1
|
Vargas JA, Finnemann SC. Probing Photoreceptor Outer Segment Phagocytosis by the RPE In Vivo: Models and Methodologies. Int J Mol Sci 2022; 23:ijms23073661. [PMID: 35409021 PMCID: PMC8998817 DOI: 10.3390/ijms23073661] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 01/27/2023] Open
Abstract
In the vertebrate retina, the light-sensitive photoreceptor rods and cones constantly undergo renewal by generating new portions of the outer segment and shedding their distal, spent tips. The neighboring RPE provides the critical function of engulfing the spent material by phagocytosis. RPE phagocytosis of shed rod outer segment fragments is a circadian process that occurs in a burst of activity shortly after daily light onset with low activity at other times, a rhythm that has been reported for many species and over 50 years. In this review, we compare studies on the rhythm and quantity of RPE phagocytosis using different in vivo model systems and assessment methods. We discuss how measurement methodology impacts the observation and analysis of RPE phagocytosis. Published studies on RPE phagocytosis investigating mice further suggest that differences in genetic background and housing conditions may affect results. Altogether, a comparison between RPE phagocytosis studies performed using differing methodology and strains of the same species is not as straightforward as previously thought.
Collapse
|
2
|
Salvador AFM, Kipnis J. Immune response after central nervous system injury. Semin Immunol 2022; 59:101629. [PMID: 35753867 DOI: 10.1016/j.smim.2022.101629] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Traumatic injuries of the central nervous system (CNS) affect millions of people worldwide, and they can lead to severely damaging consequences such as permanent disability and paralysis. Multiple factors can obstruct recovery after CNS injury. One of the most significant is the progressive neuronal death that follows the initial mechanical impact, leading to the loss of undamaged cells via a process termed secondary neurodegeneration. Efforts to define treatments that limit the spread of damage, while important, have been largely ineffectual owing to gaps in the mechanistic understanding that underlies the persisting neuronal cell death. Inflammation, with its influx of immune cells that occurs shortly after injury, has been associated with secondary neurodegeneration. However, the role of the immune system after CNS injury is far more complex. Studies have indicated that the immune response after CNS injury is detrimental, owing to immune cell-produced factors (e.g., pro-inflammatory cytokines, free radicals, neurotoxic glutamate) that worsen tissue damage. Our lab and others have also demonstrated the beneficial immune response that occurs after CNS injury, with the release of growth factors such as brain-derived growth factor (BDNF) and interleukin (IL-10) and the clearance of apoptotic and myelin debris by immune cells1-4. In this review, we first discuss the multifaceted roles of the immune system after CNS injury. We then speculate on how advancements in single-cell RNA technologies can dramatically change our understanding of the immune response, how the spinal cord meninges serve as an important site for hosting immunological processes critical for recovery, and how the origin of peripherally recruited immune cells impacts their function in the injured CNS.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
3
|
Blum K, Modestino EJ, Baron D, Brewer R, Thanos P, Elman I, Badgaiyan RD, Downs BW, Bagchi D, McLaughlin T, Bowirrat A, Roy AK, Gold MS. Endorphinergic Enhancement Attenuation of Post-traumatic Stress Disorder (PTSD) via Activation of Neuro-immunological Function in the Face of a Viral Pandemic. ACTA ACUST UNITED AC 2021; 10:86-97. [PMID: 34466374 DOI: 10.2174/2211556009999210104221215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Introduction Polymorphic gene variants, particularly the genetic determinants of low dopamine function (hypodopaminergia), are known to associate with Substance Use Disorder (SUD) and a predisposition to PTSD. Addiction research and molecular genetic applied technologies supported by the National Institutes of Health (NIH) have revealed the complex functions of brain reward circuitry and its crucial role in addiction and PTSD symptomatology. Discussion It is noteworthy that Israeli researchers compared mice with a normal immune system with mice lacking adaptive immunity and found that the incidence of PTSD increased several-fold. It is well established that raising endorphinergic function increases immune response significantly. Along these lines, Blum's work has shown that D-Phenylalanine (DPA), an enkephalinase inhibitor, increases brain endorphins in animal models and reduces stress in humans. Enkephalinase inhibition with DPA treats Post Traumatic Stress Disorder (PTSD) by restoring endorphin function. The Genetic Addiction Risk Severity (GARS) can characterize relevant phenotypes, genetic risk for stress vulnerability vs. resilience. GARS could be used to pre-test military enlistees for adaptive immunity or as part of PTSD management with customized neuronutrient supplementation upon return from deployment. Conclusion Based on GARS values, with particular emphasis on enhancing immunological function, pro-dopamine regulation may restore dopamine homeostasis. Recognition of the immune system as a "sixth sense" and assisting adaptive immunity with Precision Behavioral Management (PBM), accompanied by other supportive interventions and therapies, may shift the paradigm in treating stress disorders.
Collapse
Affiliation(s)
- Kenneth Blum
- Western University Health Sciences, Graduate School of Biomedical Sciences, Pomona, CA, USA.,Institute of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary.,Division of Precision Behavioral Management, Geneus Health, San Antonio, TX, USA.,Division of Nutrigenomics, Victory Nutrition International, Lederoch, PA., USA
| | | | - David Baron
- Western University Health Sciences, Graduate School of Biomedical Sciences, Pomona, CA, USA
| | - Raymond Brewer
- Division of Precision Behavioral Management, Geneus Health, San Antonio, TX, USA
| | - Panayotis Thanos
- Behavioral Neuropharmacology & Neuroimaging Laboratory on Addiction, Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY, USA
| | - Igor Elman
- Department of Psychiatry, Harvard School of Medicine, Cambridge MA, USA
| | - Rajendra D Badgaiyan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy I Memorial VA Hospital, San Antonio, TX. and Long School of Medicine, University of Texas Medical Center, San Antonio TX, USA
| | - B William Downs
- Division of Nutrigenomics, Victory Nutrition International, Lederoch, PA., USA
| | - Debasis Bagchi
- Division of Nutrigenomics, Victory Nutrition International, Lederoch, PA., USA.,Department of Pharmaceutical Sciences, University of Houston, School of Pharmacy, Houston, TX., USA
| | | | - Abdalla Bowirrat
- Department of Neuroscience and Genetics, Interdisciplinary Center Herzliya, Herzliya, Israel
| | - A Kenison Roy
- Department of Psychiatry, University of Tulane School of Medicine, New Orleans, LA, USA
| | - Mark S Gold
- Department of Psychiatry, Washington University, School of Medicine, St. Louis, MO., USA
| |
Collapse
|
4
|
Gillespie ER, Ruitenberg MJ. Neuroinflammation after SCI: Current Insights and Therapeutic Potential of Intravenous Immunoglobulin. J Neurotrauma 2020; 39:320-332. [PMID: 32689880 DOI: 10.1089/neu.2019.6952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Traumatic spinal cord injury (SCI) elicits a complex cascade of cellular and molecular inflammatory events. Although certain aspects of the inflammatory response are essential to wound healing and repair, post-SCI inflammation is, on balance, thought to be detrimental to recovery by causing "bystander damage" and the spread of pathology into spared but vulnerable regions of the spinal cord. Much of the research to date has therefore focused on understanding the inflammatory drivers of secondary tissue loss after SCI, to define therapeutic targets and positively modulate this response. Numerous experimental studies have demonstrated that modulation of the inflammatory response to SCI can indeed lead to significant neuroprotection and improved recovery. However, it is now also recognized that broadscale immunosuppression is not necessarily beneficial and may even carry the risk of contributing to the development of serious adverse events. Immune modulation rather than suppression is therefore now considered a more promising approach to target harmful post-traumatic inflammation following a major neurotraumatic event such as SCI. One promising immunomodulatory agent is intravenous immunoglobulin (IVIG), a plasma product that contains mostly immunoglobulin G (IgG) from thousands of healthy donors. IVIG is currently already widely used to treat a range of autoimmune diseases, but recent studies have found that it also holds great promise for treating acute neurological conditions, including SCI. This review provides an overview of the inflammatory response to SCI, immunomodulatory approaches that are currently in clinical trials, proposed mechanisms of action for IVIG therapy, and the putative relevance of these in the context of neurotraumatic events.
