, cell apoptosis and hematopoietic-related markers. RNA-seq was performed to screen signaling pathways responsible for gmfg deficiency-induced defects in HSPC initiation. The effect of gmfg on YAP sublocalization was assessed in vitro by utilizing HUVEC cell line. RESULTS We took advantage of zebrafish embryos to illustrate that loss of gmfg impaired HSPC initiation and maintenance. In gmfg-deficient embryos, the number of hemogenic endothelium and HSPCs was significantly reduced, with the accompanying decreased number of erythrocytes, myelocytes and lymphocytes. We found that blood flow modulates gmfg expression and gmfg overexpression could partially rescue the reduction of HSPCs in the absence of blood flow. Assays in zebrafish and HUVEC showed that gmfg deficiency suppressed the activity of YAP, a well-established blood flow mediator, by preventing its shuttling from cytoplasm to nucleus. During HSPC initiation, loss of gmfg resulted in Notch inactivation and the induction of Notch intracellular domain could partially restore the HSPC loss in gmfg-deficient embryos. CONCLUSIONS We conclude that gmfg mediates blood flow-induced HSPC maintenance via regulation of YAP, and contributes to HSPC initiation through the modulation of Notch signaling. Our findings reveal a brand-new aspect of gmfg function and highlight a novel mechanism for embryonic HSPC development.
Collapse
Affiliation(s)
- Honghu Li
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Qian Luo
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Shuyang Cai
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ye Meng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Wei Shan
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Yulin Xu
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- School of Medicine, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, People's Republic of China.
| | - He Huang
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
| |
Collapse
|
5
|
Fathi M, Vakili K, Yaghoobpoor S, Qadirifard MS, Kosari M, Naghsh N, Asgari taei A, Klegeris A, Dehghani M, Bahrami A, Taheri H, Mohamadkhani A, Hajibeygi R, Rezaei Tavirani M, Sayehmiri F. Pre-clinical Studies Identifying Molecular Pathways of Neuroinflammation in Parkinson's Disease: A Systematic Review. Front Aging Neurosci 2022; 14:855776. [PMID: 35912090 PMCID: PMC9327618 DOI: 10.3389/fnagi.2022.855776] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/23/2022] [Indexed: 12/09/2022] Open
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by neuroinflammation, formation of Lewy bodies, and progressive loss of dopaminergic neurons in the substantia nigra of the brain. In this review, we summarize evidence obtained by animal studies demonstrating neuroinflammation as one of the central pathogenetic mechanisms of PD. We also focus on the protein factors that initiate the development of PD and other neurodegenerative diseases. Our targeted literature search identified 40 pre-clinical in vivo and in vitro studies written in English. Nuclear factor kappa B (NF-kB) pathway is demonstrated as a common mechanism engaged by neurotoxins such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA), as well as the bacterial lipopolysaccharide (LPS). The α-synuclein protein, which plays a prominent role in PD neuropathology, may also contribute to neuroinflammation by activating mast cells. Meanwhile, 6-OHDA models of PD identify microsomal prostaglandin E synthase-1 (mPGES-1) as one of the contributors to neuroinflammatory processes in this model. Immune responses are used by the central nervous system to fight and remove pathogens; however, hyperactivated and prolonged immune responses can lead to a harmful neuroinflammatory state, which is one of the key mechanisms in the pathogenesis of PD.
Collapse
Affiliation(s)
- Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Yaghoobpoor
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadegh Qadirifard
- Department of Nursing and Midwifery, Islamic Azad University, Tehran, Iran
- Department of Nursing, Garmsar Branch, Islamic Azad University, Garmsar, Iran
| | - Mohammadreza Kosari
- The First Clinical College, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Navid Naghsh
- Department of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Afsaneh Asgari taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Andis Klegeris
- Department of Biology, Faculty of Science, University of British Columbia Okanagan Campus, Kelowna, BC, Canada
| | - Mina Dehghani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ashkan Bahrami
- Faculty of Medicine, Kashan University of Medical Science, Kashan, Iran
| | - Hamed Taheri
- Dental School, Kazan Federal University, Kazan, Russia
| | - Ashraf Mohamadkhani
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramtin Hajibeygi
- Department of Cardiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Mostafa Rezaei Tavirani
| | - Fatemeh Sayehmiri
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Fatemeh Sayehmiri
| |
Collapse
|
6
|
Ahmed ME, Selvakumar GP, Kempuraj D, Raikwar SP, Thangavel R, Bazley K, Wu K, Khan O, Khan A, Zaheer S, Iyer S, Burton C, James D, Zaheer A. Glia Maturation Factor (GMF) Regulates Microglial Expression Phenotypes and the Associated Neurological Deficits in a Mouse Model of Traumatic Brain Injury. Mol Neurobiol 2020; 57:4438-4450. [PMID: 32737763 DOI: 10.1007/s12035-020-02040-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) induces inflammatory responses through microglial activation and polarization towards a more inflammatory state that contributes to the deleterious secondary brain injury. Glia maturation factor (GMF) is a pro-inflammatory protein that is responsible for neuroinflammation following insult to the brain, such as in TBI. We hypothesized that the absence of GMF in GMF-knockout (GMF-KO) mice would regulate microglial activation state and the M1/M2 phenotypes following TBI. We used the weight drop model of TBI in C57BL/6 mice wild-type (WT) and GMF-KO mice. Immunofluorescence staining, Western blot, and ELISA assays were performed to confirm TBI-induced histopathological and neuroinflammatory changes. Behavioral analysis was done to check motor coordination ability and cognitive function. We demonstrated that the deletion of GMF in GMF-KO mice significantly limited lesion volume, attenuated neuronal loss, inhibited gliosis, and activated microglia adopted predominantly anti-inflammatory (M2) phenotypes. Using an ELISA method, we found a gradual decrease in pro-inflammatory cytokines (TNF-α and IL-6) and upregulation of anti-inflammatory cytokines (IL-4 and IL-10) in GMF-KO mice compared with WT mice, thus, promoting the transition of microglia towards a more predominantly anti-inflammatory (M2) phenotype. GMF-KO mice showed significant improvement in motor ability, memory, and cognition. Overall, our results demonstrate that GMF deficiency regulates microglial polarization, which ameliorates neuronal injury and behavioral impairments following TBI in mice and concludes that GMF is a regulator of neuroinflammation and an ideal therapeutic target for the treatment of TBI.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Osaid Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.
