1
|
Nowak-Kiczmer M, Niedziela N, Czuba ZP, Sowa P, Wierzbicki K, Lubczyński M, Adamczyk-Sowa M. A comparison of serum inflammatory parameters in progressive forms of multiple sclerosis. Mult Scler Relat Disord 2023; 79:105004. [PMID: 37738756 DOI: 10.1016/j.msard.2023.105004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/24/2023]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic, inflammatory demyelinating disease of the central nervous system. Primary progressive MS (PPMS) is diagnosed in approximately 10-15 % of MS patients. Disease-modifying therapies (DMT) are less effective in modifying the course of progressive types of MS. It seems that inflammatory processes differ in the MS subtypes. OBJECTIVES The objective of this study was to assess differences in the inflammatory parameters between PPMS and other courses of MS. MATERIALS AND METHODS A total of 84 subjects were included in the study. The study group was divided according to the course of MS into the following categories: PPMS (n = 24); SPMS-secondary progressive multiple sclerosis (n = 14); RRMS-relapsing-remitting multiple sclerosis (n = 46). PPMS patients were further divided into treated with ocrelizumab and treatment-naive groups. The concentrations of serum inflammatory parameters were evaluated. RESULTS PPMS and SPMS significantly differed in the serum levels of sCD30, gp130, sIL-6R alpha, osteopontin, pentraxin-3 and sTNF-R1. The serum concentrations of IFN-alpha2, IL-10, IL-20, IL-29 and osteopontin significantly differed between PPMS and RRMS. The serum levels of BAFF, IL-19, IL-20, pentraxin-3, s-TNF-R1 and s-TNF-R2 significantly differed between PPMS treated with ocrelizumab and treatment-naive. CONCLUSION Although inflammatory processes take part in the pathogenesis of all types of MS, they differ between MS courses. Serum inflammatory parameters seem to be promising biomarkers in helping to differentiate courses of MS, and in assessing reactions to DMT treatment. Further investigations on their usage are required.
Collapse
Affiliation(s)
- Maria Nowak-Kiczmer
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland.
| | - Natalia Niedziela
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Zenon P Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Paweł Sowa
- Department of Otorhinolaryngology and Oncological Laryngology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Krzysztof Wierzbicki
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Michał Lubczyński
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| |
Collapse
|
2
|
Ceyzériat K, Nicolaides A, Amossé Q, Fossey C, Cailly T, Fabis F, Garibotto V, Escartin C, Tournier BB, Millet P. Reactive astrocytes mediate TSPO overexpression in response to sustained CNTF exposure in the rat striatum. Mol Brain 2023; 16:57. [PMID: 37408083 PMCID: PMC10320938 DOI: 10.1186/s13041-023-01041-x] [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: 01/12/2023] [Accepted: 06/01/2023] [Indexed: 07/07/2023] Open
Abstract
The 18 kDa translocator protein (TSPO) is a classical marker of neuroinflammation targeted for in vivo molecular imaging. Microglial cells were originally thought to be the only source of TSPO overexpression but astrocytes, neurons and endothelial cells can also up-regulate TSPO depending on the pathological context. This study aims to determine the cellular origin of TSPO overexpression in a simplified model of neuroinflammation and to identify the molecular pathways involved. This is essential to better interpret TSPO molecular imaging in preclinical and clinical settings. We used lentiviral vectors (LV) to overexpress the ciliary neurotrophic factor (CNTF) in the right striatum of 2-month-old Sprague Dawley rats. A LV encoding for β-Galactosidase (LV-LacZ) was used as control. One month later, TSPO expression was measured by single-photon emission computed tomography (SPECT) imaging using [125I]CLINDE. The fluorescence-activated cell sorting to radioligand-treated tissue (FACS-RTT) method was used to quantify TSPO levels in acutely sorted astrocytes, microglia, neurons and endothelial cells. A second cohort was injected with LV-CNTF and a LV encoding suppressor of cytokine signaling 3 (SOCS3), to inhibit the JAK-STAT3 pathway specifically in astrocytes. GFAP and TSPO expressions were quantified by immunofluorescence. We measured a significant increase in TSPO signal in response to CNTF by SPECT imaging. Using FACS-RTT, we observed TSPO overexpression in reactive astrocytes (+ 153 ± 62%) but also in microglia (+ 2088 ± 500%) and neurons (+ 369 ± 117%), accompanied by an increase in TSPO binding sites per cell in those three cell populations. Endothelial cells did not contribute to TSPO signal increase. Importantly, LV-SOCS3 reduced CNTF-induced astrocyte reactivity and decreased global TSPO immunoreactivity (-71% ± 30%), suggesting that TSPO overexpression is primarily mediated by reactive astrocytes. Overall, this study reveals that CNTF induces TSPO in multiple cell types in the rat striatum, through the JAK2-STAT3 pathway in astrocytes, identifying this cell type as the primary mediator of CNTF effects neuroinflammatory processes. Our results highlight the difficulty to interpret TSPO imaging in term of cellular origin without addition cellular analysis by FACS-RTT or quantitative immunostainings. Consequently, TSPO should only be used as a global marker of neuroinflammation.
Collapse
Affiliation(s)
- Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland.
- Department of Psychiatry, University of Geneva, Avenue de la Roseraie, 64, Geneva, 1205, Switzerland.
- CIBM Center for BioMedical Imaging, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | - Alekos Nicolaides
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Avenue de la Roseraie, 64, Geneva, 1205, Switzerland
| | - Quentin Amossé
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Avenue de la Roseraie, 64, Geneva, 1205, Switzerland
| | - Christine Fossey
- Normandie Univ, UNICAEN, Centre d'Études et de Recherche sur le Médicament de Normandie (CERMN), Caen, France
| | - Thomas Cailly
- Normandie Univ, UNICAEN, Centre d'Études et de Recherche sur le Médicament de Normandie (CERMN), Caen, France
- Department of Nuclear Medicine, CHU Cote de Nacre, Caen, France
- Normandie Univ, UNICAEN, IMOGERE, Caen, France
- Institut Blood and Brain @Caen-Normandie (BB@C) Boulevard Henri Becquerel, Caen, 14074, France
| | - Frédéric Fabis
- Normandie Univ, UNICAEN, Centre d'Études et de Recherche sur le Médicament de Normandie (CERMN), Caen, France
| | - Valentina Garibotto
- CIBM Center for BioMedical Imaging, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of nuclear medicine and molecular imaging, Diagnostic Department, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, Gif-sur-Yvette, France
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Avenue de la Roseraie, 64, Geneva, 1205, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Avenue de la Roseraie, 64, Geneva, 1205, Switzerland
| |
Collapse
|
3
|
Sirkisoon SR, Wong GL, Aguayo NR, Doheny DL, Zhu D, Regua AT, Arrigo A, Manore SG, Wagner C, Thomas A, Singh R, Xing F, Jin G, Watabe K, Lo HW. Breast cancer extracellular vesicles-derived miR-1290 activates astrocytes in the brain metastatic microenvironment via the FOXA2→CNTF axis to promote progression of brain metastases. Cancer Lett 2022; 540:215726. [PMID: 35589002 PMCID: PMC9387054 DOI: 10.1016/j.canlet.2022.215726] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/21/2022] [Accepted: 05/04/2022] [Indexed: 01/09/2023]
Abstract
Mechanisms underlying breast cancer brain metastasis (BCBM) are still unclear. In this study, we observed that extracellular vesicles (EVs) secreted from breast cancer cells with increased expression of tGLI1, a BCBM-promoting transcription factor, strongly activated astrocytes. EV-derived microRNA/miRNA microarray revealed tGLI1-positive breast cancer cells highly secreted miR-1290 and miR-1246 encapsulated in EVs. Genetic knockin/knockout studies established a direct link between tGLI1 and both miRNAs. Datamining and analysis of patient samples revealed that BCBM patients had more circulating EV-miRs-1290/1246 than those without metastasis. Ectopic expression of miR-1290 or miR-1246 strongly activated astrocytes whereas their inhibitors abrogated the effect. Conditioned media from miR-1290- or miR-1246-overexpressing astrocytes promoted mammospheres. Furthermore, miRs-1290/1246 suppressed expression of FOXA2 transcription repressor, leading to CNTF cytokine secretion and subsequent activation of astrocytes. Finally, we conducted a mouse study to demonstrate that astrocytes overexpressing miR-1290, but not miR-1246, enhanced intracranial colonization and growth of breast cancer cells. Collectively, our findings demonstrate, for the first time, that breast cancer EV-derived miR-1290 and miR-1246 activate astrocytes in the brain metastatic microenvironment and that EV-derived miR-1290 promotes progression of brain metastases through the novel EV-miR-1290→FOXA2→CNTF signaling axis.
Collapse
Affiliation(s)
- Sherona R Sirkisoon
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Grace L Wong
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Noah R Aguayo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Daniel L Doheny
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Dongqin Zhu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Angelina T Regua
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Austin Arrigo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Sara G Manore
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Calvin Wagner
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Alexandra Thomas
- Department of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Breast Cancer Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Fei Xing
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Breast Cancer Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Guangxu Jin
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Breast Cancer Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Breast Cancer Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
4
|
Paron F, Barattucci S, Cappelli S, Romano M, Berlingieri C, Stuani C, Laurents D, Mompeán M, Buratti E. Unravelling the toxic effects mediated by the neurodegenerative disease-associated S375G mutation of TDP-43 and its S375E phosphomimetic variant. J Biol Chem 2022; 298:102252. [PMID: 35835219 PMCID: PMC9364110 DOI: 10.1016/j.jbc.2022.102252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 12/05/2022] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) is a nucleic acid–binding protein found in the nucleus that accumulates in the cytoplasm under pathological conditions, leading to proteinopathies, such as frontotemporal dementia and ALS. An emerging area of TDP-43 research is represented by the study of its post-translational modifications, the way they are connected to disease-associated mutations, and what this means for pathological processes. Recently, we described a novel mutation in TDP-43 in an early onset ALS case that was affecting a potential phosphorylation site in position 375 (S375G). A preliminary characterization showed that both the S375G mutation and its phosphomimetic variant, S375E, displayed altered nuclear–cytoplasmic distribution and cellular toxicity. To better investigate these effects, here we established cell lines expressing inducible WT, S375G, and S375E TDP-43 variants. Interestingly, we found that these mutants do not seem to affect well-studied aspects of TDP-43, such as RNA splicing or autoregulation, or protein conformation, dynamics, or aggregation, although they do display dysmorphic nuclear shape and cell cycle alterations. In addition, RNA-Seq analysis of these cell lines showed that although the disease-associated S375G mutation and its phosphomimetic S375E variant regulate distinct sets of genes, they have a common target in mitochondrial apoptotic genes. Taken together, our data strongly support the growing evidence that alterations in TDP-43 post-translational modifications can play a potentially important role in disease pathogenesis and provide a further link between TDP-43 pathology and mitochondrial health.