Collapse
Affiliation(s)
- Ellen R Gillespie
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Trauma, Critical Care, and Recovery, Brisbane Diamantina Health Partners, Brisbane, Australia
| |
Collapse
|
5
|
Alves de Lima K, Rustenhoven J, Kipnis J. Meningeal Immunity and Its Function in Maintenance of the Central Nervous System in Health and Disease. Annu Rev Immunol 2020; 38:597-620. [DOI: 10.1146/annurev-immunol-102319-103410] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuroimmunology, albeit a relatively established discipline, has recently sparked numerous exciting findings on microglia, the resident macrophages of the central nervous system (CNS). This review addresses meningeal immunity, a less-studied aspect of neuroimmune interactions. The meninges, a triple layer of membranes—the pia mater, arachnoid mater, and dura mater—surround the CNS, encompassing the cerebrospinal fluid produced by the choroid plexus epithelium. Unlike the adjacent brain parenchyma, the meninges contain a wide repertoire of immune cells. These constitute meningeal immunity, which is primarily concerned with immune surveillance of the CNS, and—according to recent evidence—also participates in postinjury CNS recovery, chronic neurodegenerative conditions, and even higher brain function. Meningeal immunity has recently come under the spotlight owing to the characterization of meningeal lymphatic vessels draining the CNS. Here, we review the current state of our understanding of meningeal immunity and its effects on healthy and diseased brains.
Collapse
Affiliation(s)
- Kalil Alves de Lima
- Center for Brain Immunology and Glia (BIG) and Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA;,
| | - Justin Rustenhoven
- Center for Brain Immunology and Glia (BIG) and Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA;,
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG) and Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA;,
| |
Collapse
|
6
|
Sabin KZ, Echeverri K. The role of the immune system during regeneration of the central nervous system. ACTA ACUST UNITED AC 2019; 7. [PMID: 32864529 DOI: 10.1016/j.regen.2019.100023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Central nervous system damage in mammals leads to neuronal cell death, axonal degeneration, and formation of a glial scar resulting in functional and behavioral defects. Other vertebrates, like fish and salamanders, have retained the ability to functionally regenerate after central nervous system injury. To date research from many research organisms has led to a more concise understanding of the response of local neural cells to injury. However, it has become clear that non-neural cells of the immune system play an important role in determining the tissue response to injury. In this review we briefly consider the mammalian response to injury compared to organisms with the natural ability to regenerate. We then discuss similarities and differences in how cells of the innate and adaptive immune system respond and contribute to tissue repair in various species.
Collapse
Affiliation(s)
- K Z Sabin
- Eugene Bell Center for Regenerative Biology & Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543 USA
| | - K Echeverri
- Eugene Bell Center for Regenerative Biology & Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543 USA
| |
Collapse
|
7
|
The immunological response to traumatic brain injury. J Neuroimmunol 2019; 332:112-125. [DOI: 10.1016/j.jneuroim.2019.04.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/30/2022]
|
8
|
Lu XM, Mao M, Xiao L, Yu Y, He M, Zhao GY, Tang JJ, Feng S, Li S, He CM, Wang YT. Nucleic Acid Vaccine Targeting Nogo-66 Receptor and Paired Immunoglobulin-Like Receptor B as an Immunotherapy Strategy for Spinal Cord Injury in Rats. Neurotherapeutics 2019; 16:381-393. [PMID: 30843154 PMCID: PMC6554366 DOI: 10.1007/s13311-019-00718-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Nogo-66 receptor (NgR) and paired immunoglobulin-like receptor B (PirB) are two common receptors of various myelin-associated inhibitors (MAIs) and, thus, play an important role in MAIs-induced inhibitory signalling of regeneration following spinal cord injury (SCI). Based on the concept of protective autoimmunity, vaccine approaches could induce the production of antibodies against inhibitors in myelin, such as using purified myelin, spinal cord homogenates, or MAIs receptor NgR, in order to block the inhibitory effects and promote functional recovery in SCI models. However, due to the complication of the molecules and the mechanisms involved in MAIs-mediated inhibitory signalling, these immunotherapy strategies have yielded inconsistent outcomes. Therefore, we hypothesized that the choice and modification of self-antigens, and co-regulating multiple targets, may be more effective in repairing the injured spinal cord and improving functional recovery. In this study, NgR and PirB were selected to construct a double-targeted granulocyte-macrophage colony stimulating factor-NgR-PirB (GMCSF-NgR-PirB) nucleic acid vaccine, and investigate the efficacy of this immunotherapy in a spinal cord injury model in rats. The results showed that this vaccination could stimulate the production of antibodies against NgR and PirB, block the inhibitory effects mediated by various MAIs, and promote nerve regeneration and functional recovery after spinal cord injury. These findings suggest that nucleic acid vaccination against NgR and PirB can be a promising therapeutic strategy for SCI and other central nervous system diseases and injuries.
Collapse
Affiliation(s)
- Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Min Mao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Lan Xiao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Ying Yu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Mei He
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Guo-Yan Zhao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jun-Jie Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Shuang Feng
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Sen Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Cheng-Ming He
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yong-Tang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| |
Collapse
|
9
|
Wang X, Xuan W, Zhu ZY, Li Y, Zhu H, Zhu L, Fu DY, Yang LQ, Li PY, Yu WF. The evolving role of neuro-immune interaction in brain repair after cerebral ischemic stroke. CNS Neurosci Ther 2018; 24:1100-1114. [PMID: 30350341 DOI: 10.1111/cns.13077] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/23/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke is the world's leading cause of disability with limited brain repair treatments which effectively improve long-term neurological deficits. The neuroinflammatory responses persist into the late repair phase of stroke and participate in all brain repair elements, including neurogenesis, angiogenesis, synaptogenesis, remyelination and axonal sprouting, shedding new light on post-stroke brain recovery. Resident brain glial cells, such as astrocytes not only contribute to neuroinflammation after stroke, but also secrete a wide range of trophic factors that can promote post-stroke brain repair. Alternatively, activated microglia, monocytes, and neutrophils in the innate immune system, traditionally considered as major damaging factors after stroke, have been suggested to be extensively involved in brain repair after stroke. The adaptive immune system may also have its bright side during the late regenerative phase, affecting the immune suppressive regulatory T cells and B cells. This review summarizes the recent findings in the evolving role of neuroinflammation in multiple post-stroke brain repair mechanisms and poses unanswered questions that may generate new directions for future research and give rise to novel therapeutic targets to improve stroke recovery.
Collapse
Affiliation(s)
- Xin Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Yu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hao Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dan-Yun Fu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li-Qun Yang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pei-Ying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
10
|
Filiano AJ, Gadani SP, Kipnis J. How and why do T cells and their derived cytokines affect the injured and healthy brain? Nat Rev Neurosci 2017; 18:375-384. [PMID: 28446786 PMCID: PMC5823005 DOI: 10.1038/nrn.2017.39] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The evolution of adaptive immunity provides enhanced defence against specific pathogens, as well as homeostatic immune surveillance of all tissues. Despite being 'immune privileged', the CNS uses the assistance of the immune system in physiological and pathological states. In this Opinion article, we discuss the influence of adaptive immunity on recovery after CNS injury and on cognitive and social brain function. We further extend a hypothesis that the pro-social effects of interferon-regulated genes were initially exploited by pathogens to increase host-host transmission, and that these genes were later recycled by the host to form part of an immune defence programme. In this way, the evolution of adaptive immunity may reflect a host-pathogen 'arms race'.