| |
Collapse
|
7
|
Selvakumar GP, Ahmed ME, Thangavel R, Kempuraj D, Dubova I, Raikwar SP, Zaheer S, Iyer SS, Zaheer A. A role for glia maturation factor dependent activation of mast cells and microglia in MPTP induced dopamine loss and behavioural deficits in mice. Brain Behav Immun 2020; 87:429-443. [PMID: 31982500 PMCID: PMC7316620 DOI: 10.1016/j.bbi.2020.01.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/23/2022] Open
Abstract
The molecular mechanism mediating degeneration of nigrostriatal dopaminergic neurons in Parkinson's disease (PD) is not yet fully understood. Previously, we have shown the contribution of glia maturation factor (GMF), a proinflammatory protein in dopaminergic neurodegeneration mediated by activation of mast cells (MCs). In this study, methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigrostriatal neurodegeneration and astro-glial activations were determined by western blot and immunofluorescence techniques in wild type (WT) mice, MC-deficient (MC-KO) mice and GMF-deficient (GMF-KO) mice, with or without MC reconstitution before MPTP administration. We show that GMF-KO in the MCs reduces the synergistic effects of MC and Calpain1 (calcium-activated cysteine protease enzyme)-dependent dopaminergic neuronal loss that reduces motor behavioral impairments in MPTP-treated mouse. Administration of MPTP increase in calpain-mediated proteolysis in nigral dopaminergic neurons further resulting in motor decline in mice. We found that MPTP administered WT mice exhibits oxidative stress due to significant increases in the levels of malondialdehyde, superoxide dismutase and reduction in the levels of reduced glutathione and glutathione peroxidase activity as compared with both MC-KO and GMF-KO mice. The number of TH-positive neurons in the ventral tegmental area, substantia nigra and the fibers in the striatum were significantly reduced while granulocyte macrophage colony-stimulating factor (GM-CSF), MC-Tryptase, GFAP, IBA1, Calpain1 and intracellular adhesion molecule 1 expression were significantly increased in WT mice. Similarly, tyrosine hydroxylase, dopamine transporters and vesicular monoamine transporters 2 proteins expression were significantly reduced in the SN of MPTP treated WT mice. The motor behavior as analyzed by rotarod and hang test was significantly reduced in WT mice as compared with both the MC-KO and GMF-KO mice. We conclude that GMF-dependent MC activation enhances the detrimental effect of astro-glial activation-mediated oxidative stress and neuroinflammation in the midbrain, and its inhibition may slowdown the progression of PD.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65211, United States,Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65211, United States,Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65211, United States,Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65211, United States,Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Iuliia Dubova
- Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Sudhanshu P. Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65211, United States,Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Smita Zaheer
- Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Shankar S. Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65211, United States,Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65211, United States; Department of Neurology, and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
8
|
Selvakumar GP, Ahmed ME, Iyer SS, Thangavel R, Kempuraj D, Raikwar SP, Bazley K, Wu K, Khan A, Kukulka K, Bussinger B, Zaheer S, Burton C, James D, Zaheer A. Absence of Glia Maturation Factor Protects from Axonal Injury and Motor Behavioral Impairments after Traumatic Brain Injury. Exp Neurobiol 2020; 29:230-248. [PMID: 32565489 PMCID: PMC7344375 DOI: 10.5607/en20017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) causes disability and death, accelerating the progression towards Alzheimer's disease and Parkinson's disease (PD). TBI causes serious motor and cognitive impairments, as seen in PD that arise during the period of the initial insult. However, this has been understudied relative to TBI induced neuroinflammation, motor and cognitive decline that progress towards PD. Neuronal ubiquitin-C-terminal hydrolase- L1 (UCHL1) is a thiol protease that breaks down ubiquitinated proteins and its level represents the severity of TBI. Previously, we demonstrated the molecular action of glia maturation factor (GMF); a proinflammatory protein in mediating neuroinflammation and neuronal loss. Here, we show that the weight drop method induced TBI neuropathology using behavioral tests, western blotting, and immunofluorescence techniques on sections from wild type (WT) and GMF-deficient (GMF-KO) mice. Results reveal a significant improvement in substantia nigral tyrosine hydroxylase and dopamine transporter expression with motor behavioral performance in GMF-KO mice following TBI. In addition, a significant reduction in neuroinflammation was manifested, as shown by activation of nuclear factor-kB, reduced levels of inducible nitric oxide synthase, and cyclooxygenase- 2 expressions. Likewise, neurotrophins including brain-derived neurotrophic factor and glial-derived neurotrophic factor were significantly improved in GMF-KO mice than WT 72 h post-TBI. Consistently, we found that TBI enhances GFAP and UCHL-1 expression and reduces the number of dopaminergic TH-positive neurons in WT compared to GMF-KO mice 72 h post-TBI. Interestingly, we observed a reduction of THpositive tanycytes in the median eminence of WT than GMF-KO mice. Overall, we found that absence of GMF significantly reversed these neuropathological events and improved behavioral outcome. This study provides evidence that PD-associated pathology progression can be initiated upon induction of TBI.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
9
|
Ahmed ME, Selvakumar GP, Kempuraj D, Raikwar SP, Thangavel R, Bazley K, Wu K, Khan O, Kukulka K, Bussinger B, Dubova I, Zaheer S, Govindarajan R, Iyer S, Burton C, James D, Zaheer A. Neuroinflammation Mediated by Glia Maturation Factor Exacerbates Neuronal Injury in an in vitro Model of Traumatic Brain Injury. J Neurotrauma 2020; 37:1645-1655. [PMID: 32200671 DOI: 10.1089/neu.2019.6932] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) is the primary cause of death and disability affecting over 10 million people in the industrialized world. TBI causes a wide spectrum of secondary molecular and cellular complications in the brain. However, the pathological events are still not yet fully understood. Previously, we have shown that the glia maturation factor (GMF) is a mediator of neuroinflammation in neurodegenerative diseases. To identify the potential molecular pathways accompanying TBI, we used an in vitro cell culture model of TBI. A standardized injury was induced by scalpel cut through a mixed primary cell culture of astrocytes, microglia and neurons obtained from both wild type (WT) and GMF-deficient (GMF-KO) mice. Cell culture medium and whole-cell lysates were collected at 24, 48, and 72 h after the scalpel cuts injury and probed for oxidative stress using immunofluorescence analysis. Results showed that oxidative stress markers such as glutathione and glutathione peroxidase were significantly reduced, while release of cytosolic enzyme lactate dehydrogenase along with nitric oxide and prostaglandin E2 were significantly increased in injured WT cells compared with injured GMF-KO cells. In addition, injured WT cells showed increased levels of oxidation product 4-hydroxynonenal and 8-oxo-2'-deoxyguanosine compared with injured GMF-KO cells. Further, we found that injured WT cells showed a significantly increased expression of glial fibrillary acidic protein, ionized calcium binding adaptor molecule 1, and phosphorylated ezrin/radixin/moesin proteins, and reduced microtubule associated protein expression compared with injured GMF-KO cells after injury. Collectively, our results demonstrate that GMF exacerbates the oxidative stress-mediated neuroinflammation that could be brought about by TBI-induced astroglial activation.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| | - Duraisamy Kempuraj
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| | - Sudhanshu P Raikwar
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| | - Ramasamy Thangavel
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| | - Kieran Bazley
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Kristopher Wu
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Osaid Khan
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Klaudia Kukulka
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Bret Bussinger
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Iuliia Dubova
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| | - Smita Zaheer
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Raghav Govindarajan
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Shankar Iyer
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| | | | | | - Asgar Zaheer
- Department of Neurology and School of Medicine, University of Missouri, Columbia, Missouri, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Harry S Truman Memorial Veterans Hospital, Columbia, Missouri, USA
| |
Collapse
|
10
|