Collapse
Affiliation(s)
- Francesca Paron
- Molecular Pathology, International Centre for Genetic and Engineering Biotechnology (ICGEB), Trieste, Italy
| | - Simone Barattucci
- Molecular Pathology, International Centre for Genetic and Engineering Biotechnology (ICGEB), Trieste, Italy
| | - Sara Cappelli
- Molecular Pathology, International Centre for Genetic and Engineering Biotechnology (ICGEB), Trieste, Italy
| | - Maurizio Romano
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Christian Berlingieri
- Molecular Pathology, International Centre for Genetic and Engineering Biotechnology (ICGEB), Trieste, Italy
| | - Cristiana Stuani
- Molecular Pathology, International Centre for Genetic and Engineering Biotechnology (ICGEB), Trieste, Italy
| | - Douglas Laurents
- "Rocasolano" Institute for Physical Chemistry, Spanish National Research Council, Serrano 119, 28006, Madrid, Spain
| | - Miguel Mompeán
- "Rocasolano" Institute for Physical Chemistry, Spanish National Research Council, Serrano 119, 28006, Madrid, Spain
| | - Emanuele Buratti
- Molecular Pathology, International Centre for Genetic and Engineering Biotechnology (ICGEB), Trieste, Italy.
| |
Collapse
|
5
|
Astrocytes in Multiple Sclerosis-Essential Constituents with Diverse Multifaceted Functions. Int J Mol Sci 2021; 22:ijms22115904. [PMID: 34072790 PMCID: PMC8198285 DOI: 10.3390/ijms22115904] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 11/19/2022] Open
Abstract
In multiple sclerosis (MS), astrocytes respond to the inflammatory stimulation with an early robust process of morphological, transcriptional, biochemical, and functional remodeling. Recent studies utilizing novel technologies in samples from MS patients, and in an animal model of MS, experimental autoimmune encephalomyelitis (EAE), exposed the detrimental and the beneficial, in part contradictory, functions of this heterogeneous cell population. In this review, we summarize the various roles of astrocytes in recruiting immune cells to lesion sites, engendering the inflammatory loop, and inflicting tissue damage. The roles of astrocytes in suppressing excessive inflammation and promoting neuroprotection and repair processes is also discussed. The pivotal roles played by astrocytes make them an attractive therapeutic target. Improved understanding of astrocyte function and diversity, and the mechanisms by which they are regulated may lead to the development of novel approaches to selectively block astrocytic detrimental responses and/or enhance their protective properties.
Collapse
|
6
|
Tripathi A, Pandit I, Perles A, Zhou Y, Cheng F, Dutta R. Identifying miRNAs in multiple sclerosis gray matter lesions that correlate with atrophy measures. Ann Clin Transl Neurol 2021; 8:1279-1291. [PMID: 33978322 PMCID: PMC8164853 DOI: 10.1002/acn3.51365] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/10/2021] [Accepted: 03/27/2021] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVE Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system (CNS). Though MS was initially considered to be a white matter demyelinating disease, myelin loss in cortical gray matter has been reported in all disease stages. We previously identified microRNAs (miRNAs) in white matter lesions (WMLs) that are detected in serum from MS patients. However, miRNA expression profiles in gray matter lesions (GMLs) from progressive MS brains are understudied. METHODS We used a combination of global miRNAs and gene expression profiling of GMLs and independent validation using real-time quantitative polymerase chain reaction (RT-qPCR), immuno-in situ hybridization, and immunohistochemistry. RESULTS Compared to matched myelinated gray matter (GM) regions, we identified 82 miRNAs in GMLs, of which 10 were significantly upregulated and 17 were significantly downregulated. Among these 82 miRNAs, 13 were also detected in serum and importantly were associated with brain atrophy in MS patients. The predicted target mRNAs of these miRNAs belonged to pathways associated with axonal guidance, TGF-β signaling, and FOXO signaling. Further, using state-of-the-art human protein-protein interactome network analysis, we mapped the four key GM atrophy-associated miRNAs (hsa-miR-149*, hsa-miR-20a, hsa-miR-29c, and hsa-miR-25) to their target mRNAs that were also changed in GMLs. INTERPRETATION Our study identifies miRNAs altered in GMLs in progressive MS brains that correlate with atrophy measures. As these miRNAs were also detected in sera of MS patients, these could act as markers of GML demyelination in MS.
Collapse
Affiliation(s)
- Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| | - Ishani Pandit
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Aaron Perles
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yadi Zhou
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Feixiong Cheng
- Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA.,Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Potential of N-trans feruloyl tyramine from Lycium barbarum fruit extract on neurogenesis and neurotrophins; targeting TrkA/ERK/CREB signaling pathway. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104432] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
8
|
Duncan GJ, Simkins TJ, Emery B. Neuron-Oligodendrocyte Interactions in the Structure and Integrity of Axons. Front Cell Dev Biol 2021; 9:653101. [PMID: 33763430 PMCID: PMC7982542 DOI: 10.3389/fcell.2021.653101] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
The myelination of axons by oligodendrocytes is a highly complex cell-to-cell interaction. Oligodendrocytes and axons have a reciprocal signaling relationship in which oligodendrocytes receive cues from axons that direct their myelination, and oligodendrocytes subsequently shape axonal structure and conduction. Oligodendrocytes are necessary for the maturation of excitatory domains on the axon including nodes of Ranvier, help buffer potassium, and support neuronal energy metabolism. Disruption of the oligodendrocyte-axon unit in traumatic injuries, Alzheimer's disease and demyelinating diseases such as multiple sclerosis results in axonal dysfunction and can culminate in neurodegeneration. In this review, we discuss the mechanisms by which demyelination and loss of oligodendrocytes compromise axons. We highlight the intra-axonal cascades initiated by demyelination that can result in irreversible axonal damage. Both the restoration of oligodendrocyte myelination or neuroprotective therapies targeting these intra-axonal cascades are likely to have therapeutic potential in disorders in which oligodendrocyte support of axons is disrupted.
Collapse
Affiliation(s)
- Greg J. Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Tyrell J. Simkins
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, VA Portland Health Care System, Portland, OR, United States
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
9
|
Bagnato F, Gauthier SA, Laule C, Moore GRW, Bove R, Cai Z, Cohen-Adad J, Harrison DM, Klawiter EC, Morrow SA, Öz G, Rooney WD, Smith SA, Calabresi PA, Henry RG, Oh J, Ontaneda D, Pelletier D, Reich DS, Shinohara RT, Sicotte NL. Imaging Mechanisms of Disease Progression in Multiple Sclerosis: Beyond Brain Atrophy. J Neuroimaging 2021; 30:251-266. [PMID: 32418324 DOI: 10.1111/jon.12700] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
Clinicians involved with different aspects of the care of persons with multiple sclerosis (MS) and scientists with expertise on clinical and imaging techniques convened in Dallas, TX, USA on February 27, 2019 at a North American Imaging in Multiple Sclerosis Cooperative workshop meeting. The aim of the workshop was to discuss cardinal pathobiological mechanisms implicated in the progression of MS and novel imaging techniques, beyond brain atrophy, to unravel these pathologies. Indeed, although brain volume assessment demonstrates changes linked to disease progression, identifying the biological mechanisms leading up to that volume loss are key for understanding disease mechanisms. To this end, the workshop focused on the application of advanced magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging techniques to assess and measure disease progression in both the brain and the spinal cord. Clinical translation of quantitative MRI was recognized as of vital importance, although the need to maintain a relatively short acquisition time mandated by most radiology departments remains the major obstacle toward this effort. Regarding PET, the panel agreed upon its utility to identify ongoing pathological processes. However, due to costs, required expertise, and the use of ionizing radiation, PET was not considered to be a viable option for ongoing care of persons with MS. Collaborative efforts fostering robust study designs and imaging technique standardization across scanners and centers are needed to unravel disease mechanisms leading to progression and discovering medications halting neurodegeneration and/or promoting repair.