Collapse
Affiliation(s)
- Anthony J Filiano
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Sachin P Gadani
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| |
Collapse
|
11
|
Abstract
Neuroimmunologists seek to understand the interactions between the central nervous system (CNS) and the immune system, both under homeostatic conditions and in diseases. Unanswered questions include those relating to the diversity and specificity of the meningeal T cell repertoire; the routes taken by immune cells that patrol the meninges under healthy conditions and invade the parenchyma during pathology; the opposing effects (beneficial or detrimental) of these cells on CNS function; the role of immune cells after CNS injury; and the evolutionary link between the two systems, resulting in their tight interaction and interdependence. This Review summarizes the current standing of and challenging questions related to interactions between adaptive immunity and the CNS and considers the possible directions in which these aspects of neuroimmunology will be heading over the next decade.
Collapse
Affiliation(s)
- Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
12
|
Martiñón S, García-Vences E, Toscano-Tejeida D, Flores-Romero A, Rodriguez-Barrera R, Ferrusquia M, Hernández-Muñoz RE, Ibarra A. Long-term production of BDNF and NT-3 induced by A91-immunization after spinal cord injury. BMC Neurosci 2016; 17:42. [PMID: 27364353 PMCID: PMC4928355 DOI: 10.1186/s12868-016-0267-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/03/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND After spinal cord (SC)-injury, a non-modulated immune response contributes to the damage of neural tissue. Protective autoimmunity (PA) is a T cell mediated, neuroprotective response induced after SC-injury. Immunization with neural-derived peptides (INDP), such as A91, has shown to promote-in vitro-the production of neurotrophic factors. However, the production of these molecules has not been studied at the site of injury. RESULTS In order to evaluate these issues, we performed four experiments in adult female Sprague-Dawley rats. In the first one, brain derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) concentrations were evaluated at the site of lesion 21 days after SC-injury. BDNF and NT-3 were significantly increased in INDP-treated animals. In the second experiment, proliferation of anti-A91 T cells was assessed at chronic stages of injury. In this case, we found a significant proliferation of these cells in animals subjected to SC-injury + INDP. In the third experiment, we explored the amount of BDNF and NT3 at the site of injury in the chronic phase of rats subjected to either SC-contusion (SCC; moderate or severe) or SC-transection (SCT; complete or incomplete). The animals were treated with INDP immediately after injury. Rats subjected to moderate contusion or incomplete SCT showed significantly higher levels of BDNF and NT-3 as compared to PBS-immunized ones. In rats with severe SCC and complete SCT, BDNF and NT-3 concentrations were barely detected. Finally, in the fourth experiment we assessed motor function recovery in INDP-treated rats with moderate SC-injury. Rats immunized with A91 showed a significantly higher motor recovery from the first week and up to 4 months after SC-injury. CONCLUSIONS The results of this study suggest that PA boosted by immunization with A91 after moderate SC-injury can exert its benefits even at chronic stages, as shown by long-term production of BDNF and NT-3 and a substantial improvement in motor recovery.
Collapse
Affiliation(s)
- Susana Martiñón
- Facultad de Ciencias de la Salud, Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico.,Centro de Investigación del Proyecto CAMINA A.C., Mexico, D.F., Mexico.,Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Mexico, D.F., Mexico
| | - Elisa García-Vences
- Facultad de Ciencias de la Salud, Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico
| | - Diana Toscano-Tejeida
- Facultad de Ciencias de la Salud, Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico
| | - Adrian Flores-Romero
- Facultad de Ciencias de la Salud, Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico
| | - Roxana Rodriguez-Barrera
- Facultad de Ciencias de la Salud, Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico
| | - Manuel Ferrusquia
- Facultad de Ciencias de la Salud, Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico
| | - Rolando E Hernández-Muñoz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, UNAM, Mexico, D.F., Mexico
| | - Antonio Ibarra
- Facultad de Ciencias de la Salud, Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico. .,Centro de Investigación del Proyecto CAMINA A.C., Mexico, D.F., Mexico.
| |
Collapse
|
13
|
A Common Language: How Neuroimmunological Cross Talk Regulates Adult Hippocampal Neurogenesis. Stem Cells Int 2016; 2016:1681590. [PMID: 27143977 PMCID: PMC4842066 DOI: 10.1155/2016/1681590] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/17/2016] [Indexed: 12/31/2022] Open
Abstract
Immune regulation of the brain is generally studied in the context of injury or disease. Less is known about how the immune system regulates the brain during normal brain function. Recent work has redefined the field of neuroimmunology and, as long as their recruitment and activation are well regulated, immune cells are now known to have protective properties within the central nervous system in maintaining brain health. Adult neurogenesis, the process of new neuron generation in the adult brain, is highly plastic and regulated by diverse extrinsic and intrinsic cues. Emerging research has shown that immune cells and their secreted factors can influence adult neurogenesis, both under baseline conditions and during conditions known to change neurogenesis levels, such as aging and learning in an enriched environment. This review will discuss how, under nonpathological conditions, the immune system can interact with the neural stem cells to regulate adult neurogenesis with particular focus on the hippocampus—a region crucial for learning and memory.
Collapse
|
14
|
Russo R, Varano GP, Adornetto A, Nucci C, Corasaniti MT, Bagetta G, Morrone LA. Retinal ganglion cell death in glaucoma: Exploring the role of neuroinflammation. Eur J Pharmacol 2016; 787:134-42. [PMID: 27044433 DOI: 10.1016/j.ejphar.2016.03.064] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 03/10/2016] [Accepted: 03/31/2016] [Indexed: 01/06/2023]
Abstract
In clinical glaucoma, as well as in experimental models, the loss of retinal ganglion cells occurs by apoptosis. This final event is preceded by inflammatory responses involving the activation of innate and adaptive immunity, with retinal and optic nerve resident glial cells acting as major players. Here we review the current literature on the role of neuroinflammation in neurodegeneration, focusing on the inflammatory molecular mechanisms involved in the pathogenesis and progression of the optic neuropathy.
Collapse
Affiliation(s)
- Rossella Russo
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy.
| | - Giuseppe Pasquale Varano
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Annagrazia Adornetto
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Carlo Nucci
- Ophthalmology Unit, Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome Italy
| | | | - Giacinto Bagetta
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy; University Center for Adaptive Disorders and Head Pain, Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, Arcavacata di Rende, Italy
| | - Luigi Antonio Morrone
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy; University Center for Adaptive Disorders and Head Pain, Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
15
|
Ley K. 2015 Russell Ross Memorial Lecture in Vascular Biology: Protective Autoimmunity in Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:429-38. [PMID: 26821946 PMCID: PMC4970520 DOI: 10.1161/atvbaha.115.306009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/18/2016] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is an inflammatory disease of the arterial wall. It is accompanied by an autoimmune response against apolipoprotein B-100, the core protein of low-density lipoprotein, which manifests as CD4 T cell and antibody responses. To assess the role of the autoimmune response in atherosclerosis, the nature of the CD4 T cell response against apolipoprotein B-100 was studied with and without vaccination with major histocompatibility complex-II-restricted apolipoprotein B-100 peptides. The immunologic basis of autoimmunity in atherosclerosis is discussed in the framework of theories of adaptive immunity. Older vaccination approaches are also discussed. Vaccinating Apoe(-/-) mice with major histocompatibility complex-II-restricted apolipoprotein B-100 peptides reduces atheroma burden in the aorta by ≈40%. The protective mechanism likely includes secretion of interleukin-10. Protective autoimmunity limits atherosclerosis in mice and suggests potential for developing preventative and therapeutic vaccines for humans.