Kempuraj D, Selvakumar GP, Thangavel R, Ahmed ME, Zaheer S, Kumar KK, Yelam A, Kaur H, Dubova I, Raikwar SP, Iyer SS, Zaheer A. Glia Maturation Factor and Mast Cell-Dependent Expression of Inflammatory Mediators and Proteinase Activated Receptor-2 in Neuroinflammation. J Alzheimers Dis 2019; 66:1117-1129. [PMID: 30372685 DOI: 10.3233/jad-180786] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is characterized by the presence of inflammation-mediated dopaminergic neurodegeneration in the substantia nigra. Inflammatory mediators from activated microglia, astrocytes, neurons, T-cells and mast cells mediate neuroinflammation and neurodegeneration. Administration of neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induces PD like motor deficits in rodents. 1-methyl-4-phenylpyridinium (MPP+), a toxic metabolite of MPTP activates glial cells, neurons and mast cells to release neuroinflammatory mediators. Glia maturation factor (GMF), mast cells and proteinase activated receptor-2 (PAR-2) are implicated in neuroinflammation. Alpha-synuclein which induces neurodegeneration increases PAR-2 expression in the brain. However, the exact mechanisms are not yet understood. In this study, we quantified inflammatory mediators in the brains of MPTP-administered wild type (Wt), GMF-knockout (GMF-KO), and mast cell knockout (MC-KO) mice. Additionally, we analyzed the effect of MPP+, GMF, and mast cell proteases on PAR-2 expression in astrocytes and neurons in vitro. Results show that the levels of interleukin-1beta (IL-1β), tumor necrosis factor-alpha (TNF-α), and the chemokine (C-C motif) ligand 2 (CCL2) were lesser in the brains of GMF-KO mice and MC-KO mice when compared to Wt mice brain after MPTP administration. Incubation of astrocytes and neurons with MPP+, GMF, and mouse mast cell protease-6 (MMCP-6) and MMCP-7 increased the expression of PAR-2. Our studies show that the absence of mast cells and GMF reduce the expression of neuroinflammatory mediators in the brain. We conclude that GMF along with mast cell interactions with glial cells and neurons during neuroinflammation can be explored as a new therapeutic target for PD and other neuroinflammatory disorders.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.,Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.,Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.,Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.,Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Smita Zaheer
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Keerthana Kuppamma Kumar
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Anudeep Yelam
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Harleen Kaur
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Iuliia Dubova
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.,Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.,Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.,Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
11
|
Selvakumar GP, Iyer SS, Kempuraj D, Ahmed ME, Thangavel R, Dubova I, Raikwar SP, Zaheer S, Zaheer A. Molecular Association of Glia Maturation Factor with the Autophagic Machinery in Rat Dopaminergic Neurons: a Role for Endoplasmic Reticulum Stress and MAPK Activation. Mol Neurobiol 2018; 56:3865-3881. [PMID: 30218400 DOI: 10.1007/s12035-018-1340-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is one of the several neurodegenerative diseases where accumulation of aggregated proteins like α-synuclein occurs. Dysfunction in autophagy leading to this protein build-up and subsequent dopaminergic neurodegeneration may be one of the causes of PD. The mechanisms that impair autophagy remain poorly understood. 1-Methyl-4-phenylpiridium ion (MPP+) is a neurotoxin that induces experimental PD in vitro. Our studies have shown that glia maturation factor (GMF), a brain-localized inflammatory protein, induces dopaminergic neurodegeneration in PD and that suppression of GMF prevents MPP+-induced loss of dopaminergic neurons. In the present study, we demonstrate a molecular action of GMF on the autophagic machinery resulting in dopaminergic neuronal loss and propose GMF-mediated autophagic dysfunction as one of the contributing factors in PD progression. Using dopaminergic N27 neurons, primary neurons from wild type (WT), and GMF-deficient (GMF-KO) mice, we show that GMF and MPP+ enhanced expression of MAPKs increased the mammalian target of rapamycin (mTOR) activation and endoplasmic reticulum stress markers such as phospho-eukaryotic translation initiation factor 2 alpha kinase 3 (p-PERK) and inositol-requiring enzyme 1α (IRE1α). Further, GMF and MPP+ reduced Beclin 1, focal adhesion kinase (FAK) family-interacting protein of 200 kD (FIP200), and autophagy-related proteins (ATGs) 3, 5, 7, 16L, and 12. The combined results demonstrate that GMF affects autophagy through autophagosome formation with significantly reduced lysosomal-associated membrane protein 1/2, and the number of autophagic acidic vesicles. Using primary neurons, we show that MPP+ treatment leads to differential expression and localization of p62/sequestosome and in GMF-KO neurons, there was a marked increase in p62 staining implying autophagy deficiency with very little co-localization of α-synuclein and p62 as compared with WT neurons. Collectively, this study provides a bidirectional role for GMF in executing dopaminergic neuronal death mediated by autophagy that is relevant to PD.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Iuliia Dubova
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Smita Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA. .,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA.
| |
Collapse
|
12
|
Kempuraj D, Thangavel R, Selvakumar GP, Ahmed ME, Zaheer S, Raikwar SP, Zahoor H, Saeed D, Dubova I, Giler G, Herr S, Iyer SS, Zaheer A. Mast Cell Proteases Activate Astrocytes and Glia-Neurons and Release Interleukin-33 by Activating p38 and ERK1/2 MAPKs and NF-κB. Mol Neurobiol 2018; 56:1681-1693. [PMID: 29916143 DOI: 10.1007/s12035-018-1177-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022]
Abstract
Inflammatory mediators released from activated microglia, astrocytes, neurons, and mast cells mediate neuroinflammation. Parkinson's disease (PD) is characterized by inflammation-dependent dopaminergic neurodegeneration in substantia nigra. 1-Methyl-4-phenylpyridinium (MPP+), a metabolite of parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), induces inflammatory mediators' release from brain cells and mast cells. Brain cells' interaction with mast cells is implicated in neuroinflammation. However, the exact mechanisms involved are not yet clearly understood. Mouse fetal brain-derived cultured primary astrocytes and glia-neurons were incubated with mouse mast cell protease-6 (MMCP-6) and MMCP-7, and mouse bone marrow-derived mast cells (BMMCs) were incubated with MPP+ and brain protein glia maturation factor (GMF). Interleukin-33 (IL-33) released from these cells was quantitated by enzyme-linked immunosorbent assay. Both MMCP-6 and MMCP-7 induced IL-33 release from astrocytes and glia-neurons. MPP+ and GMF were used as a positive control-induced IL-33 and reactive oxygen species expression in mast cells. Mast cell proteases and MPP+ activate p38 and extracellular signal-regulated kinases 1/2 (ERK1/2), mitogen-activated protein kinases (MAPKs), and transcription factor nuclear factor-kappa B (NF-κB) in astrocytes, glia-neurons, or mast cells. Addition of BMMCs from wt mice and transduction with adeno-GMF show higher chemokine (C-C motif) ligand 2 (CCL2) release. MPP+ activated glial cells and reduced microtubule-associated protein 2 (MAP-2) expression indicating neurodegeneration. IL-33 expression increased in the midbrain and striatum of PD brains as compared with age- and sex-matched control subjects. Glial cells and neurons interact with mast cells and accelerate neuroinflammation and these interactions can be explored as a new therapeutic target to treat PD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA.
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Gvindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Haris Zahoor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Daniyal Saeed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Iuliia Dubova
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Gema Giler
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shelby Herr
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA.
| |
Collapse
|
13
|
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
|
14
|
De Sanctis C, Bellenchi GC, Viggiano D. A meta-analytic approach to genes that are associated with impaired and elevated spatial memory performance. Psychiatry Res 2018; 261:508-516. [PMID: 29395873 DOI: 10.1016/j.psychres.2018.01.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 12/11/2022]
Abstract
Spatial memory deficits are a common hallmark of psychiatric conditions, possibly due to a genetic predisposition. Thus, unravelling the relationship between genes and memory might suggest novel therapeutic targets and pathogenetic pathways. Genetic deletions are known to lead to memory deficits (post-deletion "forgetfulness" genes, PDF), or, in few instances to improve spatial memory (post-deletion "hypermnesic" genes, PDH). To assess this topic, we performed a meta-analytic approach on memory behavior in knock-out mice. We screened 300 studies from PubMed and retrieved 87 genes tested for possible effects on spatial memory. This database was crossed with the Allen Brain Atlas (brain distribution) and the Enrichr (gene function) databases. The results show that PDF genes have higher expression level in several ventral brain structures, particularly the encephalic trunk and in the hypothalamus. Moreover, part of these genes are implicated in synaptic functions. Conversely, the PDH genes are associated to G-protein coupled receptors downstream signalling. Some candidate drugs were also found to interfere with some of the PDH genes, further suggesting that this approach might help in identifying drugs to improve memory performance in psychiatric conditions.