Collapse
Affiliation(s)
- Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Susan A Gauthier
- Judith Jaffe Multiple Sclerosis Center, Department of Neurology, Feil Family Brain and Mind Institute, and Department of Radiology, Weill Cornell Medicine, New York, NY
| | - Cornelia Laule
- Department of Radiology, Pathology, and Laboratory Medicine, Department of Physics and Astronomy, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - George R Wayne Moore
- Department of Pathology and Laboratory Medicine, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Riley Bove
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University, New Haven, CT
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal and Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Quebec, Canada
| | - Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD
| | - Eric C Klawiter
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sarah A Morrow
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Gülin Öz
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN
| | - William D Rooney
- Advanced Imaging Research Center, Departments of Biomedical Engineering, Neurology, and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | - Seth A Smith
- Radiology and Radiological Sciences and Vanderbilt University Imaging Institute, Vanderbilt University Medical Center, and Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Roland G Henry
- Departments of Neurology, Radiology and Biomedical Imaging, and the UC San Francisco & Berkeley Bioengineering Graduate Group, University of California San Francisco, San Francisco, CA
| | - Jiwon Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Neurology, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Daniel Pelletier
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
| | -
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
10
|
Talebi M, Majdi A, Nasiri E, Naseri A, Sadigh-Eteghad S. The correlation between circulating inflammatory, oxidative stress, and neurotrophic factors level with the cognitive outcomes in multiple sclerosis patients. Neurol Sci 2020; 42:2291-2300. [DOI: 10.1007/s10072-020-04807-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/06/2020] [Indexed: 10/23/2022]
|
11
|
Linnerbauer M, Rothhammer V. Protective Functions of Reactive Astrocytes Following Central Nervous System Insult. Front Immunol 2020; 11:573256. [PMID: 33117368 PMCID: PMC7561408 DOI: 10.3389/fimmu.2020.573256] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022] Open
Abstract
Astrocytes play important roles in numerous central nervous system disorders including autoimmune inflammatory, hypoxic, and degenerative diseases such as Multiple Sclerosis, ischemic stroke, and Alzheimer’s disease. Depending on the spatial and temporal context, activated astrocytes may contribute to the pathogenesis, progression, and recovery of disease. Recent progress in the dissection of transcriptional responses to varying forms of central nervous system insult has shed light on the mechanisms that govern the complexity of reactive astrocyte functions. While a large body of research focuses on the pathogenic effects of reactive astrocytes, little is known about how they limit inflammation and contribute to tissue regeneration. However, these protective astrocyte pathways might be of relevance for the understanding of the underlying pathology in disease and may lead to novel targeted approaches to treat autoimmune inflammatory and degenerative disorders of the central nervous system. In this review article, we have revisited the emerging concept of protective astrocyte functions and discuss their role in the recovery from inflammatory and ischemic disease as well as their role in degenerative disorders. Focusing on soluble astrocyte derived mediators, we aggregate the existing knowledge on astrocyte functions in the maintenance of homeostasis as well as their reparative and tissue-protective function after acute lesions and in neurodegenerative disorders. Finally, we give an outlook of how these mediators may guide future therapeutic strategies to tackle yet untreatable disorders of the central nervous system.
Collapse
Affiliation(s)
- Mathias Linnerbauer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Veit Rothhammer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Kin K, Yasuhara T, Kameda M, Tomita Y, Umakoshi M, Kuwahara K, Kin I, Kidani N, Morimoto J, Okazaki M, Sasaki T, Tajiri N, Borlongan CV, Date I. Cell encapsulation enhances antidepressant effect of the mesenchymal stem cells and counteracts depressive-like behavior of treatment-resistant depressed rats. Mol Psychiatry 2020; 25:1202-1214. [PMID: 30108315 DOI: 10.1038/s41380-018-0208-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/05/2018] [Accepted: 06/20/2018] [Indexed: 12/20/2022]
Abstract
Despite the advances in pharmacological therapies, only the half of depressed patients respond to currently available treatment. Thus, the need for further investigation and development of effective therapies, especially those designed for treatment-resistant depression, has been sorely needed. Although antidepressant effects of mesenchymal stem cells (MSCs) have been reported, the potential benefit of this cell therapy on treatment-resistant depression is unknown. Cell encapsulation may enhance the survival rate of grafted cells, but the therapeutic effects and mechanisms mediating encapsulation of MSCs remain unexplored. Here, we showed that encapsulation enhanced the antidepressant effects of MSCs by attenuating depressive-like behavior of Wistar Kyoto (WKY) rats, which are considered as a promising animal model of treatment-resistant depression. The implantation of encapsulated MSCs (eMSCs) into the lateral ventricle counteracted depressive-like behavior and enhanced the endogenous neurogenesis in the subventricular zone (SVZ) and the dentate gyrus (DG) of the hippocampus, whereas the implantation of MSCs without encapsulation or the implantation of eMSCs into the striatum did not show such ameliorative effects. eMSCs displayed robust and stable secretion of vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor, fibroblast growth factor-2, and ciliary neurotrophic factor (CNTF), and the implantation of eMSCs into the lateral ventricle activated relevant pathways associated with these growth factors. Additionally, eMSCs upregulated intrinsic expression of VEGF and CNTF and their receptors. This study suggests that the implantation of eMSCs into the lateral ventricle exerted antidepressant effects likely acting via neurogenic pathways, supporting their utility for depression treatment.
Collapse
Affiliation(s)
- Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan.
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Masahiro Kameda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Yousuke Tomita
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Michiari Umakoshi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Ken Kuwahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Ittetsu Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Naoya Kidani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Jun Morimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Mihoko Okazaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Naoki Tajiri
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan.,Department of Psychology, Kibi International University Graduate School of Psychology, 8, iga-cho, takahashi-shi, Okayama, 716-8508, Japan
| | - Cesario V Borlongan
- Department of Neurosurgery, University of South Florida College Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| |
Collapse
|
13
|
Magliozzi R, Howell OW, Durrenberger P, Aricò E, James R, Cruciani C, Reeves C, Roncaroli F, Nicholas R, Reynolds R. Meningeal inflammation changes the balance of TNF signalling in cortical grey matter in multiple sclerosis. J Neuroinflammation 2019; 16:259. [PMID: 31810488 PMCID: PMC6898969 DOI: 10.1186/s12974-019-1650-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022] Open
Abstract
Background Recent studies of cortical pathology in secondary progressive multiple sclerosis have shown that a more severe clinical course and the presence of extended subpial grey matter lesions with significant neuronal/glial loss and microglial activation are associated with meningeal inflammation, including the presence of lymphoid-like structures in the subarachnoid space in a proportion of cases. Methods To investigate the molecular consequences of pro-inflammatory and cytotoxic molecules diffusing from the meninges into the underlying grey matter, we carried out gene expression profiling analysis of the motor cortex from 20 post-mortem multiple sclerosis brains with and without substantial meningeal inflammation and 10 non-neurological controls. Results Gene expression profiling of grey matter lesions and normal appearing grey matter not only confirmed the substantial pathological cell changes, which were greatest in multiple sclerosis cases with increased meningeal inflammation, but also demonstrated the upregulation of multiple genes/pathways associated with the inflammatory response. In particular, genes involved in tumour necrosis factor (TNF) signalling were significantly deregulated in MS cases compared with controls. Increased meningeal inflammation was found to be associated with a shift in the balance of TNF signalling away from TNFR1/TNFR2 and NFkB-mediated anti-apoptotic pathways towards TNFR1- and RIPK3-mediated pro-apoptotic/pro-necroptotic signalling in the grey matter, which was confirmed by RT-PCR analysis. TNFR1 was found expressed preferentially on neurons and oligodendrocytes in MS cortical grey matter, whereas TNFR2 was predominantly expressed by astrocytes and microglia. Conclusions We suggest that the inflammatory milieu generated in the subarachnoid space of the multiple sclerosis meninges by infiltrating immune cells leads to increased demyelinating and neurodegenerative pathology in the underlying grey matter due to changes in the balance of TNF signalling.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Department of Brain Sciences, Department of Medicine, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK. .,Neurology Unit, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Policlinico G.B. Rossi, P.le L.A. Scuro, 10, 37134, Verona, Italy.
| | - Owain William Howell
- Department of Brain Sciences, Department of Medicine, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.,Institute for Life Sciences, Swansea University, Swansea, Wales
| | - Pascal Durrenberger
- Department of Brain Sciences, Department of Medicine, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Eleonora Aricò
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Rachel James
- Department of Brain Sciences, Department of Medicine, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Carolina Cruciani
- Department of Brain Sciences, Department of Medicine, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | | | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Richard Nicholas
- Department of Brain Sciences, Department of Medicine, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Richard Reynolds
- Department of Brain Sciences, Department of Medicine, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
14
|
Pöyhönen S, Er S, Domanskyi A, Airavaara M. Effects of Neurotrophic Factors in Glial Cells in the Central Nervous System: Expression and Properties in Neurodegeneration and Injury. Front Physiol 2019; 10:486. [PMID: 31105589 PMCID: PMC6499070 DOI: 10.3389/fphys.2019.00486] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 04/08/2019] [Indexed: 12/28/2022] Open
Abstract
Astrocytes, oligodendrocytes, and microglia are abundant cell types found in the central nervous system and have been shown to play crucial roles in regulating both normal and disease states. An increasing amount of evidence points to the critical importance of glia in mediating neurodegeneration in Alzheimer’s and Parkinson’s diseases (AD, PD), and in ischemic stroke, where microglia are involved in initial tissue clearance, and astrocytes in the subsequent formation of a glial scar. The importance of these cells for neuronal survival has previously been studied in co-culture experiments and the search for neurotrophic factors (NTFs) initiated after finding that the addition of conditioned media from astrocyte cultures could support the survival of primary neurons in vitro. This led to the discovery of the potent dopamine neurotrophic factor, glial cell line-derived neurotrophic factor (GDNF). In this review, we focus on the relationship between glia and NTFs including neurotrophins, GDNF-family ligands, CNTF family, and CDNF/MANF-family proteins. We describe their expression in astrocytes, oligodendrocytes and their precursors (NG2-positive cells, OPCs), and microglia during development and in the adult brain. Furthermore, we review existing data on the glial phenotypes of NTF knockout mice and follow NTF expression patterns and their effects on glia in disease models such as AD, PD, stroke, and retinal degeneration.