Collapse
Affiliation(s)
- Klaus Ley
- From the La Jolla Institute for Allergy & Immunology and Department of Bioengineering, UCSD, La Jolla, CA
| |
Collapse
|
16
|
Ellwardt E, Walsh JT, Kipnis J, Zipp F. Understanding the Role of T Cells in CNS Homeostasis. Trends Immunol 2016; 37:154-165. [DOI: 10.1016/j.it.2015.12.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/14/2015] [Accepted: 12/14/2015] [Indexed: 01/16/2023]
|
17
|
Hu JG, Shi LL, Chen YJ, Xie XM, Zhang N, Zhu AY, Jiang ZS, Feng YF, Zhang C, Xi J, Lü HZ. Differential effects of myelin basic protein-activated Th1 and Th2 cells on the local immune microenvironment of injured spinal cord. Exp Neurol 2016; 277:190-201. [PMID: 26772636 DOI: 10.1016/j.expneurol.2016.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/01/2016] [Accepted: 01/04/2016] [Indexed: 01/22/2023]
Abstract
Myelin basic protein (MBP) activated T cells (MBP-T) play an important role in the damage and repair process of the central nervous system (CNS). However, whether these cells play a beneficial or detrimental role is still a matter of debate. Although some studies showed that MBP-T cells are mainly helper T (Th) cells, their subtypes are still not very clear. One possible explanation for MBP-T immunization leading to conflicting results may be the different subtypes of T cells are responsible for distinct effects. In this study, the Th1 and Th2 type MBP-T cells (MBP-Th1 and -Th2) were polarized in vitro, and their effects on the local immune microenvironment and tissue repair of spinal cord injury (SCI) after adoptive immunization were investigated. In MBP-Th1 cell transferred rats, the high levels of pro-inflammatory cells (Th1 cells and M1 macrophages) and cytokines (IFN-γ, TNF-α, -β, IL-1β) were detected in the injured spinal cord; however, the anti-inflammatory cells (Th2 cells, regulatory T cells, and M2 macrophages) and cytokines (IL-4, -10, and -13) were found in MBP-Th2 cell transferred animals. MBP-Th2 cell transfer resulted in decreased lesion volume, increased myelination of axons, and preservation of neurons. This was accompanied by significant locomotor improvement. These results indicate that MBP-Th2 adoptive transfer has beneficial effects on the injured spinal cord, in which the increased number of Th2 cells may alter the local microenvironment from one primarily populated by Th1 and M1 cells to another dominated by Th2, Treg, and M2 cells and is conducive for SCI repair.
Collapse
Affiliation(s)
- Jian-Guo Hu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Ling-Ling Shi
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Yue-Juan Chen
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Xiu-Mei Xie
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Nan Zhang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - An-You Zhu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Zheng-Song Jiang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Yi-Fan Feng
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Chen Zhang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Jin Xi
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - He-Zuo Lü
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Department of Immunology, Bengbu Medical College, Anhui 233030, PR China.
| |
Collapse
|
18
|
Zeira O, Asiag N, Aralla M, Ghezzi E, Pettinari L, Martinelli L, Zahirpour D, Dumas MP, Lupi D, Scaccia S, Konar M, Cantile C. Adult autologous mesenchymal stem cells for the treatment of suspected non-infectious inflammatory diseases of the canine central nervous system: safety, feasibility and preliminary clinical findings. J Neuroinflammation 2015; 12:181. [PMID: 26415563 PMCID: PMC4587680 DOI: 10.1186/s12974-015-0402-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/22/2015] [Indexed: 01/29/2023] Open
Abstract
Background Non-infectious inflammatory diseases of the canine central nervous system (CNS) are common idiopathic disorders grouped under the term meningoencephalomyelitis of unknown origin (MUO). Ante mortem diagnosis is achieved via assessment of clinical signs, magnetic resonance imaging (MRI), and cerebrospinal fluid (CSF) analysis, but the definitive diagnosis needs histopathological examination. MUO are mostly considered as autoimmune CNS disorders, so that suppressing the immune reaction is the best management method for patients. Mesenchymal stem cells (MSCs) are under investigation to treat autoimmune and degenerative disorders due to their immunomodulatory and regenerative properties. This study aims to verify the safety, feasibility, and efficacy of MSCs treatment in canine idiopathic autoimmune inflammatory disorders of the CNS. Methods Eight dogs presented with acute onset and rapid progression of multifocal neurological signs were selected to the study. In all patients’ physical and neurological examinations, MRI and CSF analyses were performed. Clinical diagnosis in all cases was MUO. All selected dogs responded initially to immunosuppressive drugs (prednisone and a combination of prednisolone and cytosine arabinoside) but developed undesirable side effects. For all eight dogs, the owners considered euthanasia but accepted cell therapy as a last possibility. Autologous bone marrow MSCs (BMMSCs), isolated, cultured, and expanded, were administered by intrathecal (IT) injection in the cisterna magna intravenously (IV) and by intra-arterial (IA) injection in the right carotid artery. Adverse effects and clinical response were monitored for 6 months up to 2-year follow-up. Results The use of autologous BMMSCs in dogs with MUO was safe for IT, IV, and IA injections. No major short- or long-term adverse effects were registered. All the dogs presented early improvement in their general and neurological conditions, with particular effect on cervical pain. The group of dogs treated by IT+IA administration showed a shorter time of reaction to therapy compared to the group treated by IT+IV administration. Conclusions MSCs treatment in dogs affected by MOU is safe and feasible. A larger group of dogs is needed to confirm these results as well as CNS histology in order to better understand the underlying mechanisms.
Collapse
Affiliation(s)
- Offer Zeira
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Nimrod Asiag
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Marina Aralla
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Erica Ghezzi
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Letizia Pettinari
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Laura Martinelli
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Daniele Zahirpour
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Maria Pia Dumas
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Davide Lupi
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Simone Scaccia
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Martin Konar
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Carlo Cantile
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, 56124, Pisa, Italy.
| |
Collapse
|
19
|
Chen L, Yao Y, Wei C, Sun Y, Ma X, Zhang R, Xu X, Hao J. T cell immunity to glatiramer acetate ameliorates cognitive deficits induced by chronic cerebral hypoperfusion by modulating the microenvironment. Sci Rep 2015; 5:14308. [PMID: 26391515 PMCID: PMC4585746 DOI: 10.1038/srep14308] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/24/2015] [Indexed: 11/09/2022] Open
Abstract
Vascular dementia (VaD) is a progressive and highly prevalent disorder. However, in a very large majority of cases, a milieu of cellular and molecular events common for multiple neurodegenerative diseases is involved. Our work focused on whether the immunomodulating effect of glatiramer acetate (GA) could restore normalcy to the microenvironment and ameliorate cognitive decline induced by chronic cerebral hypoperfusion. We assessed cognitive function by rats’ performance in a Morris water maze (MWM), electrophysiological recordings and by pathologic changes. The results suggest that GA reduced cognitive deficits by reestablishing an optimal microenvironment such as increasing expression of the brain-derived neurotrophic factor (BDNF) and modulating the Th1/Th2 cytokine balance in the hippocampus. When microenvironmental homeostasis is restored, cholinergic activity becomes involved in ameliorating cellular damage. Since vaccination with GA can boost “protective autoimmunity” in this way, a similar strategy may have therapeutic potential for alleviating VaD disease.