Collapse
Affiliation(s)
- Claudia De Sanctis
- IRCCS Neuromed, Pozzilli, IS 86077, Italy; Department of Medicine and Health Sciences, University of Molise, Via De Sanctis, Campobasso 86100, Italy
| | | | - Davide Viggiano
- Department of Medicine and Health Sciences, University of Molise, Via De Sanctis, Campobasso 86100, Italy.
| |
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
|
Kempuraj D, Selvakumar GP, Zaheer S, Thangavel R, Ahmed ME, Raikwar S, Govindarajan R, Iyer S, Zaheer A. Cross-Talk between Glia, Neurons and Mast Cells in Neuroinflammation Associated with Parkinson's Disease. J Neuroimmune Pharmacol 2017; 13:100-112. [PMID: 28952015 DOI: 10.1007/s11481-017-9766-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/13/2017] [Indexed: 01/28/2023]
Abstract
Parkinson's disease (PD) is a progressive movement disorder characterized by neuroinflammation and dopaminergic neurodegeneration in the brain. 1-methyl-4-phenylpyridinium (MPP+), a metabolite of the parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces the release of inflammatory mediators from glial cells and neurons. Glia maturation factor (GMF), a brain proinflammatory protein, MPP+, and mast cell-derived inflammatory mediators induce neurodegeneration which eventually leads to PD. However, the precise mechanisms underlying interaction between glial cells, neurons and mast cells in PD still remain elusive. In the present study, mouse bone marrow-derived mast cells (BMMCs) and mouse fetal brain-derived mixed glia/neurons, astrocytes and neurons were incubated with MPP+, GMF and mast cell-derived inflammatory mediators mouse mast cell protease-6 (MMCP-6), MMCP-7 or tryptase/brain-specific serine protease-4 (tryptase/BSSP-4). Inflammatory mediators released from these cells in the culture medium were quantitated by enzyme-linked immunosorbent assay. Neurodegeneration was quantified by measuring total neurite outgrowth following microtubule-associated protein-2 immunocytochemistry. MPP+-induced significant neurodegeneration with reduced total neurite outgrowth. MPP+induced the release of tryptase/BSSP-4 from the mouse mast cells, and tryptase/BSSP-4 induced chemokine (C-C motif) ligand 2 (CCL2) release from astrocytes and glia/neurons. Overall our results suggest that MPP+, GMF, MMCP-6 or MMCP-7 stimulate glia/neurons, astrocytes or neurons to release CCL2 and matrix metalloproteinase-3. Additionally, CD40L expression is increased in BMMCs after incubation with MPP+ in a co-culture system consisting of BMMCs and glia/neurons. We propose that mast cell interaction with glial cells and neurons during neuroinflammation can be explored as a new therapeutic target for PD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Govindhasamy Pushpavathi Selvakumar
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Ramasamy Thangavel
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Mohammad Ejaz Ahmed
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Sudhanshu Raikwar
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Raghav Govindarajan
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Shankar Iyer
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Asgar Zaheer
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA. .,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA.
| |
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
|
Kempuraj D, Thangavel R, Yang E, Pattani S, Zaheer S, Santillan DA, Santillan MK, Zaheer A. Dopaminergic Toxin 1-Methyl-4-Phenylpyridinium, Proteins α-Synuclein and Glia Maturation Factor Activate Mast Cells and Release Inflammatory Mediators. PLoS One 2015; 10:e0135776. [PMID: 26275153 PMCID: PMC4537263 DOI: 10.1371/journal.pone.0135776] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/24/2015] [Indexed: 12/23/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the presence of Lewy bodies and degeneration of dopaminergic neurons. 1-methyl-4-phenylpyridinium (MPP+), a metabolite of neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and Lewy body component α-synuclein activates glia in PD pathogenesis. Mast cells and glia maturation factor (GMF) are implicated in neuroinflammatory conditions including Multiple Sclerosis. However, the role of mast cells in PD is not yet known. We have analyzed the effect of recombinant GMF, MPP+, α-synuclein and interleukin-33 (IL-33) on mouse bone marrow-derived cultured mast cells (BMMCs), human umbilical cord blood-derived cultured mast cells (hCBMCs) and mouse brain-derived cultured astrocytes by quantifying cytokines/chemokines released using ELISA or by detecting the expression of co-stimulatory molecules CD40 and CD40L by flow cytometry. GMF significantly released chemokine (C-C motif) ligand 2 (CCL2) from BMMCs but its release was reduced in BMMCs from GMF knockout mice. GMF, α-synuclein and MPP+ released IL-1β, β-hexosaminidase from BMMCs, and IL-8 from hCBMCs. GMF released CCL5, and IL-33- induced the expression of GMF from hCBMCs. Novel GMF expression was detected in hCBMCs and BMMCs by immunocytochemistry. GMF released tumor necrosis factor-alpha (TNF-α) from mouse astrocytes, and this release was greater in BMMC- astrocyte coculture than in individual cultures. Flow cytometry results showed increased IL-33 expression by GMF and MPP+, and GMF-induced CD40 expression in astrocytes. Proinflammatory mediator release by GMF, MPP+ and α-synuclein, as well as GMF expression by mast cells indicate a potential therapeutic target for neurodegenerative diseases including PD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Veterans Affairs Health Care System, Iowa City, Iowa, United States of America
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Ramasamy Thangavel
- Veterans Affairs Health Care System, Iowa City, Iowa, United States of America
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Evert Yang
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Sagar Pattani
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Smita Zaheer
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Donna A. Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Mark K. Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Asgar Zaheer
- Veterans Affairs Health Care System, Iowa City, Iowa, United States of America
- Department of Neurology, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
19
|
Yang Y, Lei C, Feng H, Sui JF. The neural circuitry and molecular mechanisms underlying delay and trace eyeblink conditioning in mice. Behav Brain Res 2014; 278:307-14. [PMID: 25448430 DOI: 10.1016/j.bbr.2014.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/28/2014] [Accepted: 10/02/2014] [Indexed: 11/30/2022]
Abstract
Classical eyeblink conditioning (EBC), a simple form of associative learning, has long been served as a model for motor learning and modulation. The neural circuitry of EBC has been studied in detail in rabbits. However, its underlying molecular mechanisms remain unclear. The advent of mouse transgenics has generated new perspectives on the studies of the neural substrates and molecular mechanisms essential for EBC. Results about EBC in mice differ in some aspects from those obtained in other mammals. Here, we review the current studies about the neural circuitry and molecular mechanisms underlying delay and trace EBC in mice. We conclude that brainstem-cerebellar circuit plays an essential role in DEC while the amygdala modulates this process, and that the medial prefrontal cortex (mPFC) as a candidate is involved in the extra-cerebellar mechanism underlying delay eyeblink conditioning (DEC) in mice. We propose the Amygdala-Cerebellum-Prefrontal Cortex-Dynamic-Conditioning Model (ACPDC model) for DEC in mice. As to trace eyeblink conditioning (TEC), the forebrain regions may play an essential role in it, whereas cerebellar cortex seems to be out of the neural circuitry in mice. Moreover, the molecular mechanisms underlying DEC and TEC in mice differ from each other. This review provides some new information and perspectives for further research on EBC.