Collapse
Affiliation(s)
- Suvi Pöyhönen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Safak Er
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
Tripathi A, Volsko C, Datta U, Regev K, Dutta R. Expression of disease-related miRNAs in white-matter lesions of progressive multiple sclerosis brains. Ann Clin Transl Neurol 2019; 6:854-862. [PMID: 31139683 PMCID: PMC6530016 DOI: 10.1002/acn3.750] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/26/2019] [Accepted: 02/11/2019] [Indexed: 01/15/2023] Open
Abstract
Background MicroRNA (miRNA) expression in the serum of multiple sclerosis (MS) patients has been correlated with white matter (WM) magnetic resonance imaging (MRI) abnormalities. The expression levels and cellular specificity of the target genes of these miRNAs are unknown in MS brain. Objective The aim of this study was to analyze and validate the expression of miRNAs, previously reported as dysregulated in sera of MS patients, in white‐matter lesions (WMLs) of progressive MS brains. Methods We performed global miRNA expression profiling analysis in demyelinated WMLs of progressive MS brains (n = 5) and compared the significantly altered miRNAs to previously identified miRNAs from sera of MS patients. Top dysregulated miRNAs common between the two datasets were validated in an independent cohort of MS brains by quantitative PCR (qPCR) and in situ hybridization. Results Among the miRNAs that were significantly changed in WML tissues, 11 were similar to pathogenic and 12 were common to protective miRNAs previously identified in sera and correlating with WM MRI abnormalities. Importantly, the expression levels of 58% of the protective miRNAs (7 of 12) were decreased in MS lesions compared to surrounding normal‐appearing tissue. Target genes of these miRNAs were also altered in MS lesions and queries of cell‐specific databases identified astrocytes and microglia as the key cellular expressers of these genes in MS brains. Conclusions We identified miRNAs that correlate with MRI abnormalities in lesioned tissue from MS brains.
Collapse
Affiliation(s)
- Ajai Tripathi
- Department of Neurosciences Cleveland Clinic Cleveland Ohio
| | | | - Ushasi Datta
- Department of Neurosciences Cleveland Clinic Cleveland Ohio
| | - Keren Regev
- Department of Neurology Tel Aviv Sourasky Medical Center Tel Aviv Israel
| | - Ranjan Dutta
- Department of Neurosciences Cleveland Clinic Cleveland Ohio
| |
Collapse
|
16
|
Haindl MT, Köck U, Zeitelhofer‐Adzemovic M, Fazekas F, Hochmeister S. The formation of a glial scar does not prohibit remyelination in an animal model of multiple sclerosis. Glia 2019; 67:467-481. [PMID: 30484905 PMCID: PMC6588096 DOI: 10.1002/glia.23556] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 01/12/2023]
Abstract
The role of astrocytes in the pathophysiology of multiple sclerosis (MS) is discussed controversially. Especially the formation of the glial scar is often believed to act as a barrier for remyelination. At the same time, astrocytes are known to produce factors that influence oligodendrocyte precursor cell (OPC) survival. To explore these mechanisms, we investigated the astrocytic reaction in an animal model induced by immunization with myelin oligodendrocyte glycoprotein (MOG) in Dark Agouti (DA) rats, which mimics most of the histological features of MS. We correlated the astroglial reaction by immunohistochemistry (IHC) for glial fibrillary acidic protein (GFAP) to the remyelination capacity by in situ hybridization for mRNA of proteolipid protein (PLP), indicative of OPCs, over the full course of the disease. PLP mRNA peaked in early remyelinating lesions while the amount of GFAP positive astrocytes was highest in remyelinated lesions. In shadow plaques, we found at the same time all features of a glial scar and numbers of OPCs and mature oligodendrocytes, which were nearly equal to that in unaffected white matter areas. To assess the plaque environment, we furthermore quantitatively analyzed factors expressed by astrocytes previously suggested to influence remyelination. From our data, we conclude that remyelination occurs despite an abundant glial reaction in this animal model. The different patterns of astrocytic factors and the occurrence of different astrocytic phenotypes during lesion evolution furthermore indicate a finely regulated, balanced astrocytic involvement leading to successful repair.
Collapse
Affiliation(s)
| | - Ulrike Köck
- Center for Brain ResearchMedical University of ViennaViennaAustria
| | | | - Franz Fazekas
- Department of NeurologyMedical University of GrazGrazAustria
| | | |
Collapse
|
17
|
Li H, Chen L, Ma X, Cui P, Lang W, Hao J. Shared Gene Expression Between Multiple Sclerosis and Ischemic Stroke. Front Genet 2019; 9:598. [PMID: 30809253 PMCID: PMC6379658 DOI: 10.3389/fgene.2018.00598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/15/2018] [Indexed: 01/22/2023] Open
Abstract
Patients with multiple sclerosis (MS) appear to have an increased risk of ischemic stroke (IS). Although MS and IS have very different phenotypes, gene-based and pathway-based analyses of large-scale genome-wide association studies (GWAS) have increasingly enhanced our understanding of these two diseases. Whether there are common molecular mechanisms connecting MS and IS is still unclear. Here, we describe the outcome of gene-based test and pathway-based analysis of GWAS datasets that explored potential gene expression links between MS and IS. After identifying significant gene sets individually of MS and IS, we performed pathway-based analysis in four biological pathway databases (KEGG, PANTHER, REACTOME, and WikiPathways) and GO categories. We discovered that there were 9 shared pathways between MS and IS in KEGG, 2 in PANTHER, 14 in REACTOME, 1 in WikiPathways, and 194 in GO annotations (p < 0.05). These results provide an improved understanding about possible shared mechanisms and treatments strategies for MS and IS. They also provide some basis for further studies of how these two diseases are linked at the molecular level.
Collapse
Affiliation(s)
- He Li
- Department of Neurology and Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Ministry of Education and Tianjin City, Tianjin, China
| | - Lin Chen
- Department of Neurology and Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Ministry of Education and Tianjin City, Tianjin, China
| | - Xiaofeng Ma
- Department of Neurology and Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Ministry of Education and Tianjin City, Tianjin, China
| | - Pan Cui
- Department of Neurology and Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Ministry of Education and Tianjin City, Tianjin, China
| | - Wenjing Lang
- Department of Neurology and Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Ministry of Education and Tianjin City, Tianjin, China
| | - Junwei Hao
- Department of Neurology and Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
18
|
Young TL, Zychowski KE, Denson JL, Campen MJ. Blood-brain barrier at the interface of air pollution-associated neurotoxicity and neuroinflammation. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
19
|
Kin K, Yasuhara T, Borlongan CV, Date I. Encapsulated stem cells ameliorate depressive-like behavior via growth factor secretion. Brain Circ 2018; 4:128-132. [PMID: 30450420 PMCID: PMC6187943 DOI: 10.4103/bc.bc_17_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/04/2018] [Accepted: 09/10/2018] [Indexed: 12/28/2022] Open
Abstract
As prevalence of depression continues to rise around the world, there remains a stagnation of available treatments as the affected population grows. The subset of treatment-resistant depression also is on the rise highlighting the need for innovative treatments to address this issue. Mesenchymal stem cells (MSCs) have been reported to attenuate depression-like behaviors, however, the effects of encapsulation of MSCs have yet to be investigated. Encapsulation of MSCs exhibited prolonged survival of exogenous cell injection accompanied with increased secretion of neurotrophic factors including vascular endothelial growth factor, ciliary neurotrophic factor, and others. The enhanced expression of these factors highlights the ability of encapsulated MSCs to upregulate the respective signaling pathways, which are associated with depression pathology and activation of neurogenesis. This treatment identifies a promising therapeutic option for depression, specifically treatment-resistant depression. Further, evaluation of long-term effects of the treatment is warranted. This paper is a review article. Referred literature in this paper has been listed in the references section. The datasets supporting the conclusions of this article are available online by searching various databases including PubMed. Some original themes in this article come from the laboratory practice in our research center and the authors' experiences.
Collapse
Affiliation(s)
- Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, US
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| |
Collapse
|
20
|
Magliozzi R, Reynolds R, Calabrese M. MRI of cortical lesions and its use in studying their role in MS pathogenesis and disease course. Brain Pathol 2018; 28:735-742. [PMID: 30020563 PMCID: PMC8028295 DOI: 10.1111/bpa.12642] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/21/2018] [Indexed: 02/01/2023] Open
Abstract
Cortical grey matter (GM) demyelination is present from the earliest stages of multiple sclerosis (MS) and is associated with physical deficits and cognitive impairment. In particular, the rate of disability progression in MS, both in the relapsing and progressive phases, appears to be strictly associated with degenerative GM demyelination and diffuse cortical atrophy. In the last decade, several histopathological studies and advanced radiological methodologies have contributed to better identify the exact involvement/load of cortical pathology in MS, even if the specific inflammatory features and the precise cell and molecular mechanisms of GM demyelination and neurodegeneration in MS remain still not fully understood. It has been proposed that a combined neuropathology, imaging and molecular approach may help to define a more detailed characterization and precise assessment of the heterogeneous features of GM injury and inflammation in MS. This, in turn, will possibly identify specific imaging and biohumoral (cerebrospinal fluid/serum) correlates of cortical pathology that may have an important role in predicting and monitor the disease evolution.
Collapse
Affiliation(s)
- R. Magliozzi
- Neurology BDepartment of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
- Division of Brain SciencesDepartment of MedicineImperial College LondonLondonUnited Kingdom
| | - R. Reynolds
- Division of Brain SciencesDepartment of MedicineImperial College LondonLondonUnited Kingdom
| | - M. Calabrese
- Neurology BDepartment of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| |
Collapse
|
21
|
Kirschen GW, Liu H, Lang T, Liang X, Ge S, Xiong Q. The radial organization of neuronal primary cilia is acutely disrupted by seizure and ischemic brain injury. FRONTIERS IN BIOLOGY 2017; 12:124-138. [PMID: 28473847 PMCID: PMC5412953 DOI: 10.1007/s11515-017-1447-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Neuronal primary cilia are sensory organelles that are critically involved in the proper growth, development, and function of the central nervous system (CNS). Recent work also suggests that they signal in the context of CNS injury, and that abnormal ciliary signaling may be implicated in neurological diseases. METHODS We quantified the distribution of neuronal primary cilia alignment throughout the normal adult mouse brain by immunohistochemical staining for the primary cilia marker adenylyl cyclase III (ACIII) and measuring the angles of primary cilia with respect to global and local coordinate planes. We then introduced two different models of acute brain insult-temporal lobe seizure and cerebral ischemia, and re-examined neuronal primary cilia distribution, as well as ciliary lengths and the proportion of neurons harboring cilia. RESULTS Under basal conditions, cortical cilia align themselves radially with respect to the cortical surface, while cilia in the dentate gyrus align themselves radially with respect to the granule cell layer. Cilia of neurons in the striatum and thalamus, by contrast, exhibit a wide distribution of ciliary arrangements. In both cases of acute brain insult, primary cilia alignment was significantly disrupted in a region-specific manner, with areas affected by the insult preferentially disrupted. Further, the two models promoted differential effects on ciliary lengths, while only the ischemia model decreased the proportion of ciliated cells. CONCLUSIONS These findings provide evidence for the regional anatomical organization of neuronal primary cilia in the adult brain and suggest that various brain insults may disrupt this organization.