Collapse
Affiliation(s)
- Li Chen
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yang Yao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Changjuan Wei
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yanan Sun
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaofeng Ma
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Rongxin Zhang
- Center for Basic Medical Science Research, Tianjin Medical University, Tianjin 300070, China
| | - Xiaolin Xu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300060, China
| | - Junwei Hao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
20
|
Mac Nair CE, Nickells RW. Neuroinflammation in Glaucoma and Optic Nerve Damage. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 134:343-63. [PMID: 26310164 DOI: 10.1016/bs.pmbts.2015.06.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Glaucoma is a group of optic neuropathies characterized by the degeneration of retinal ganglion cell axons and somas, ultimately preventing light signals in the retina from reaching the brain. Glaucoma is a leading cause of blindness in the world, and treatment options for patients remain limited and minimally efficacious. A number of mechanisms have been linked to glaucomatous pathophysiology. A leading role is now attributed to neuroinflammatory conditions generated by the resident innate immune cells in the optic nerve and retina. Since the eye is immune privileged, the adaptation of these innate immune cells, termed glia, is crucial following trauma. In this chapter, we discuss the mechanisms associated with normal glial function in a healthy eye, and how changes in glial activation can contribute to the process of glaucomatous neurodegeneration in both the optic nerve and retina.
Collapse
Affiliation(s)
- Caitlin E Mac Nair
- Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Robert W Nickells
- Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
21
|
Experimentally Induced Mammalian Models of Glaucoma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:281214. [PMID: 26064891 PMCID: PMC4433635 DOI: 10.1155/2015/281214] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/01/2014] [Indexed: 11/17/2022]
Abstract
A wide variety of animal models have been used to study glaucoma. Although these models provide valuable information about the disease, there is still no ideal model for studying glaucoma due to its complex pathogenesis. Animal models for glaucoma are pivotal for clarifying glaucoma etiology and for developing novel therapeutic strategies to halt disease progression. In this review paper, we summarize some of the major findings obtained in various glaucoma models and examine the strengths and limitations of these models.
Collapse
|
22
|
Kostic M, Stojanovic I, Marjanovic G, Zivkovic N, Cvetanovic A. Deleterious versus protective autoimmunity in multiple sclerosis. Cell Immunol 2015; 296:122-32. [PMID: 25944389 DOI: 10.1016/j.cellimm.2015.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/18/2015] [Accepted: 04/22/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disorder of central nervous system, in which myelin specific CD4(+) T cells have a central role in orchestrating pathological events involved in disease pathogenesis. There is compelling evidence that Th1, Th9 and Th17 cells, separately or in cooperation, could mediate deleterious autoimmune response in MS. However, the phenotype differences between Th cell subpopulations initially employed in MS pathogenesis are mainly reflected in the different patterns of inflammation introduction, which results in the development of characteristic pathological features (blood-brain barrier disruption, demyelination and neurodegeneration), clinically presented with MS symptoms. Although, autoimmunity was traditionally seen as deleterious, some studies indicated that autoimmunity mediated by Th2 cells and T regulatory cells could be protective by nature. The concept of protective autoimmunity in MS pathogenesis is still poorly understood, but could be of great importance in better understanding of MS immunology and therefore, creating better therapeutic strategies.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia.
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Goran Marjanovic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Ana Cvetanovic
- Clinic of Oncology, Clinical Centre, Blvd. Dr. Zorana Djindjica 48, 18000 Nis, Serbia
| |
Collapse
|
23
|
Sharma TP, Liu Y, Wordinger RJ, Pang IH, Clark AF. Neuritin 1 promotes retinal ganglion cell survival and axonal regeneration following optic nerve crush. Cell Death Dis 2015; 6:e1661. [PMID: 25719245 PMCID: PMC4669798 DOI: 10.1038/cddis.2015.22] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 12/11/2014] [Accepted: 01/06/2015] [Indexed: 12/16/2022]
Abstract
Neuritin 1 (Nrn1) is an extracellular glycophosphatidylinositol-linked protein that stimulates axonal plasticity, dendritic arborization and synapse maturation in the central nervous system (CNS). The purpose of this study was to evaluate the neuroprotective and axogenic properties of Nrn1 on axotomized retinal ganglion cells (RGCs) in vitro and on the in vivo optic nerve crush (ONC) mouse model. Axotomized cultured RGCs treated with recombinant hNRN1 significantly increased survival of RGCs by 21% (n=6–7, P<0.01) and neurite outgrowth in RGCs by 141% compared to controls (n=15, P<0.05). RGC transduction with AAV2-CAG–hNRN1 prior to ONC promoted RGC survival (450%, n=3–7, P<0.05) and significantly preserved RGC function by 70% until 28 days post crush (dpc) (n=6, P<0.05) compared with the control AAV2-CAG–green fluorescent protein transduction group. Significantly elevated levels of RGC marker, RNA binding protein with multiple splicing (Rbpms; 73%, n=5–8, P<0.001) and growth cone marker, growth-associated protein 43 (Gap43; 36%, n=3, P<0.01) were observed 28 dpc in the retinas of the treatment group compared with the control group. Significant increase in Gap43 (100%, n=5–6, P<0.05) expression was observed within the optic nerves of the AAV2–hNRN1 group compared to controls. In conclusion, Nrn1 exhibited neuroprotective, regenerative effects and preserved RGC function on axotomized RGCs in vitro and after axonal injury in vivo. Nrn1 is a potential therapeutic target for CNS neurodegenerative diseases.
Collapse
Affiliation(s)
- T P Sharma
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Y Liu
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - R J Wordinger
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - I-H Pang
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA
| | - A F Clark
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
24
|
Walsh JT, Hendrix S, Boato F, Smirnov I, Zheng J, Lukens JR, Gadani S, Hechler D, Gölz G, Rosenberger K, Kammertöns T, Vogt J, Vogelaar C, Siffrin V, Radjavi A, Fernandez-Castaneda A, Gaultier A, Gold R, Kanneganti TD, Nitsch R, Zipp F, Kipnis J. MHCII-independent CD4+ T cells protect injured CNS neurons via IL-4. J Clin Invest 2015; 125:699-714. [PMID: 25607842 PMCID: PMC4319416 DOI: 10.1172/jci76210] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 12/02/2014] [Indexed: 12/13/2022] Open
Abstract
A body of experimental evidence suggests that T cells mediate neuroprotection following CNS injury; however, the antigen specificity of these T cells and how they mediate neuroprotection are unknown. Here, we have provided evidence that T cell-mediated neuroprotection after CNS injury can occur independently of major histocompatibility class II (MHCII) signaling to T cell receptors (TCRs). Using two murine models of CNS injury, we determined that damage-associated molecular mediators that originate from injured CNS tissue induce a population of neuroprotective, IL-4-producing T cells in an antigen-independent fashion. Compared with wild-type mice, IL-4-deficient animals had decreased functional recovery following CNS injury; however, transfer of CD4+ T cells from wild-type mice, but not from IL-4-deficient mice, enhanced neuronal survival. Using a culture-based system, we determined that T cell-derived IL-4 protects and induces recovery of injured neurons by activation of neuronal IL-4 receptors, which potentiated neurotrophin signaling via the AKT and MAPK pathways. Together, these findings demonstrate that damage-associated molecules from the injured CNS induce a neuroprotective T cell response that is independent of MHCII/TCR interactions and is MyD88 dependent. Moreover, our results indicate that IL-4 mediates neuroprotection and recovery of the injured CNS and suggest that strategies to enhance IL-4-producing CD4+ T cells have potential to attenuate axonal damage in the course of CNS injury in trauma, inflammation, or neurodegeneration.