Collapse
Affiliation(s)
- Yi Yang
- Department of Physiology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, PR China; Experimental Center of Basic Medicine, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, PR China
| | - Chen Lei
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, PR China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Jian-feng Sui
- Department of Physiology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, PR China; Experimental Center of Basic Medicine, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, PR China.
| |
Collapse
|
20
|
Clinical course of myelin oligodendrocyte glycoprotein 35-55 induced experimental autoimmune encephalomyelitis is aggravated by glia maturation factor. Neurochem Int 2011; 60:215-9. [PMID: 22226840 DOI: 10.1016/j.neuint.2011.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 12/20/2011] [Accepted: 12/22/2011] [Indexed: 11/22/2022]
Abstract
The role of glia maturation factor (GMF) in myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide-induced experimental autoimmune encephalomyelitis (EAE) was investigated using GMF-deficient (GMF-KO) mice. We demonstrate that GMF-KO mice were resistant to the MOG 35-55 peptide-induced EAE as compared to wild type (Wt) mice (two in eight versus 10 in 10). Next, we examined the effect of administration of recombinant human GMF (rGMF) on MOG 35-55 peptide-induced EAE in mice. Daily administration of rGMF, staring days 1-14, resulted in significant exacerbation of clinical symptoms. Following rGMF injections, both GMF-KO (six in eight) and Wt mice (eight in eight) developed severe EAE (maximal clinical score of 3.5-4.0) with high frequency. The histological examination revealed severe infiltration of inflammatory cells in the spinal cord of MOG-immunized Wt mice while the resistance to EAE in GMF-KO mice was characterized by the absence of inflammatory cells. Administration of rGMF in Wt mice and GMF-KO mice resulted in a significant increase in infiltrating cells in the spinal cord following MOG-immunizations. We also evaluated cytokines and chemokines production as parameters of severity of inflammation in the spinal cord of Wt versus GMF-KO mice with and without GMF-reconstitution following MOG-immunizations. Cytokines (TNF-α, IFN-γ, IL-1β, IL-6) and chemokines (CCL2, CCL3, CXCL10, GM-CSF) production were significantly greater in Wt mice than in GMF-KO mice following MOG-immunization. Furthermore, the reconstitution experiment with rGMF showed that the administration of rGMF in both, Wt mice and GMF-KO mice produced significant increase in the GMF-mediated cytokine/chemokine production.
Collapse
|
21
|
Poukkula M, Kremneva E, Serlachius M, Lappalainen P. Actin-depolymerizing factor homology domain: a conserved fold performing diverse roles in cytoskeletal dynamics. Cytoskeleton (Hoboken) 2011; 68:471-90. [PMID: 21850706 DOI: 10.1002/cm.20530] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 06/29/2011] [Accepted: 08/05/2011] [Indexed: 11/09/2022]
Abstract
Actin filaments form contractile and protrusive structures that play central roles in many processes such as cell migration, morphogenesis, endocytosis, and cytokinesis. During these processes, the dynamics of the actin filaments are precisely regulated by a large array of actin-binding proteins. The actin-depolymerizing factor homology (ADF-H) domain is a structurally conserved protein motif, which promotes cytoskeletal dynamics by interacting with monomeric and/or filamentous actin, and with the Arp2/3 complex. Despite their structural homology, the five classes of ADF-H domain proteins display distinct biochemical activities and cellular roles, only parts of which are currently understood. ADF/cofilin promotes disassembly of aged actin filaments, whereas twinfilin inhibits actin filament assembly via sequestering actin monomers and interacting with filament barbed ends. GMF does not interact with actin, but instead binds Arp2/3 complex and promotes dissociation of Arp2/3-mediated filament branches. Abp1 and drebrin are multidomain proteins that interact with actin filaments and regulate the activities of other proteins during various actin-dependent processes. The exact function of coactosin is currently incompletely understood. In this review article, we discuss the biochemical functions, cellular roles, and regulation of the five groups of ADF-H domain proteins.
Collapse
Affiliation(s)
- Minna Poukkula
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Finland
| | | | | | | |
Collapse
|
22
|
Zaheer S, Wu Y, Sahu SK, Zaheer A. Suppression of neuro inflammation in experimental autoimmune encephalomyelitis by glia maturation factor antibody. Brain Res 2011; 1373:230-9. [PMID: 21146509 PMCID: PMC3026915 DOI: 10.1016/j.brainres.2010.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 12/01/2010] [Accepted: 12/02/2010] [Indexed: 01/06/2023]
Abstract
Glia maturation factor (GMF), a protein primarily localized in the central nervous system (CNS) was isolated, sequenced and cloned in our laboratory. We previously demonstrated that GMF mediates the experimental autoimmune encephalomyelitis (EAE)-induced production of pro-inflammatory cytokines and chemokines in the central nervous system of mice. In the present study we show that immunization with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55) caused an early onset (days 7-9 post immunization) and severe EAE with a mean peak score of 3.5 ± 0.5 in mice. Neutralization of GMF with four injections of anti-GMF antibody 5 to 11 days post immunization delayed the time of onset (days 12-14 post immunization) and significantly reduced the severity of EAE (mean peak score of 1.5 ± 0.4). Consistent with these clinical scores, histological examination of the CNS of these mice revealed profound differences between GMF-antibody treated mice and isotype matched control-antibody treated mice. Histological analysis of the spinal cord and brain showed severe inflammation and demyelination in the control antibody-treated mice whereas significantly reduced inflammation and demyelination was detected in GMF-antibody-treated mice at days 8, 16, and 24 post immunization. The decreased incidence and reduced severity of EAE in GMF-antibody-treated mice was consistent with the significantly reduced expressions of pro-inflammatory cytokines and chemokines. Our overall results demonstrate that neutralization of endogenous GMF with an affinity purified GMF antibody significantly decreased the inflammation, severity and progression of immunization induced active, passive and relapsing-remitting EAE. Treatment of mice with isotype-matched control antibody did not have any effect on EAE. Taken together, these results demonstrate the critical role of GMF in EAE, and GMF antibody as a potent anti-inflammatory therapeutic agent for effectively suppressing EAE in mouse models of major types of multiple sclerosis (MS).
Collapse
Affiliation(s)
- Smita Zaheer
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA 52242
| | - Yanghong Wu
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA 52242
| | - Shailendra K. Sahu
- Veterans Affair Medical Center, Iowa City, IA 52242
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA 52242
| | - Asgar Zaheer
- Veterans Affair Medical Center, Iowa City, IA 52242
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
23
|
Zaheer S, Wu Y, Sahu SK, Zaheer A. Overexpression of glia maturation factor reinstates susceptibility to myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis in glia maturation factor deficient mice. Neurobiol Dis 2010; 40:593-8. [PMID: 20696246 PMCID: PMC2955779 DOI: 10.1016/j.nbd.2010.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 07/26/2010] [Accepted: 08/01/2010] [Indexed: 11/21/2022] Open
Abstract
Glia maturation factor (GMF), a primarily CNS localized protein was discovered and characterized in our laboratory. We previously demonstrated that GMF is the upstream regulator for excessive production and release of proinflammatory cytokines/chemokines in brain cells leading to the destruction of oligodendrocytes, the myelin forming cells, and neurons. We also reported that mice lacking endogenous GMF (GMF-deficient, GMF-KO) were resistant to myelin oligodendrocyte glycoprotein peptide 35-55 (MOG(35-55)) induced EAE, since immunization induced only delayed EAE with diminished severity. In the present study we show that a replication-defective adenovirus-GMF construct caused expression of GMF in CNS of GMF-KO mice and reinstated MOG(35-55) induced early and severe EAE. Our results show that MOG(35-55) immunization caused only a muted EAE and inflammation/demyelination in mice lacking endogenous GMF. The diminished incidence of EAE in GMF-KO mice was consistent with the significantly reduced expressions of cytokines/chemokines. The muted severity of EAE in GMF-KO mice was restored to full blown levels upon reintroduction of GMF using an adeno-GMF-virus (Adv-GMF) vector. Consistent with the clinical findings, histological examination of the CNS of mice with EAE revealed profound differences between wild type (Wt), GMF-KO, and GMF-KO mice with re-introduced GMF (GMF-KO+Adv-GMF). Spinal cord sections from mice with EAE were analyzed for the infiltration of mononuclear cells (inflammation) and myelin loss (demyelination). In Wt mice, 40% of spinal cord quadrants were positive for demyelination and 45% of spinal cord quadrants were positive for inflammation at the peak of EAE. Drastically reduced infiltrates (15%) and demyelination (10%) were found in GMF-KO mice that developed reduced severity of EAE. Upon GMF reintroduction in GMF-KO mice, MOG(35-55) immunization caused extensive monocytes infiltration (48%) and demyelination (46%), similar to that observed in the immunized Wt mice. The levels of cytokine/chemokine in the spinal cords of mice at three time points, corresponding to the onset, peak severity and recovery period of EAE, show a distinct pattern of very large increases in IFN-γ, TNF-α, GM-CSF and MCP-1 in Wt and GMF-KO+Adv-GMF mice compared to GMF-KO and GMF-KO+Adv-LacZ mice.