Collapse
Affiliation(s)
- Gregory W. Kirschen
- Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY 11794, USA
- Molecular & Cellular Pharmacology Program, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Hanxiao Liu
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Tracy Lang
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA
- Simons Summer Research Program (SSRP)
| | - Xuelin Liang
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Shaoyu Ge
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
22
|
Hollingsworth E, Khouri J, Imitola J. Endogenous repair and development inspired therapy of neurodegeneration in progressive multiple sclerosis. Expert Rev Neurother 2017; 17:611-629. [DOI: 10.1080/14737175.2017.1287564] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Ethan Hollingsworth
- Laboratory for Neural Stem Cells and Functional Neurogenetics, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
- Division of Neuroimmunology and Multiple Sclerosis and Departments of Neurology and Neuroscience. The Ohio State, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
| | - Jamil Khouri
- Laboratory for Neural Stem Cells and Functional Neurogenetics, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
- Division of Neuroimmunology and Multiple Sclerosis and Departments of Neurology and Neuroscience. The Ohio State, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
| | - Jaime Imitola
- Laboratory for Neural Stem Cells and Functional Neurogenetics, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
- Division of Neuroimmunology and Multiple Sclerosis and Departments of Neurology and Neuroscience. The Ohio State, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
| |
Collapse
|
23
|
|
24
|
Angiogenic factors are associated with multiple sclerosis. J Neuroimmunol 2016; 301:88-93. [DOI: 10.1016/j.jneuroim.2016.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 12/20/2022]
|
25
|
Waller R, Woodroofe MN, Wharton SB, Ince PG, Francese S, Heath PR, Cudzich-Madry A, Thomas RH, Rounding N, Sharrack B, Simpson JE. Gene expression profiling of the astrocyte transcriptome in multiple sclerosis normal appearing white matter reveals a neuroprotective role. J Neuroimmunol 2016; 299:139-146. [DOI: 10.1016/j.jneuroim.2016.09.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 01/03/2023]
|
26
|
Huang Y, Dreyfus CF. The role of growth factors as a therapeutic approach to demyelinating disease. Exp Neurol 2016; 283:531-40. [PMID: 27016070 PMCID: PMC5010931 DOI: 10.1016/j.expneurol.2016.02.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/19/2016] [Accepted: 02/24/2016] [Indexed: 01/19/2023]
Abstract
A variety of growth factors are being explored as therapeutic agents relevant to the axonal and oligodendroglial deficits that occur as a result of demyelinating lesions such as are evident in Multiple Sclerosis (MS). This review focuses on five such proteins that are present in the lesion site and impact oligodendrocyte regeneration. It then presents approaches that are being exploited to manipulate the lesion environment affiliated with multiple neurodegenerative diseases and suggests that the utility of these approaches can extend to demyelination. Challenges are to further understand the roles of specific growth factors on a cellular and tissue level. Emerging technologies can then be employed to optimize the use of growth factors to ameliorate the deficits associated with demyelinating degenerative diseases.
Collapse
Affiliation(s)
- Yangyang Huang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ 08854, USA.
| | - Cheryl F Dreyfus
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ 08854, USA.
| |
Collapse
|
27
|
Steelman AJ, Zhou Y, Koito H, Kim S, Payne HR, Lu QR, Li J. Activation of oligodendroglial Stat3 is required for efficient remyelination. Neurobiol Dis 2016; 91:336-46. [PMID: 27060559 DOI: 10.1016/j.nbd.2016.03.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 03/11/2016] [Accepted: 03/30/2016] [Indexed: 01/26/2023] Open
Abstract
Multiple sclerosis is the most prevalent demyelinating disease of the central nervous system (CNS) and is histologically characterized by perivascular demyelination as well as neurodegeneration. While the degree of axonal damage is correlated with clinical disability, it is believed that remyelination can protect axons from degeneration and slow disease progression. Therefore, understanding the intricacies associated with myelination and remyelination may lead to therapeutics that can enhance the remyelination process and slow axon degeneration and loss of function. Ciliary neurotrophic factor (CNTF) family cytokines such as leukemia inhibitory factor (LIF) and interleukin 11 (IL-11) are known to promote oligodendrocyte maturation and remyelination in experimental models of demyelination. Because CNTF family member binding to the gp130 receptor results in activation of the JAK2/Stat3 pathway we investigated the necessity of oligodendroglial Stat3 in transducing the signal required for myelination and remyelination. We found that Stat3 activation in the CNS coincides with myelination during development. Stimulation of oligodendrocyte precursor cells (OPCs) with CNTF or LIF promoted OPC survival and final differentiation, which was completely abolished by pharmacologic blockade of Stat3 activation with JAK2 inhibitor. Similarly, genetic ablation of Stat3 in oligodendrocyte lineage cells prevented CNTF-induced OPC differentiation in culture. In vivo, while oligodendroglial Stat3 signaling appears to be dispensable for developmental CNS myelination, it is required for oligodendrocyte regeneration and efficient remyelination after toxin-induced focal demyelination in the adult brain. Our data suggest a critical function for oligodendroglial Stat3 signaling in myelin repair.
Collapse
Affiliation(s)
- Andrew J Steelman
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, United States; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, United States
| | - Yun Zhou
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, United States
| | - Hisami Koito
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, United States
| | - SunJa Kim
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, United States; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, United States
| | - H Ross Payne
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, United States
| | - Q Richard Lu
- Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, United States
| | - Jianrong Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, United States; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
28
|
Zeis T, Allaman I, Gentner M, Schroder K, Tschopp J, Magistretti PJ, Schaeren-Wiemers N. Metabolic gene expression changes in astrocytes in Multiple Sclerosis cerebral cortex are indicative of immune-mediated signaling. Brain Behav Immun 2015; 48:313-25. [PMID: 25937052 DOI: 10.1016/j.bbi.2015.04.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/10/2015] [Accepted: 04/11/2015] [Indexed: 01/22/2023] Open
Abstract
Emerging as an important correlate of neurological dysfunction in Multiple Sclerosis (MS), extended focal and diffuse gray matter abnormalities have been found and linked to clinical manifestations such as seizures, fatigue and cognitive dysfunction. To investigate possible underlying mechanisms we analyzed the molecular alterations in histopathological normal appearing cortical gray matter (NAGM) in MS. By performing a differential gene expression analysis of NAGM of control and MS cases we identified reduced transcription of astrocyte specific genes involved in the astrocyte-neuron lactate shuttle (ANLS) and the glutamate-glutamine cycle (GGC). Additional quantitative immunohistochemical analysis demonstrating a CX43 loss in MS NAGM confirmed a crucial involvement of astrocytes and emphasizes their importance in MS pathogenesis. Concurrently, a Toll-like/IL-1β signaling expression signature was detected in MS NAGM, indicating that immune-related signaling might be responsible for the downregulation of ANLS and GGC gene expression in MS NAGM. Indeed, challenging astrocytes with immune stimuli such as IL-1β and LPS reduced their ANLS and GGC gene expression in vitro. The detected upregulation of IL1B in MS NAGM suggests inflammasome priming. For this reason, astrocyte cultures were treated with ATP and ATP/LPS as for inflammasome activation. This treatment led to a reduction of ANLS and GGC gene expression in a comparable manner. To investigate potential sources for ANLS and GGC downregulation in MS NAGM, we first performed an adjuvant-driven stimulation of the peripheral immune system in C57Bl/6 mice in vivo. This led to similar gene expression changes in spinal cord demonstrating that peripheral immune signals might be one source for astrocytic gene expression changes in the brain. IL1B upregulation in MS NAGM itself points to a possible endogenous signaling process leading to ANLS and GGC downregulation. This is supported by our findings that, among others, MS NAGM astrocytes express inflammasome components and that astrocytes are capable to release Il-1β in-vitro. Altogether, our data suggests that immune signaling of immune- and/or central nervous system origin drives alterations in astrocytic ANLS and GGC gene regulation in the MS NAGM. Such a mechanism might underlie cortical brain dysfunctions frequently encountered in MS patients.
Collapse
Affiliation(s)
- T Zeis
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - I Allaman
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - M Gentner
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - K Schroder
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland; Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
| | - J Tschopp
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - P J Magistretti
- Division of Biological and Environmental Sciences and Engineering, KAUST, Thuwal, Saudi Arabia; Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland; Centre de Neurosciences Psychiatriques, CHUV, Département de Psychiatrie, Site de Cery, CH-1008 Prilly/Lausanne, Switzerland
| | - N Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| |
Collapse
|
29
|
Mallucci G, Peruzzotti-Jametti L, Bernstock JD, Pluchino S. The role of immune cells, glia and neurons in white and gray matter pathology in multiple sclerosis. Prog Neurobiol 2015; 127-128:1-22. [PMID: 25802011 PMCID: PMC4578232 DOI: 10.1016/j.pneurobio.2015.02.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/24/2014] [Accepted: 02/27/2015] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis is one of the most common causes of chronic neurological disability beginning in early to middle adult life. Multiple sclerosis is idiopathic in nature, yet increasing correlative evidence supports a strong association between one's genetic predisposition, the environment and the immune system. Symptoms of multiple sclerosis have primarily been shown to result from a disruption in the integrity of myelinated tracts within the white matter of the central nervous system. However, recent research has also highlighted the hitherto underappreciated involvement of gray matter in multiple sclerosis disease pathophysiology, which may be especially relevant when considering the accumulation of irreversible damage and progressive disability. This review aims at providing a comprehensive overview of the interplay between inflammation, glial/neuronal damage and regeneration throughout the course of multiple sclerosis via the analysis of both white and gray matter lesional pathology. Further, we describe the common pathological mechanisms underlying both relapsing and progressive forms of multiple sclerosis, and analyze how current (as well as future) treatments may interact and/or interfere with its pathology. Understanding the putative mechanisms that drive disease pathogenesis will be key in helping to develop effective therapeutic strategies to prevent, mitigate, and treat the diverse morbidities associated with multiple sclerosis.