Collapse
Affiliation(s)
- James T. Walsh
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Sven Hendrix
- Department of Morphology and BIOMED Institute, Hasselt University, Diepenbeek, Belgium
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | - Francesco Boato
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Igor Smirnov
- Center for Brain Immunology and Glia
- Department of Neuroscience
| | - Jingjing Zheng
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Institute of Neurosciences, Fourth Military Medical University, Xi’an, China
| | - John R. Lukens
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sachin Gadani
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel Hechler
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | - Greta Gölz
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | - Karen Rosenberger
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | | | - Johannes Vogt
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christina Vogelaar
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Volker Siffrin
- Department of Neurology, Focus Program Translational Neuroscience and Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ali Radjavi
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Microbiology, Immunology and Infectious Diseases, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | | | - Alban Gaultier
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital/Ruhr-University Bochum, Bochum, Germany
| | | | - Robert Nitsch
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience and Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Graduate Program in Microbiology, Immunology and Infectious Diseases, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
25
|
Kumaran AM, Sundar G, Chye LT. Traumatic optic neuropathy: a review. Craniomaxillofac Trauma Reconstr 2014; 8:31-41. [PMID: 25709751 DOI: 10.1055/s-0034-1393734] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 05/03/2014] [Indexed: 01/08/2023] Open
Abstract
The aim of this article is to evaluate current literature on investigation and management of traumatic optic neuropathy (TON), propose recommendations for diagnosis and management, and explore novel future treatments. TON, though uncommon, causes substantial visual loss. Without clear guidelines, there is much ambiguity regarding its diagnosis and management. Investigation and treatment (conservative, medical, surgical, and combined) vary widely between centers. Electronic databases PubMed, MEDLINE, PROSPERO, CENTRAL, and EMBASE were searched for content that matched "Traumatic optic neuropathy." Articles with abstracts and full text available, published in the past 10 years, written English and limited to human adults, were selected. All study designs were acceptable except case reports and case series with fewer 10 patients. All abstracts were then evaluated for relevance. References of these studies were evaluated and if also relevant, included. A total of 2,686 articles were retrieved and 43 examined for relevance. Of these, 23 articles were included. TON is a clinical diagnosis. Visual-evoked potential is useful in diagnosis and prognosis. Computed tomography demonstrates canal fractures and concomitant injuries. Magnetic resonance images should be reserved for select and stable patients. Conservative treatment is appropriate in mild TON. Steroids are of questionable benefit and may be harmful. Surgery should be reserved for patients with radiological evidence of compression and individualized.
Collapse
Affiliation(s)
| | - Gangadhara Sundar
- Department of Ophthalmology, National University Hospital, National University Health System, Singapore
| | - Lim Thiam Chye
- Division of Plastic, Reconstructive and Aesthetic Surgery, National University Hospital, National University Health System, Singapore
| |
Collapse
|
26
|
Coates JR, Jeffery ND. Perspectives on Meningoencephalomyelitis of Unknown Origin. Vet Clin North Am Small Anim Pract 2014; 44:1157-85. [DOI: 10.1016/j.cvsm.2014.07.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
27
|
Heuss ND, Pierson MJ, Montaniel KRC, McPherson SW, Lehmann U, Hussong SA, Ferrington DA, Low WC, Gregerson DS. Retinal dendritic cell recruitment, but not function, was inhibited in MyD88 and TRIF deficient mice. J Neuroinflammation 2014; 11:143. [PMID: 25116321 PMCID: PMC4149240 DOI: 10.1186/s12974-014-0143-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 07/29/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Immune system cells are known to affect loss of neurons due to injury or disease. Recruitment of immune cells following retinal/CNS injury has been shown to affect the health and survival of neurons in several models. We detected close, physical contact between dendritic cells and retinal ganglion cells following an optic nerve crush, and sought to understand the underlying mechanisms. METHODS CD11c-DTR/GFP mice producing a chimeric protein of diphtheria toxin receptor (DTR) and GFP from a transgenic CD11c promoter were used in conjunction with mice deficient in MyD88 and/or TRIF. Retinal ganglion cell injury was induced by an optic nerve crush, and the resulting interactions of the GFPhi cells and retinal ganglion cells were examined. RESULTS Recruitment of GFPhi dendritic cells to the retina was significantly compromised in MyD88 and TRIF knockout mice. GFPhi dendritic cells played a significant role in clearing fluorescent-labeled retinal ganglion cells post-injury in the CD11c-DTR/GFP mice. In the TRIF and MyD88 deficient mice, the resting level of GFPhi dendritic cells was lower, and their influx was reduced following the optic nerve crush injury. The reduction in GFPhi dendritic cell numbers led to their replacement in the uptake of fluorescent-labeled debris by GFPlo microglia/macrophages. Depletion of GFPhi dendritic cells by treatment with diphtheria toxin also led to their displacement by GFPlo microglia/macrophages, which then assumed close contact with the injured neurons. CONCLUSIONS The contribution of recruited cells to the injury response was substantial, and regulated by MyD88 and TRIF. However, the presence of these adaptor proteins was not required for interaction with neurons, or the phagocytosis of debris. The data suggested a two-niche model in which resident microglia were maintained at a constant level post-optic nerve crush, while the injury-stimulated recruitment of dendritic cells and macrophages led to their transient appearance in numbers equivalent to or greater than the resident microglia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dale S Gregerson
- Department of Ophthalmology & Visual Neurosciences, University of Minnesota, Lions Research Bldg, Rm 314, 2001 6th St SE, Minneapolis 55455, MN, USA.
| |
Collapse
|
28
|
Sharma TP, McDowell CM, Liu Y, Wagner AH, Thole D, Faga BP, Wordinger RJ, Braun TA, Clark AF. Optic nerve crush induces spatial and temporal gene expression patterns in retina and optic nerve of BALB/cJ mice. Mol Neurodegener 2014; 9:14. [PMID: 24767545 PMCID: PMC4113182 DOI: 10.1186/1750-1326-9-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 04/18/2014] [Indexed: 12/18/2022] Open
Abstract
Background Central nervous system (CNS) trauma and neurodegenerative disorders trigger a cascade of cellular and molecular events resulting in neuronal apoptosis and regenerative failure. The pathogenic mechanisms and gene expression changes associated with these detrimental events can be effectively studied using a rodent optic nerve crush (ONC) model. The purpose of this study was to use a mouse ONC model to: (a) evaluate changes in retina and optic nerve (ON) gene expression, (b) identify neurodegenerative pathogenic pathways and (c) discover potential new therapeutic targets. Results Only 54% of total neurons survived in the ganglion cell layer (GCL) 28 days post crush. Using Bayesian Estimation of Temporal Regulation (BETR) gene expression analysis, we identified significantly altered expression of 1,723 and 2,110 genes in the retina and ON, respectively. Meta-analysis of altered gene expression (≥1.5, ≤-1.5, p < 0.05) using Partek and DAVID demonstrated 28 up and 20 down-regulated retinal gene clusters and 57 up and 41 down-regulated optic nerve clusters. Regulated gene clusters included regenerative change, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. Expression of selected genes (Vsnl1, Syt1, Synpr and Nrn1) from retinal and ON neuronal clusters were quantitatively and qualitatively examined for their relation to axonal neurodegeneration by immunohistochemistry and qRT-PCR. Conclusion A number of detrimental gene expression changes occur that contribute to trauma-induced neurodegeneration after injury to ON axons. Nrn1 (synaptic plasticity gene), Synpr and Syt1 (synaptic vesicle fusion genes), and Vsnl1 (neuron differentiation associated gene) were a few of the potentially unique genes identified that were down-regulated spatially and temporally in our rodent ONC model. Bioinformatic meta-analysis identified significant tissue-specific and time-dependent gene clusters associated with regenerative changes, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. These ONC induced neuronal loss and regenerative failure associated clusters can be extrapolated to changes occurring in other forms of CNS trauma or in clinical neurodegenerative pathological settings. In conclusion, this study identified potential therapeutic targets to address two key mechanisms of CNS trauma and neurodegeneration: neuronal loss and regenerative failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Abbot F Clark
- North Texas Eye Research Institute, Ft, Worth, TX USA.