Collapse
Affiliation(s)
- Smita Zaheer
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA
| | - Yanghong Wu
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA
| | - Shailendra K. Sahu
- Veterans Affair Medical Center, Iowa City, IA
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA
| | - Asgar Zaheer
- Veterans Affair Medical Center, Iowa City, IA
- Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA
| |
Collapse
|
24
|
Nakano K, Kuwayama H, Kawasaki M, Numata O, Takaine M. GMF is an evolutionarily developed Adf/cofilin-super family protein involved in the Arp2/3 complex-mediated organization of the actin cytoskeleton. Cytoskeleton (Hoboken) 2010; 67:373-82. [PMID: 20517925 DOI: 10.1002/cm.20451] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Actin-depolymerizing factor (ADF)/cofilin is widely expressed in eukaryotes and plays a central role in reorganizing the actin cytoskeleton by disassembling actin filaments. The ADF-homologous domain (ADF-H) is conserved in several other actin-modulating proteins such as twinfilin, Abp1/drebrin, and coactosin. Although these proteins interact with actin via ADF-H, their effects on actin are not identical to each other. Here, we report a novel ADF/cofilin-super family protein, Gmf1 (Glia maturation factor-like protein 1), from the fission yeast Schizosaccharomyces pombe. Gmf1 is a component of actin patches, which are located on the cell cortex and required for endocytosis, and may be involved in the control of the disassembly of actin patches since its overexpression diminishes them. We provide evidence that Gmf1 binds weakly if at all to actin, but it associates with actin-related protein (Arp) 2/3 complex and suppresses its functions such as the promotion of actin polymerization and branching filaments. Importantly, Arp2/3 complex-suppressing activity is conserved among GMF-family proteins from other organisms. Given the functional plasticity of ADF-H, GMF-family proteins possibly have changed their target from conventional actin to Arps through molecular evolution.
Collapse
Affiliation(s)
- Kentaro Nakano
- Department of Structural Biosciences, Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennohdai, Tsukuba, Ibaraki, Japan.
| | | | | | | | | |
Collapse
|
25
|
Abstract
Ocular involvement in muscular dystrophy ranges from structural defects to abnormal electroretinograms. While the mechanisms underlying the abnormal retinal physiology in patients are not understood, it is thought that alpha-dystroglycan extracellular interactions are critical for normal visual function. Here we show that beta-dystroglycan anchors dystrophin and the inward rectifying K(+) channel Kir4.1 at glial endfeet and that disruption of dystrophin and potassium channel clustering in dystroglycan mutant mice is associated with an attenuation of the electroretinogram b-wave. Glial-specific inactivation of dystroglycan or deletion of the cytoplasmic domain of beta-dystroglycan was sufficient to attenuate the electroretinogram b-wave. Unexpectedly, deletion of the beta-dystroglycan cytoplasmic domain did not disrupt the laminar structure of the retina. In contrast to the role of alpha-dystroglycan extracellular interactions during early development of the CNS, beta-dystroglycan intracellular interactions are important for visual function but not the laminar development of the retina.
Collapse
|
26
|
Qi H, Ma J, Liu YM, Yang L, Peng L, Wang H, Chen HZ. Allostatic tumor-burden induces depression-associated changes in hepatoma-bearing mice. J Neurooncol 2009; 94:367-72. [PMID: 19381448 DOI: 10.1007/s11060-009-9887-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Accepted: 04/02/2009] [Indexed: 01/13/2023]
Abstract
The high incidence of depression among cancer patients has created the need for deeper insights into its underlying pathophysiological mechanism. The aim of the current study is to demonstrate the presence of depressive states due to allostatic tumor-burden in the animal model, and if possible to elucidate the mechanism(s) by which the tumor might induce the depressive state. Stress-related behavioral responses and tumor weight were evaluated 6, 8, and 12 days after implantation of H22 hepatoma cells into the right flank of mice. Brain changes associated with depression were also observed 12 days after tumor implantation. The mice that were subjected to tumor-burden exhibited a significant depression-like state which manifested as behavioral changes including prolonged time spent immobile in the tail suspension test and reduced spontaneous motor activity with tumor progression. These behavior indices changed significantly 12 days after tumor implantation and were improved by the antidepressant fluoxetine. Correspondingly, the activity of superoxide dismutase (SOD) in the cerebrum and the expression of glia maturation factor beta (GMF-beta) and brain-derived neurotrophic factor (BDNF) in the hippocampus were significantly decreased in tumor-bearing mice, and these decreases were also reversed by fluoxetine. Taken together, these data indicate that tumor-burden might induce depressive-related behavior and brain changes, and suggest that antidepressants might not only palliate depression symptoms but also modify disease processes in the auxiliary treatment of cancer.
Collapse
Affiliation(s)
- Hong Qi
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Hayashi S, Okamoto N, Makita Y, Hata A, Imoto I, Inazawa J. Heterozygous deletion at 14q22.1-q22.3 including the BMP4 gene in a patient with psychomotor retardation, congenital corneal opacity and feet polysyndactyly. Am J Med Genet A 2008; 146A:2905-10. [PMID: 18925664 DOI: 10.1002/ajmg.a.32519] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Here we report on a 1-year-old Japanese girl with psychomotor retardation, bilateral congenital corneal opacity and bilateral postaxial polysyndactyly of the feet. Although she had a normal female karyotype, our in-house bacterial artificial chromosome (BAC)-based array-CGH analysis successfully detected at least a 2.7-Mb heterozygous deletion at 14q22.1-q22.3 harboring 18 protein-coding genes. Among the genes, BMP4 was a candidate for the gene causing the abnormalities of both the eye and digits. It was previously reported that the BMP family was correlated with the morphogenesis of digits and ocular development, and Bmp4 heterozygous null mice revealed skeletal abnormalities including polydactyly and ocular anterior segment abnormalities. Patients with a deletion including BMP4 also hadabnormalities of the eye and digits. These previous reports support that a haplo-insufficiency of the BMP4 gene likely caused the congenital ocular and digit abnormalities. Moreover, among the other genes contained in the deletion, GMFB is a candidate for the gene responsible for the psychomotor retardation.