Collapse
Affiliation(s)
- Giulia Mallucci
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
- Department of Brain and Behavioural Sciences, National Neurological Institute C. Mondino, University of Pavia, 27100 Pavia, Italy
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
| | - Joshua D. Bernstock
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bldg10/Rm5B06, MSC 1401, 10 Center Drive, Bethesda, MD 20892, USA
| | - Stefano Pluchino
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
| |
Collapse
|
30
|
Leukemia inhibitory factor tips the immune balance towards regulatory T cells in multiple sclerosis. Brain Behav Immun 2015; 45:180-8. [PMID: 25514345 DOI: 10.1016/j.bbi.2014.11.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/08/2014] [Accepted: 11/20/2014] [Indexed: 12/21/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS), for which current treatments are unable to prevent disease progression. Based on its neuroprotective and neuroregenerating properties, leukemia inhibitory factor (LIF), a member of the interleukin-6 (IL-6) cytokine family, is proposed as a novel candidate for MS therapy. However, its effect on the autoimmune response remains unclear. In this study, we determined how LIF modulates T cell responses that play a crucial role in the pathogenesis of MS. We demonstrate that expression of the LIF receptor was strongly increased on immune cells of MS patients. LIF treatment potently boosted the number of regulatory T cells (Tregs) in CD4(+) T cells isolated from healthy controls and MS patients with low serum levels of IL-6. Moreover, IL-6 signaling was reduced in the donors that responded to LIF treatment in vitro. Our data together with previous findings revealing that IL-6 inhibits Treg development, suggest an opposing function of LIF and IL-6. In a preclinical animal model of MS we shifted the LIF/IL-6 balance in favor of LIF by CNS-targeted overexpression. This increased the number of Tregs in the CNS during active autoimmune responses and reduced disease symptoms. In conclusion, our data show that LIF downregulates the autoimmune response by enhancing Treg numbers, providing further impetus for the use of LIF as a novel treatment for MS and other autoimmune diseases.
Collapse
|
31
|
Itkonen JM, Urtti A, Bird LE, Sarkhel S. Codon optimization and factorial screening for enhanced soluble expression of human ciliary neurotrophic factor in Escherichia coli. BMC Biotechnol 2014; 14:92. [PMID: 25394427 PMCID: PMC4237735 DOI: 10.1186/s12896-014-0092-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 10/21/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurotrophic factors influence survival, differentiation, proliferation and death of neuronal cells within the central nervous system. Human ciliary neurotrophic factor (hCNTF) has neuroprotective properties and is also known to influence energy balance. Consequently, hCNTF has potential therapeutic applications in neurodegenerative, obesity and diabetes related disorders. Clinical and biological applications of hCNTF necessitate a recombinant expression system to produce large amounts of functional protein in soluble form. Earlier attempts to express hCNTF in Escherichia coli (E. coli) were limited by low amounts and the need to refold from inclusion bodies. RESULTS In this report, we describe a strategy to effectively identify constructs and conditions for soluble expression of hCNTF in E. coli. Small-scale expression screening with soluble fusion tags identified many conditions that yielded soluble expression. Codon optimized 6-His-hCNTF construct showed soluble expression in all the conditions tested. Large-scale culture of the 6-His-hCNTF construct yielded high (10 - 20 fold) soluble expression (8 - 9 fold) as compared to earlier published reports. Functional activity of recombinant 6-His-hCNTF produced was confirmed by its binding to hCNTF receptor (hCNTFRα) with an EC50 = 36 nM. CONCLUSION Our results highlight the combination of codon optimization and screening soluble fusion tags as a successful strategy for high yielding soluble expression of hCNTF in E. coli. Codon optimization of the hCNTF sequence seems to be sufficient for soluble expression of hCNTF. The combined approach of codon optimization and soluble fusion tag screen can be an effective strategy for soluble expression of pharmaceutical proteins in E. coli.
Collapse
|
32
|
Jonas A, Thiem S, Kuhlmann T, Wagener R, Aszodi A, Nowell C, Hagemeier K, Laverick L, Perreau V, Jokubaitis V, Emery B, Kilpatrick T, Butzkueven H, Gresle M. Axonally derived matrilin-2 induces proinflammatory responses that exacerbate autoimmune neuroinflammation. J Clin Invest 2014; 124:5042-56. [PMID: 25329699 DOI: 10.1172/jci71385] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/11/2014] [Indexed: 01/14/2023] Open
Abstract
In patients with multiple sclerosis (MS) and mice with experimental autoimmune encephalomyelitis (EAE), inflammatory axonal injury is a major determinant of disability; however, the drivers of this injury are incompletely understood. Here, we used the EAE model and determined that the extracellular matrix protein matrilin-2 (MATN2) is an endogenous neuronal molecule that is regulated in association with inflammatory axonal injury. Compared with WT mice, mice harboring a deletion of Matn2 exhibited reduced disease severity and axon damage following induction of EAE. Evaluation of neuron-macrophage cocultures revealed that exogenous MATN2 specifically signals through TLR4 and directly induces expression of proinflammatory genes in macrophages, promoting axonal damage. Moreover, the MATN2-induced proinflammatory response was attenuated greatly in macrophages from Myd88 KO mice. Examination of brain sections from patients with MS revealed that MATN2 is expressed in lesions but not in normal-appearing white matter. Together, our results indicate that MATN2 is a deleterious endogenous neuroaxonal injury response signal that activates innate immune cells and could contribute to early axonal damage in CNS inflammatory diseases like MS.
Collapse
|
33
|
Dutta R. Gene expression changes underlying cortical pathology: clues to understanding neurological disability in multiple sclerosis. Mult Scler 2014; 19:1249-54. [PMID: 23996595 DOI: 10.1177/1352458513500554] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system with an unknown etiology. The clinical disease course is variable, with the majority of patients experiencing reversible episodes of neurological disability in the third or fourth decade of life, eventually followed by a state of irreversible progression. Continuous axonal and neuronal loss is thought to be the major cause of this progression. Over the last decade, extensive research has targeted the gray matter and its role in MS pathogenesis. While pathological and imaging studies have begun to reveal important clues about the role of cortical pathology, gene expression studies in MS cortex are still emerging. Microarray-based comparative gene expression profiling provides a snapshot of genes underlying a particular condition and has been performed using brain tissues from patients with progressive MS. In this review, we summarize existing data from gene expression changes in cortical tissues from MS brains and how they may provide clues to the pathogenesis.
Collapse
Affiliation(s)
- Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW This review will explore two new aspects of the involvement of viruses in multiple sclerosis pathogenesis. The first aspect is the complex interactions between viruses. The second aspect is the proposal of a mechanism by which autoreactive T cells are able to escape thymic selection and potentially recognize self and a pathogen. RECENT FINDINGS With regard to viruses, recent work has demonstrated that one virus may enhance the replication of another virus, potentially leading to an increase in inflammation and disease progression. Also, interactions between human endogenous retroviruses, which likely do not replicate, and certain herpes viruses, may also play a role in disease pathogenesis. Mechanistically, T cells expressing dual T-cell receptors would be able to recognize self and a foreign antigen specifically. Therefore, human endogenous retroviruses potentially play a role in multiple sclerosis pathogenesis, and both interactions between multiple viruses and autoreactive CD8(+) T cells with dual T-cell receptors may play a role in the pathogenesis of the disease. SUMMARY The complex interactions between multiple viral infections, either within the central nervous system or in the periphery, and the host immune response to viral infection may be such that a variety of viral specificities result in the activation of T cells that recognize self and induce multiple sclerosis. Therefore, it is unlikely that any one microbe will be determined to be the causative agent of multiple sclerosis as reflected by the number of potential triggering mechanisms of the disease.
Collapse
|
35
|
Criste G, Trapp B, Dutta R. Axonal loss in multiple sclerosis: causes and mechanisms. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:101-13. [PMID: 24507515 DOI: 10.1016/b978-0-444-52001-2.00005-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system and the leading cause of non-traumatic neurologic disability in young adults in the United States and Europe. The disease course is variable and starts with reversible episodes of neurologic disability which transforms into continuous and irreversible neurologic decline. It is well established that loss of axons and neurons is the major cause of the progressive neurologic decline that most MS patients endure. Current hypotheses support primary inflammatory demyelination as the underlying cause of axonal loss during earlier stages in MS. The transition to progressive disease course is thought to occur when a threshold of neuronal and axonal loss is reached and the compensatory capacity of the central nervous system is surpassed. Available immunomodulatory therapies are of little benefit to MS after entering this irreversible phase of the disease. Elucidation of mechanisms that are responsible for axonal loss is therefore essential for the development of therapies directed to stop neurologic decline in MS patients. The current chapter reviews existing data on mechanisms of axonal pathology in MS.