| |
Collapse
|
29
|
Liu NK, Deng LX, Zhang YP, Lu QB, Wang XF, Hu JG, Oakes E, Bonventre JV, Shields CB, Xu XM. Cytosolic phospholipase A2 protein as a novel therapeutic target for spinal cord injury. Ann Neurol 2014; 75:644-58. [PMID: 24623140 PMCID: PMC4320750 DOI: 10.1002/ana.24134] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 02/28/2014] [Accepted: 03/10/2014] [Indexed: 12/17/2022]
Abstract
Objective The objective of this study was to investigate whether cytosolic phospholipase A2 (cPLA2), an important isoform of PLA2 that mediates the release of arachidonic acid, plays a role in the pathogenesis of spinal cord injury (SCI). Methods A combination of molecular, histological, immunohistochemical, and behavioral assessments were used to test whether blocking cPLA2 activation pharmacologically or genetically reduced cell death, protected spinal cord tissue, and improved behavioral recovery after a contusive SCI performed at the 10th thoracic level in adult mice. Results SCI significantly increased cPLA2 expression and activation. Activated cPLA2 was localized mainly in neurons and oligodendrocytes. Notably, the SCI-induced cPLA2 activation was mediated by the extracellular signal-regulated kinase signaling pathway. In vitro, activation of cPLA2 by ceramide-1-phosphate or A23187 induced spinal neuronal death, which was substantially reversed by arachidonyl trifluoromethyl ketone, a cPLA2 inhibitor. Remarkably, blocking cPLA2 pharmacologically at 30 minutes postinjury or genetically deleting cPLA2 in mice ameliorated motor deficits, and reduced cell loss and tissue damage after SCI. Interpretation cPLA2 may play a key role in the pathogenesis of SCI, at least in the C57BL/6 mouse, and as such could be an attractive therapeutic target for ameliorating secondary tissue damage and promoting recovery of function after SCI.
Collapse
Affiliation(s)
- Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
del Barco DG, Berlanga J, Penton E, Hardiman O, Montero E. Boosting controlled autoimmunity: a new therapeutic target for CNS disorders. Expert Rev Neurother 2014; 8:819-25. [DOI: 10.1586/14737175.8.5.819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Radjavi A, Smirnov I, Kipnis J. Brain antigen-reactive CD4+ T cells are sufficient to support learning behavior in mice with limited T cell repertoire. Brain Behav Immun 2014; 35:58-63. [PMID: 24012647 PMCID: PMC3858511 DOI: 10.1016/j.bbi.2013.08.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/16/2013] [Accepted: 08/28/2013] [Indexed: 01/18/2023] Open
Abstract
Numerous methods of T cell depletion lead to impairment of learning and memory function in mice. While adoptive transfer of whole splenocytes rescues learning behavior impairments, the precise sub-population and antigenic specificity of the T cells mediating the rescue remains unknown. Using several transgenic mouse models in combination with adoptive transfers, we demonstrate the necessity of an antigen-specific CD4(+) T cell compartment in normal spatial learning and memory, as measured by the Morris water maze (MWM). Moreover, transfer of a monoclonal T cell population reactive to the central nervous system (CNS) antigen, myelin oligodendrocyte glycoprotein (MOG), was sufficient to improve cognitive task performance in otherwise impaired OTII mice, raising the possibility that the antigen-specificity requirement of pro-cognitive T cells may be directed against CNS-derived self-antigens.
Collapse
Affiliation(s)
- Ali Radjavi
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Immunology Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| | - Igor Smirnov
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Immunology Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
32
|
Schwartz M, Baruch K. The resolution of neuroinflammation in neurodegeneration: leukocyte recruitment via the choroid plexus. EMBO J 2013; 33:7-22. [PMID: 24357543 DOI: 10.1002/embj.201386609] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inflammation is an integral part of the body's physiological repair mechanism, unless it remains unresolved and becomes pathological, as evident in the progressive nature of neurodegeneration. Based on studies from outside the central nervous system (CNS), it is now understood that the resolution of inflammation is an active process, which is dependent on well-orchestrated innate and adaptive immune responses. Due to the immunologically privileged status of the CNS, such resolution mechanism has been mostly ignored. Here, we discuss resolution of neuroinflammation as a process that depends on a network of immune cells operating in a tightly regulated sequence, involving the brain's choroid plexus (CP), a unique neuro-immunological interface, positioned to integrate signals it receives from the CNS parenchyma with signals coming from circulating immune cells, and to function as an on-alert gate for selective recruitment of inflammation-resolving leukocytes to the inflamed CNS parenchyma. Finally, we propose that functional dysregulation of the CP reflects a common underlying mechanism in the pathophysiology of neurodegenerative diseases, and can thus serve as a potential novel target for therapy.
Collapse
Affiliation(s)
- Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
33
|
Qian S, Tang Y, Cheng L, Sun X, Tian J, Zhou C. Interaction of copolymer-1-activated T cells and microglia in retinal ganglion cell protection. Clin Exp Ophthalmol 2013; 41:881-90. [PMID: 23566072 DOI: 10.1111/ceo.12110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Accepted: 03/14/2013] [Indexed: 12/14/2022]
Affiliation(s)
- Shaohong Qian
- Department of Ophthalmology; EENT Hospital of Fudan University; Shanghai China
| | - Yating Tang
- Department of Ophthalmology; EENT Hospital of Fudan University; Shanghai China
| | - Lina Cheng
- Department of Ophthalmology; no. 2 Hospital of Xi'an; Shanxi China
| | - Xinghuai Sun
- Department of Ophthalmology; EENT Hospital of Fudan University; Shanghai China
| | - Jie Tian
- Central Lab; EENT Hospital of Fudan University; Shanghai China
| | - Chuandi Zhou
- Department of Ophthalmology; EENT Hospital of Fudan University; Shanghai China
| |
Collapse
|
34
|
Smirnov I, Walsh JT, Kipnis J. Chronic mild stress eliminates the neuroprotective effect of Copaxone after CNS injury. Brain Behav Immun 2013; 31:177-82. [PMID: 23295266 PMCID: PMC3633726 DOI: 10.1016/j.bbi.2012.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/26/2012] [Accepted: 12/23/2012] [Indexed: 01/01/2023] Open
Abstract
Copolymer (Cop)-1, also known as glatiramer acetate, is an active compound of Copaxone, a drug widely used by patients with multiple sclerosis (MS). Copaxone functions in MS through two mechanisms of action, namely immunomodulation and neuroprotection. Because the immune system is suppressed or altered in depressed individuals, and since depression is often associated with neurological conditions, we were interested in examining whether the neuroprotective effect of Copaxone persists under conditions of stress-induced depressive behavior. We exposed mice to unpredictable chronic mild stress for 4 weeks and then treated them with three doses of Copaxone at 3-day intervals, with the last dose given immediately before the mice underwent a crush injury to the optic nerve. Whereas nonstressed mice exhibited a strong neuroprotective response after Copaxone treatment, this effect was completely absent in mice that underwent chronic mild stress. Interestingly, when Copaxone was combined with Prozac, the neuroprotective effect of Copaxone was regained, suggesting that chronic mild stress interferes with the neuroprotective effect of Copaxone. These results may shed a light on mechanism of action of Copaxone and lead to new combined therapies for neurodegenerative and neuroinflammatory disorders.