Collapse
Affiliation(s)
- Shin Hayashi
- Department of Molecular Cytogenetics, Medical Research Institute and School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
28
|
Weitzdörfer R, Höger H, Burda G, Pollak A, Lubec G. Differences in Hippocampal Protein Expression at 3 Days, 3 Weeks, and 3 Months Following Induction of Perinatal Asphyxia in the Rat. J Proteome Res 2008; 7:1945-52. [DOI: 10.1021/pr700835y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Rachel Weitzdörfer
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Harald Höger
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Gudrun Burda
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Arnold Pollak
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Gert Lubec
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| |
Collapse
|
29
|
Gerits N, Kostenko S, Shiryaev A, Johannessen M, Moens U. Relations between the mitogen-activated protein kinase and the cAMP-dependent protein kinase pathways: comradeship and hostility. Cell Signal 2008; 20:1592-607. [PMID: 18423978 DOI: 10.1016/j.cellsig.2008.02.022] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 02/28/2008] [Accepted: 02/29/2008] [Indexed: 01/05/2023]
Abstract
Inter- and intracellular communications and responses to environmental changes are pivotal for the orchestrated and harmonious operation of multi-cellular organisms. These well-tuned functions in living organisms are mediated by the action of signal transduction pathways, which are responsible for receiving a signal, transmitting and amplifying it, and eliciting the appropriate cellular responses. Mammalian cells posses numerous signal transduction pathways that, rather than acting in solitude, interconnect with each other, a phenomenon referred to as cross-talk. This allows cells to regulate the distribution, duration, intensity and specificity of the response. The cAMP/cAMP-dependent protein kinase (PKA) pathway and the mitogen-activated protein kinase (MAPK) cascades modulate common processes in the cell and multiple levels of cross-talk between these signalling pathways have been described. The first- and best-characterized interconnections are the PKA-dependent inhibition of the MAPKs ERK1/2 mediated by RAF-1, and PKA-induced activation of ERK1/2 interceded through B-RAF. Recently, novel interactions between components of these pathways and new mechanisms for cross-talk have been elucidated. This review discusses both known and novel interactions between compounds of the cAMP/PKA and MAPKs signalling pathways in mammalian cells.
Collapse
Affiliation(s)
- Nancy Gerits
- Department of Microbiology and Virology, University of Tromsø, N-9037 Tromsø, Norway
| | | | | | | | | |
Collapse
|
30
|
Patel S, Sinha A, Singh MP. Identification of differentially expressed proteins in striatum of maneb-and paraquat-induced Parkinson's disease phenotype in mouse. Neurotoxicol Teratol 2007; 29:578-85. [PMID: 17532186 DOI: 10.1016/j.ntt.2007.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 04/11/2007] [Accepted: 04/19/2007] [Indexed: 10/23/2022]
Abstract
Behavioral, phenotypic and biochemical changes induced by maneb+paraquat (MB+PQ) in experimental animals have shown their role in the etiologies of Parkinson's disease (PD); however, MB+PQ induced neuronal damage at genome and proteome level have not yet been clearly understood. The present study was undertaken to investigate the differential protein expression patterns in control and MB+PQ treated mouse striatum and to identify differentially expressed proteins. Animals were treated with and without MB+PQ, twice a week for three, six and nine weeks and proteome profiles of striatum were compared. Three differentially expressed proteins were identified as complexin-I, alpha-enolase and glia maturation factor-beta (GMF-beta) using 2D-PAGE and mass spectrometry. The differential expressions were also confirmed at transcription level by semi-quantitative RT-PCR. The results suggest the involvement of complexin-I, alpha-enolase and GMF-beta in MB+PQ induced PD phenotype in mouse.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Western
- Chromatography, Liquid
- Electrophoresis, Polyacrylamide Gel
- Fungicides, Industrial/toxicity
- Herbicides/toxicity
- Male
- Maneb/toxicity
- Mass Spectrometry
- Mice
- Mice, Inbred C57BL
- Molecular Sequence Data
- Neostriatum/drug effects
- Neostriatum/metabolism
- Nerve Tissue Proteins/biosynthesis
- Paraquat/toxicity
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/metabolism
- Parkinson Disease, Secondary/psychology
- Peptides/analysis
- Phenotype
- RNA/biosynthesis
- RNA/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, Protein
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Suman Patel
- Industrial Toxicology Research Centre (ITRC), Mahatma Gandhi Marg, Post Box-80, Lucknow-226 001, UP, India
| | | | | |
Collapse
|
31
|
Zaheer S, Wu Y, Bassett J, Yang B, Zaheer A. Glia Maturation Factor Regulation of STAT Expression: A Novel Mechanism in Experimental Autoimmune Encephalomyelitis. Neurochem Res 2007; 32:2123-31. [PMID: 17551829 DOI: 10.1007/s11064-007-9383-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Accepted: 05/09/2007] [Indexed: 10/23/2022]
Abstract
Inflammatory cytokines are implemented in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. We previously demonstrated that glia maturation factor (GMF), a brain protein, isolated, sequenced and cloned in our laboratory, induce expression of proinflammatory cytokine/chemokine in the central nervous system (CNS). We found GMF-deficient (knockout) mice relatively resistant to EAE development after immunization with encephalitogenic MOG peptide 35-55. Consistent with these findings, the expression of proinflammatory cytokines in CNS of mice with EAE differed profoundly between wild type and GMF-knockout mice. In the present study we examined the expressions of six murine signal transducers and activators of transcription (STATs) genes, which are known to regulate the cytokine-dependent signal transduction pathways in autoimmune inflammation. The expressions of STATs genes were evaluated in the brains and spinal cords of wild type and GMF-knockout mice at the peak of EAE by quantitative real-time RT-PCR. Compared to GMF-knockout mice, the expressions of STAT1, STAT2, STAT3, STAT4, STAT5, and STAT6 genes were significantly (P < 0.001) upregulated in the wild type mice exhibiting EAE symptoms. The results are consistent with the diminished development of EAE in the GMF-knockout mice. A significant suppression of STATs expression in GMF-knockout mice suggests GMF as an upstream effector of JAK/STAT signaling.
Collapse
Affiliation(s)
- Smita Zaheer
- Veterans Affairs Medical Center, Iowa City, IA, USA.
| | | | | | | | | |
Collapse
|
32
|
Menon K, Wu Y, Haas J, Sahu SK, Yang B, Zaheer A. Diminished degradation of myelin basic protein by anti-sulfatide antibody and interferon-gamma in myelin from glia maturation factor-deficient mice. Neurosci Res 2007; 58:156-63. [PMID: 17383764 PMCID: PMC1992520 DOI: 10.1016/j.neures.2007.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 01/23/2007] [Accepted: 02/15/2007] [Indexed: 10/23/2022]
Abstract
In this study we show the effect of anti-sulfatide (RmAb) antibodies and inflammatory cytokines, TNF-alpha and IFN-gamma in inducing myelin basic protein (MBP) degradation in myelin isolated from control wild type (WT) and glia maturation factor (GMF)-deficient (GMF-KO) mice. GMF was not detected in isolated myelin from WT and GMF-KO mice although it is present in brains of WT mice. Our results show that calcium-dependent neutral protease activity caused significantly elevated degradation of 18.5 and/or 17.5kDa isoforms of MBP in WT myelin treated with RmAb or IFN-gamma. In contrast, MBP degradation in isolated myelin from GMF-KO mice remained unaffected following treatment with RmAb, IFN-gamma, or GM-CSF. Neither the 14kDa isoform of MBP nor proteolipid protein (PLP) showed an elevated degradation compared to controls. A virtual absence of GM-CSF, TNF-alpha and IFN-gamma in GMF-KO brain compared to WT was also evident when the animals were challenged with MOG 35-55. Additionally, the myelin from GMF-KO mice showed difference in distribution of myelin oligodendrocyte glycoprotein (MOG) and beta-tubulin in a sucrose density gradient myelin-axolemmal fractions compared to WT. Taken together, our data suggests a role for GMF in the biochemical organization of myelin and thereby its effect on MBP degradation induced by RmAb and IFN-gamma.