Collapse
Affiliation(s)
- Gerson Criste
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bruce Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
36
|
Benjamins JA. Direct effects of secretory products of immune cells on neurons and glia. J Neurol Sci 2013; 333:30-6. [DOI: 10.1016/j.jns.2013.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 05/30/2013] [Accepted: 06/02/2013] [Indexed: 11/29/2022]
|
37
|
Saji E, Arakawa M, Yanagawa K, Toyoshima Y, Yokoseki A, Okamoto K, Otsuki M, Akazawa K, Kakita A, Takahashi H, Nishizawa M, Kawachi I. Cognitive impairment and cortical degeneration in neuromyelitis optica. Ann Neurol 2013; 73:65-76. [PMID: 23378324 DOI: 10.1002/ana.23721] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 07/09/2012] [Accepted: 07/30/2012] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Neuromyelitis optica spectrum disorder (NMOsd) is an inflammatory and demyelinating syndrome characterized by optic neuritis and myelitis. Several magnetization transfer magnetic resonance imaging (MRI) studies have revealed abnormalities in normal-appearing gray matter in NMOsd. The aim of this study is to elucidate the characteristics and pathogenesis of cognitive impairment and neurodegeneration in NMOsd brains. METHODS Fourteen Japanese patients with serologically verified NMOsd, 17 patients with multiple sclerosis (MS), and 37 healthy controls were assessed with the Rao's Brief Repeatable Battery of Neuropsychological Tests (BRBN). Using 128 tissue blocks from 6 other cases of NMOsd, 3 cases of MS, and 4 controls without central nervous system involvement, we performed quantitative analysis of cortical neuronal loss and layer-specific changes in NMOsd. RESULTS In BRBN assessments, 57% of NMOsd patients and 47% of MS patients had impaired performance on at least 3 cognitive tests. Cognitive impairment in NMOsd was common even in the limited form of disease, indicating that NMOsd may progress insidiously from early stages of disease. Neuropathological assessments showed neuronal loss in cortical layers II, III, and IV, with nonlytic reaction of aquaporin-4 (AQP4)-negative astrocytes in layer I, massive activated microglia in layer II, and meningeal inflammation in NMOsd brains. All NMO cases showed no evidence of cortical demyelination. INTERPRETATION We demonstrate cognitive impairment and substantial cortical neuronal loss with unique AQP4 dynamics in astrocytes in NMOsd. These data indicate pathological processes consisting not only of inflammatory demyelinating events characterized by pattern-specific loss of AQP4 immunoreactivity but also cortical neurodegeneration in NMOsd brains.
Collapse
Affiliation(s)
- Etsuji Saji
- Department of Neurology, Niigata University, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Vernerey J, Macchi M, Magalon K, Cayre M, Durbec P. Ciliary neurotrophic factor controls progenitor migration during remyelination in the adult rodent brain. J Neurosci 2013; 33:3240-50. [PMID: 23407977 PMCID: PMC6619230 DOI: 10.1523/jneurosci.2579-12.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 12/03/2012] [Accepted: 12/16/2012] [Indexed: 11/21/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) has been shown to be expressed after brain lesions and in particular after demyelination. Here, we addressed the role of this cytokine in the regulation of neural progenitor migration in the adult rodent brain. Using an acute model of demyelination, we show that CNTF is strongly re-expressed after lesion and is involved in the postlesional mobilization of endogenous progenitors that participate in the myelin regenerative process. We show that CNTF controls the migration of subventricular zone (SVZ)-derived neural progenitors toward the demyelinated corpus callosum. Furthermore, an ectopic source of CNTF in adult healthy brains changes SVZ-derived neural progenitors' migratory behavior that migrate toward the source by activation of the Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway. Using various in vitro assays (Boyden chambers, explants, and video time-lapse imaging), we demonstrate that CNTF controls the directed migration of SVZ-derived progenitors and oligodendrocyte precursors. Altogether, these results demonstrate that in addition to its neuroprotective activity and its role in progenitor survival and maturation, CNTF acts as a chemoattractant and participates in the recruitment of endogenous progenitors during myelin repair.
Collapse
Affiliation(s)
- Julien Vernerey
- Aix-Marseille Université, and
- Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille de Luminy, Unité Mixte de Recherche 7288, 13288 Marseille, France
| | - Magali Macchi
- Aix-Marseille Université, and
- Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille de Luminy, Unité Mixte de Recherche 7288, 13288 Marseille, France
| | - Karine Magalon
- Aix-Marseille Université, and
- Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille de Luminy, Unité Mixte de Recherche 7288, 13288 Marseille, France
| | - Myriam Cayre
- Aix-Marseille Université, and
- Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille de Luminy, Unité Mixte de Recherche 7288, 13288 Marseille, France
| | - Pascale Durbec
- Aix-Marseille Université, and
- Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille de Luminy, Unité Mixte de Recherche 7288, 13288 Marseille, France
| |
Collapse
|
39
|
Age-dependent modulation of cortical transcriptomes in spinal cord injury and repair. PLoS One 2012; 7:e49812. [PMID: 23236355 PMCID: PMC3517588 DOI: 10.1371/journal.pone.0049812] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 10/16/2012] [Indexed: 11/19/2022] Open
Abstract
Both injury and aging of the central nervous system reportedly produce profound changes in gene expression. Therefore, aging may interfere with the success of therapeutic interventions which were tailored for young patients. Using genome-scale transcriptional profiling, we identified distinct age-dependent expression profiles in rat sensorimotor cortex during acute, subacute and chronic phases of spinal cord injury (SCI). Aging affects the cortical transcriptomes triggered by transection of the corticospinal tract as there was only a small overlap between the significantly lesion-regulated genes in both age groups. Over-representation analysis of the lesion-regulated genes revealed that, in addition to biological processes in common, such as lipid metabolism, others, such as activation of complement cascade, were specific for aged animals. When a recently developed treatment to suppress fibrotic scarring (anti-scarring treatment AST) was applied to the injured spinal cord of aged (22 months) and young (2 months) rats, we found that the cortical gene expression in old rats was modulated to resemble regeneration-associated profiles of young animals including the up-regulation of known repair promoting growth and transcription factors at 35 dpo. In combination with recent immunohistochemical findings demonstrating regenerative axon growth upon AST in aged animals, the present investigation on the level of gene expression strongly supports the feasibility of a successful AST therapy in elderly patients.
Collapse
|
40
|
Ng JMJ, Chen MJ, Leung JYK, Peng ZF, Manikandan J, Qi RZ, Chuah MI, West AK, Vickers JC, Lu J, Cheung NS, Chung RS. Transcriptional insights on the regenerative mechanics of axotomized neurons in vitro. J Cell Mol Med 2012; 16:789-811. [PMID: 21711447 PMCID: PMC3822849 DOI: 10.1111/j.1582-4934.2011.01361.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Axotomized neurons have the innate ability to undergo regenerative sprouting but this is often impeded by the inhibitory central nervous system environment. To gain mechanistic insights into the key molecular determinates that specifically underlie neuronal regeneration at a transcriptomic level, we have undertaken a DNA microarray study on mature cortical neuronal clusters maintained in vitro at 8, 15, 24 and 48 hrs following complete axonal severance. A total of 305 genes, each with a minimum fold change of ±1.5 for at least one out of the four time points and which achieved statistical significance (one-way ANOVA, P < 0.05), were identified by DAVID and classified into 14 different functional clusters according to Gene Ontology. From our data, we conclude that post-injury regenerative sprouting is an intricate process that requires two distinct pathways. Firstly, it involves restructuring of the neurite cytoskeleton, determined by compound actin and microtubule dynamics, protein trafficking and concomitant modulation of both guidance cues and neurotrophic factors. Secondly, it elicits a cell survival response whereby genes are regulated to protect against oxidative stress, inflammation and cellular ion imbalance. Our data reveal that neurons have the capability to fight insults by elevating biological antioxidants, regulating secondary messengers, suppressing apoptotic genes, controlling ion-associated processes and by expressing cell cycle proteins that, in the context of neuronal injury, could potentially have functions outside their normal role in cell division. Overall, vigilant control of cell survival responses against pernicious secondary processes is vital to avoid cell death and ensure successful neurite regeneration.
Collapse
Affiliation(s)
- Jian Ming Jeremy Ng
- Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Gresle MM, Alexandrou E, Wu Q, Egan G, Jokubaitis V, Ayers M, Jonas A, Doherty W, Friedhuber A, Shaw G, Sendtner M, Emery B, Kilpatrick T, Butzkueven H. Leukemia inhibitory factor protects axons in experimental autoimmune encephalomyelitis via an oligodendrocyte-independent mechanism. PLoS One 2012; 7:e47379. [PMID: 23077604 PMCID: PMC3471848 DOI: 10.1371/journal.pone.0047379] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 09/12/2012] [Indexed: 01/13/2023] Open
Abstract
Leukemia inhibitory factor (LIF) and Ciliary Neurotrophic factor (CNTF) are members of the interleukin-6 family of cytokines, defined by use of the gp130 molecule as an obligate receptor. In the murine experimental autoimmune encephalomyelitis (EAE) model, antagonism of LIF and genetic deletion of CNTF worsen disease. The potential mechanism of action of these cytokines in EAE is complex, as gp130 is expressed by all neural cells, and could involve immuno-modulation, reduction of oligodendrocyte injury, neuronal protection, or a combination of these actions. In this study we aim to investigate whether the beneficial effects of CNTF/LIF signalling in EAE are associated with axonal protection; and whether this requires signalling through oligodendrocytes. We induced MOG35–55 EAE in CNTF, LIF and double knockout mice. On a CNTF null background, LIF knockout was associated with increased EAE severity (EAE grade 2.1±0.14 vs 2.6±0.19; P<0.05). These mice also showed increased axonal damage relative to LIF heterozygous mice, as indicated by decreased optic nerve parallel diffusivity on MRI (1540±207 µm2−/s vs 1310±175 µm2−/s; P<0.05), and optic nerve (−12.5%) and spinal cord (−16%) axon densities; and increased serum neurofilament-H levels (2.5 fold increase). No differences in inflammatory cell numbers or peripheral auto-immune T-cell priming were evident. Oligodendrocyte-targeted gp130 knockout mice showed that disruption of CNTF/LIF signalling in these cells has no effect on acute EAE severity. These studies demonstrate that endogenous CNTF and LIF act centrally to protect axons from acute inflammatory destruction via an oligodendrocyte-independent mechanism.