Collapse
|
33
|
Zaheer A, Sahu SK, Wu Y, Zaheer A, Haas J, Lee K, Yang B. Diminished cytokine and chemokine expression in the central nervous system of GMF-deficient mice with experimental autoimmune encephalomyelitis. Brain Res 2007; 1144:239-47. [PMID: 17316572 PMCID: PMC1899479 DOI: 10.1016/j.brainres.2007.01.075] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 01/24/2007] [Accepted: 01/24/2007] [Indexed: 10/23/2022]
Abstract
Pro-inflammatory cytokines/chemokines are implemented in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model with clinical and pathological similarities to multiple sclerosis. We have previously shown that over-expression of glia maturation factor (GMF) in glial cells cause excessive production and secretion of pro-inflammatory cytokines/chemokines sufficient to destroy the myelin-forming oligodendroglial cell in vitro. In this present investigation, we evaluate the expression of pro-inflammatory mediators in the central nervous system (CNS) of GMF+/+ (wild type) mice and GMF-/- (GMF-knockout) mice at the peak of EAE induced by immunization with MOG 35-55 peptide. GMF+/+ (Wt) mice developed severe EAE with a maximal mean clinical score of 3.6+/-0.5 by day 16 post-immunization, whereas GMF-KO mice showed significantly delayed EAE with an average onset on day 26 pi with reduced mean clinical score of 1.3+/-0.3. Three of fifteen Wt mice as compared to none of GMF-KO mice died of EAE. Encephalitogenic cells from Wt mice transferred to recipient GMF-KO mice caused very mild and with low incidence of EAE. We determined the differences in the expression of cytokines, IFN-gamma, TNF-alpha, IL-1 beta, IL-6, IL-4, IL-10, and chemokines, MIP-1, MIP-2, IP-10, MCP-1, GM-CSF mRNA by quantitative real-time RT-PCR in brain and spinal cord. Our results demonstrate significantly low levels of pro-inflammatory cytokines/chemokines in the CNS of GMF-KO mice and increased expression in Wt mice with EAE. Our data suggest that GMF play a critical role in CNS inflammation.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Central Nervous System/metabolism
- Chemokines/metabolism
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Enzyme-Linked Immunosorbent Assay/methods
- Gene Expression Regulation/genetics
- Gene Expression Regulation/physiology
- Glia Maturation Factor/deficiency
- Glycoproteins
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments
- RNA, Messenger/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction/methods
Collapse
Affiliation(s)
- Asgar Zaheer
- Veterans Affair Medical Center, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Zaheer A, Zaheer S, Sahu SK, Yang B, Lim R. Reduced severity of experimental autoimmune encephalomyelitis in GMF-deficient mice. Neurochem Res 2006; 32:39-47. [PMID: 17151915 DOI: 10.1007/s11064-006-9220-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Accepted: 11/03/2006] [Indexed: 11/26/2022]
Abstract
Glia maturation factor (GMF), a highly conserved brain-specific protein, isolated, sequenced and cloned in our laboratory. Overexpression of GMF in astrocytes induces the production and secretion of granulocyte-macrophage-colony stimulating factor (GM-CSF), and subsequent immune activation of microglia, expression of several proinflammatory genes including major histocompatibility complex proteins, IL-1beta, and MIP-1beta, all associated with the development of experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. Based on GMF's ability to activate microglia and induce well-established proinflammatory mediators, including GM-CSF, we hypothesize that GMF is involved in the pathogenesis of inflammatory disease EAE. In this present investigation, using GMF-deficient mice, we study the role of GMF and how the lack of GMF affects the EAE disease. Our results show a significant decrease in incidence, delay in onset, and reduced severity of EAE in GMF-deficient mice, and support the hypothesis that GMF plays a major role in the pathogenesis of disease.
Collapse
Affiliation(s)
- Asgar Zaheer
- Veterans Affair Medical Center, Iowa City, IA, USA.
| | | | | | | | | |
Collapse
|
35
|
Baldwin RM, Garratt-Lalonde M, Parolin DAE, Krzyzanowski PM, Andrade MA, Lorimer IAJ. Protection of glioblastoma cells from cisplatin cytotoxicity via protein kinase Ciota-mediated attenuation of p38 MAP kinase signaling. Oncogene 2006; 25:2909-19. [PMID: 16331246 DOI: 10.1038/sj.onc.1209312] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Glioblastoma multiforme is an aggressive form of brain cancer that responds poorly to chemotherapy and is generally incurable. The basis for the poor response of this cancer to chemotherapy is not well understood. The atypical protein kinases C (PKCiota and PKCzeta) have previously been implicated in leukaemia cell chemoresistance. To assess the role of atypical PKC in glioblastoma cell chemoresistance, RNA interference was used to deplete human glioblastoma cells of PKCiota. Transfection of cells with either of two different RNA duplexes specific for PKCiota caused a partial sensitisation to cell death induced by the chemotherapy agent cisplatin. To screen for possible mechanisms for PKCiota-mediated chemoresistance, microarray analysis of gene expression was performed on RNA from glioblastoma cells that were either untreated or depleted of PKCiota. This identified sets of genes that were regulated either positively or negatively by PKCiota. Within the set of genes that were negatively regulated by PKCiota, the function of the gene coding for GMFbeta, an enhancer of p38 mitogen-activated protein kinase (MAP kinase) signaling, was investigated further, as the p38 MAP kinase pathway has been previously identified as a key mediator of cisplatin cytotoxicity. The expression of both GMFbeta mRNA and protein increased upon PKCiota depletion, and this was accompanied by an increase in cisplatin-activated p38 MAP kinase signaling. Transient overexpression of GMFbeta increased cisplatin-activated p38 MAP kinase signaling and also sensitised cells to cisplatin cytotoxicity. The increase in cisplatin cytotoxicity seen with PKCiota depletion was blocked by the p38 MAP kinase inhibitor SKF86002. These data show that PKCiota can confer partial resistance to cisplatin in glioblastoma cells by suppressing GMFbeta-mediated enhancement of p38 MAP kinase signaling.
Collapse
Affiliation(s)
- R M Baldwin
- Ottawa Health Research Institute, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
36
|
Zaheer A, Haas JT, Reyes C, Mathur SN, Yang B, Lim R. GMF-knockout mice are unable to induce brain-derived neurotrophic factor after exercise. Neurochem Res 2006; 31:579-84. [PMID: 16758368 DOI: 10.1007/s11064-006-9049-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2006] [Indexed: 12/01/2022]
Abstract
We earlier reported that overexpression of glia maturation factor (GMF) in cultured astrocytes enhances the production of brain-derived neurotrophic factor (BDNF). The current study was conducted to find out whether BDNF production is impaired in animals devoid of GMF. To this end GMF-knockout (KO) mice were subjected to exercise and the neurotrophin mRNAs were determined by real-time RT-PCR. Compared to wild-type (WT) mice, there is a decrease in exercise-induced BDNF in the KO mice. The observation was correlated with the finding that, in WT mice, exercise increases GMF expression. The results are consistent with the hypothesis that GMF is necessary for exercise-induction of BDNF, and that GMF may promote neuroprotection through BDNF production.
Collapse
Affiliation(s)
- Asgar Zaheer
- Department of Neurology (Division of Neurochemistry and Neurobiology), Veterans Affairs Medical Center and University of Iowa College of Medicine, Iowa City, 52242, USA.
| | | | | | | | | | | |
Collapse
|