Collapse
Affiliation(s)
- Melissa M Gresle
- Department of Medicine, The Melbourne Brain Centre at the Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Common transcriptional signatures in brain tissue from patients with HIV-associated neurocognitive disorders, Alzheimer's disease, and Multiple Sclerosis. J Neuroimmune Pharmacol 2012; 7:914-26. [PMID: 23065460 PMCID: PMC3515772 DOI: 10.1007/s11481-012-9409-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 09/26/2012] [Indexed: 01/01/2023]
Abstract
HIV-Associated Neurocognitive Disorders (HAND) is a common manifestation of HIV infection that afflicts about 50 % of HIV-positive individuals. As people with access to antiretroviral treatments live longer, HAND can be found in increasing segments of populations at risk for other chronic, neurodegenerative conditions such as Alzheimer’s disease (AD) and Multiple Sclerosis (MS). If brain diseases of diverse etiologies utilize similar biological pathways in the brain, they may coexist in a patient and possibly exacerbate neuropathogenesis and morbidity. To test this proposition, we conducted comparative meta-analysis of selected publicly available microarray datasets from brain tissues of patients with HAND, AD, and MS. In pair-wise and three-way analyses, we found a large number of dysregulated genes and biological processes common to either HAND and AD or HAND and MS, or to all three diseases. The common characteristic of all three diseases was up-regulation of broadly ranging immune responses in the brain. In addition, HAND and AD share down-modulation of processes involved, among others, in synaptic transmission and cell-cell signaling while HAND and MS share defective processes of neurogenesis and calcium/calmodulin-dependent protein kinase activity. Our approach could provide insight into the identification of common disease mechanisms and better intervention strategies for complex neurocognitive disorders.
Collapse
|
43
|
Aung LL, Brooks A, Greenberg SA, Rosenberg ML, Dhib-Jalbut S, Balashov KE. Multiple sclerosis-linked and interferon-beta-regulated gene expression in plasmacytoid dendritic cells. J Neuroimmunol 2012; 250:99-105. [PMID: 22688425 DOI: 10.1016/j.jneuroim.2012.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/14/2012] [Accepted: 05/18/2012] [Indexed: 01/05/2023]
Abstract
The cause of multiple sclerosis (MS) is not known and the mechanism of interferon-beta, a disease-modifying treatment, is not well-understood. We studied gene expression in plasmacytoid dendritic cells (pDCs), antigen-presenting cells implicated in MS pathogenesis. PDCs were separated from healthy donors and MS patients at two time points: before and after initiation of treatment with interferon-beta. Expression of selected MS-linked and interferon-beta-regulated genes was validated with single assays. We have identified 60 genes which were abnormally expressed in MS patients and were corrected after treatment. These genes could be studied as potential MS biomarkers and possible therapeutic targets in MS.
Collapse
Affiliation(s)
- Latt Latt Aung
- Department of Neurology, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | | | | | | | | | | |
Collapse
|
44
|
Dutta R, Trapp BD. Gene expression profiling in multiple sclerosis brain. Neurobiol Dis 2012; 45:108-14. [PMID: 21147224 PMCID: PMC3066282 DOI: 10.1016/j.nbd.2010.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 11/29/2010] [Accepted: 12/02/2010] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system and the leading cause of non-traumatic neurological disability in young adults in the United States and Europe. The clinical disease course is variable and starts with reversible episodes of neurological disability in the third or fourth decade of life. Microarray-based comparative gene profiling provides a snapshot of genes underlying a particular condition. Several large scale microarray studies have been conducted using brain tissue from MS patients. In this review, we summarize existing data from different gene expression profiling studies and how they relate to understand the pathogenesis of MS.
Collapse
Affiliation(s)
- Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
45
|
Cytokines regulate neuronal gene expression: Differential effects of Th1, Th2 and monocyte/macrophage cytokines. J Neuroimmunol 2011; 238:19-33. [DOI: 10.1016/j.jneuroim.2011.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 06/16/2011] [Accepted: 06/17/2011] [Indexed: 12/19/2022]
|
46
|
Multiple sclerosis as a neurodegenerative disease: pathology, mechanisms and therapeutic implications. Curr Opin Neurol 2011; 24:224-9. [PMID: 21455066 DOI: 10.1097/wco.0b013e328346056f] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) treatments targeting the inflammatory nature of the disease have become increasingly effective in recent years. However, our efforts at targeting the progressive disease phase have so far been largely unsuccessful. This has led to the hypothesis that disease mechanisms independent of an adaptive immune response contribute to disease progression and closely resemble neurodegeneration. RECENT FINDINGS Nonfocal, diffuse changes in the MS brain, especially axonal loss and mitochondrial dysfunction, prove better correlates of disability than total lesion load and have been associated with disease progression. Molecular changes in nondemyelinated MS tissue also suggest that alterations in the MS brain are widespread and consist of pro-inflammatory as well as anti-inflammatory responses. However, local lymphocytic inflammation and microglial activation are salient features of the chronic disease, and T-cell-mediated inflammation contributes to tissue damage. In addition, neuroaxonal cytoskeletal alterations have been associated with disease progression. SUMMARY Our knowledge of the molecular mechanisms leading to neuroaxonal damage and demise in MS is steadily increasing. Experimental therapies targeting neuroaxonal ionic imbalances and energy metabolism in part show promising results. A better understanding of the molecular mechanisms underlying chronic progression will substantially aid the development of new treatment strategies.
Collapse
|
47
|
Dutta R, Chang A, Doud MK, Kidd GJ, Ribaudo MV, Young EA, Fox RJ, Staugaitis SM, Trapp BD. Demyelination causes synaptic alterations in hippocampi from multiple sclerosis patients. Ann Neurol 2011; 69:445-54. [PMID: 21446020 DOI: 10.1002/ana.22337] [Citation(s) in RCA: 235] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the human central nervous system. Although the clinical impact of gray matter pathology in MS brains is unknown, 30 to 40% of MS patients demonstrate memory impairment. The molecular basis of this memory dysfunction has not yet been investigated in MS patients. METHODS To investigate possible mechanisms of memory impairment in MS patients, we compared morphological and molecular changes in myelinated and demyelinated hippocampi from postmortem MS brains. RESULTS Demyelinated hippocampi had minimal neuronal loss but significant decreases in synaptic density. Neuronal proteins essential for axonal transport, synaptic plasticity, glutamate neurotransmission, glutamate homeostasis, and memory/learning were significantly decreased in demyelinated hippocampi, but not in demyelinated motor cortices from MS brains. INTERPRETATION Collectively, these data support hippocampal demyelination as a cause of synaptic alterations in MS patients and establish that the neuronal genes regulated by myelination reflect specific functions of neuronal subpopulations.
Collapse
Affiliation(s)
- Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lindquist S, Hassinger S, Lindquist JA, Sailer M. The balance of pro-inflammatory and trophic factors in multiple sclerosis patients: effects of acute relapse and immunomodulatory treatment. Mult Scler 2011; 17:851-66. [DOI: 10.1177/1352458511399797] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: In multiple sclerosis inflammation is primarily injurious to the central nervous system, but its therapeutic suppression might inhibit repair-promoting factors. Objectives: We aimed at better describing the complexity of biological effects during an acute relapse and analysed the effects of intervention with high-dose i.v. glucocorticoids and immunomodulatory treatment with interferon-beta (IFNβ). Methods: We studied the intracellular expression levels of the pro-inflammatory mediators tumour necrosis factor alpha (TNFα) and inducible nitric oxide synthase (iNOS) together with the neurotrophins ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) in freshly isolated peripheral blood mononuclear cells of multiple sclerosis patients during an acute relapse, after intervention with i.v. methylprednisolone and at baseline, using a highly quantitative flow-cytometric approach. Results: We demonstrated the expression of CNTF in human leucocytes. We showed that CNTF levels differed in acutely relapsing multiple sclerosis patients compared with controls and increased after corticosteroid treatment. CNTF can counteract the toxicity of TNFα towards oligodendrocytes and we found TNFα increased during acute relapses. Following corticosteroids, neither TNFα nor iNOS expression was reduced. Levels of BDNF were not affected by glucocorticoids, but increased during IFNβ therapy. However, IFNβ also increased the expression of iNOS and major histocompatibility complex class I (MHC-I), underlining its immunomodulatory potential. Conclusions: Multiple sclerosis patients might benefit from reparative, and not solely from anti-inflammatory, effects of glucocorticoids. Interactive effects of glucocorticoid- and IFNβ-treatment need to be considered to improve neuroprotection and remyelination resulting from immunomodulatory treatment.
Collapse
Affiliation(s)
- Sabine Lindquist
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | - Sarah Hassinger
- University Clinic for Neurology, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jonathan A Lindquist
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke-University, Magdeburg, Germany
- Co-senior authors
| | - Michael Sailer
- University Clinic for Neurology, Otto-von-Guericke-University, Magdeburg, Germany
- Centre for Neurological Rehabilitation, Magdeburg, Germany
- Co-senior authors
| |
Collapse
|
49
|
Dutta R, Trapp BD. Mechanisms of neuronal dysfunction and degeneration in multiple sclerosis. Prog Neurobiol 2010; 93:1-12. [PMID: 20946934 DOI: 10.1016/j.pneurobio.2010.09.005] [Citation(s) in RCA: 318] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 09/14/2010] [Accepted: 09/30/2010] [Indexed: 01/18/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system. Due to its high prevalence, MS is the leading cause of non-traumatic neurological disability in young adults in the United States and Europe. The clinical disease course is variable and starts with reversible episodes of neurological disability in the third or fourth decade of life. This transforms into a disease of continuous and irreversible neurological decline by the sixth or seventh decade. Available therapies for MS patients have little benefit for patients who enter this irreversible phase of the disease. It is well established that irreversible loss of axons and neurons are the major cause of the irreversible and progressive neurological decline that most MS patients endure. This review discusses the etiology, mechanisms and progress made in determining the cause of axonal and neuronal loss in MS.
Collapse
Affiliation(s)
- Ranjan Dutta
- Department of Neurosciences/NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | |
Collapse
|
50
|
Sättler MB, Bähr M. Future neuroprotective strategies. Exp Neurol 2010; 225:40-7. [DOI: 10.1016/j.expneurol.2009.08.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 08/18/2009] [Accepted: 08/19/2009] [Indexed: 12/27/2022]
|