Collapse
Affiliation(s)
- Igor Smirnov
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - James T. Walsh
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA,Graduate Program in Neuroscience and Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA,Graduate Program in Neuroscience and Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA,Correspondence should be addressed to J.K. () Tel: 434-982-3858, Fax: (434)-982-4380
| |
Collapse
|
35
|
CD4 positive T helper cells contribute to retinal ganglion cell death in mouse model of ischemia reperfusion injury. Exp Eye Res 2013; 115:131-9. [PMID: 23792169 DOI: 10.1016/j.exer.2013.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 04/23/2013] [Accepted: 06/10/2013] [Indexed: 11/23/2022]
Abstract
Neuron degeneration is a common pathological process associated with many disease conditions in the central nervous system including retina. Although immune responses have been proposed as one potential element in triggering neural damage, the mechanism of action of specific immune components underlying the pathogenesis is unclear. In this study we focus on adaptive immune activities to evaluate CD4 positive helper cells in the retinal ganglion cell (RGC) degeneration in response to transient retinal ischemic/reperfusion (I/R) injury. Transient retinal ischemia was induced in four mouse strains with different immune backgrounds, including wild type mice from C57BL/6 and BABL/c strains, severe combined immunodeficient (SCID) mice lacking T and B lymphocytes, SCID mice with transferred wild type CD4+ T cells, and the STAT6 deficient mice without T helper 2 (TH2) cells. In SCID mice RGCs showed a strong resistance to cell death in response to I/R injury (89% ± 3% of the survival cells in contralateral eye) compared with C57BL/6 (p = 0.018) and BALB/C (p = 0.038) wild types. By transferring the mature CD4+ T cells from matched wild type into SCID mice, the resistance of RGCs to injury was significantly compromised (p < 0.05). Furthermore a significant resistance of RGCs to cell death (p < 0.05) accompanied with an overexpression of STAT1 and STAT3 was confirmed in STAT6 deficient mice in response to I/R injury compared with the wild type controls, indicating that TH2 cells maturation might be involved in RGC damage. Adaptive immunity carried by CD4 T cells plays an essential role in RGC degeneration.
Collapse
|
36
|
Eisenkraft A, Falk A, Finkelstein A. The role of glutamate and the immune system in organophosphate-induced CNS damage. Neurotox Res 2013; 24:265-79. [PMID: 23532600 DOI: 10.1007/s12640-013-9388-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/10/2013] [Accepted: 03/15/2013] [Indexed: 12/12/2022]
Abstract
Organophosphate (OP) poisoning is associated with long-lasting neurological damage, which is attributed mainly to the excessive levels of glutamate caused by the intoxication. Glutamate toxicity, however, is not specific to OP poisoning, and is linked to propagation of damage in both acute and chronic neurodegenerative conditions in the central nervous system (CNS). In addition to acute excitotoxic effects of glutamate, there is now a growing amount of evidence of its intricate immunomodulatory effects in the brain, involving both the innate and the adaptive immune systems. Moreover, it was demonstrated that immunomodulatory treatments, aimed at regulating the interaction between the resident immune cells of the brain (microglia) and the peripheral immune system, can support buffering of excessive levels of glutamate and restoration of the homeostasis. In this review, we will discuss the role of glutamate as an excitotoxic agent in the acute phase of OP poisoning, and the possible functions it may have as both a neuroprotectant and an immunomodulator in the sub-acute and chronic phases of OP poisoning. In addition, we will describe the novel immune-based neuroprotective strategies aimed at counteracting the long-term neurodegenerative effects of glutamate in the CNS.
Collapse
|
37
|
Wang K, Chao R, Guo QN, Liu MY, Liang HP, Liu P, Zhao JH. Expressions of some neurotrophins and neurotrophic cytokines at site of spinal cord injury in mice after vaccination with dendritic cells pulsed with homogenate proteins. Neuroimmunomodulation 2013; 20:87-98. [PMID: 23257628 DOI: 10.1159/000345522] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 10/22/2012] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Immune cells are key mediators of secondary damage following spinal cord injury (SCI), and dendritic cell (DC)-based vaccines have received considerable interest for treatment of SCI. We previously showed that vaccination with DCs pulsed with homogenate proteins of the spinal cord (hpDCs) promotes functional recovery from SCI in mice. However, the underlying molecular mechanisms remain unclear. Here, changes of neurotrophins, cytokines and T cells at the site of SCI in mice after vaccination with hpDCs were investigated and correlated with recovery from SCI. METHODS hpDCs, DCs (control) or PBS (control) were injected intraperitoneally into injured mouse spinal cords. Functional recovery of the spinal cord was measured weekly using the Basso Mouse Scale (BMS) and confirmed by histological and immunohistochemical analysis. Brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), interleukin-4 (IL-4) and interferon-γ (IFN-γ) levels in T cell culture supernatants and spinal cord tissues were determined by ELISA. RESULTS Eighty-four days after immunization, the BMS score of the hpDCs group (6.92 ± 0.20) was significantly higher than those of the DCs and PBS groups (p < 0.01). Meanwhile, the injury area and number of cysts in the hpDCs group decreased significantly compared with control groups. BDNF, NT-3, IL-4 and IFN-γ levels at the injured site as well as BDNF and NT-3 levels in the supernatant of cultured T cells from the hpDCs group were significantly higher than in control groups (p < 0.05). CONCLUSION These results reveal that vaccination with hpDCs can promote SCI repair potentially by upregulating BDNF, NT-3, IL-4 and IFN-γ at the injury site.
Collapse
Affiliation(s)
- Ke Wang
- Department of Spine Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University, Chongqing, PR China
| | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Abstract
Interactions between the central nervous system and the immune system have been studied primarily in the context of pathology, popularizing the view that interplay between these two systems is inherently detrimental. However, recent experimental data have demonstrated productive neuroimmune interactions that occur under normal physiological conditions. In this Essay, we outline our current understanding of contemporary neuroimmunology, describe a working model of T cell function in support of learning and memory, and offer ideas regarding the selective advantages of immune-mediated effects on brain function.
Collapse
Affiliation(s)
- Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | |
Collapse
|
40
|
Wang HJ, Hu JG, Shen L, Wang R, Wang QY, Zhang C, Xi J, Zhou JS, Lü HZ. Passive Immunization With Myelin Basic Protein Activated T Cells Suppresses Axonal Dieback but Does Not Promote Axonal Regeneration Following Spinal Cord Hemisection in Adult Rats. Int J Neurosci 2012; 122:458-65. [DOI: 10.3109/00207454.2012.678443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Hu JG, Shen L, Wang R, Wang QY, Zhang C, Xi J, Ma SF, Zhou JS, Lü HZ. Effects of Olig2-overexpressing neural stem cells and myelin basic protein-activated T cells on recovery from spinal cord injury. Neurotherapeutics 2012; 9:422-45. [PMID: 22173726 PMCID: PMC3337015 DOI: 10.1007/s13311-011-0090-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Neural stem cell (NSC) transplantation is a major focus of current research for treatment of spinal cord injury (SCI). However, it is very important to promote the survival and differentiation of NSCs into myelinating oligodendrocytes (OLs). In this study, myelin basic protein-activated T (MBP-T) cells were passively immunized to improve the SCI microenvironment. Olig2-overexpressing NSCs were infected with a lentivirus carrying the enhanced green fluorescent protein (GFP) reporter gene to generate Olig2-GFP-NSCs that were transplanted into the injured site to differentiate into OLs. Transferred MBP-T cells infiltrated the injured spinal cord, produced neurotrophic factors, and induced the differentiation of resident microglia and/or infiltrating blood monocytes into an "alternatively activated" anti-inflammatory macrophage phenotype by producing interleukin-13. As a result, the survival of transplanted NSCs increased fivefold in MBP-T cell-transferred rats compared with that of the vehicle-treated control. In addition, the differentiation of MBP-positive OLs increased 12-fold in Olig2-GFP-NSC-transplanted rats compared with that of GFP-NSC-transplanted controls. In the MBP-T cell and Olig2-GFP-NSC combined group, the number of OL-remyelinated axons significantly increased compared with those of all other groups. However, a significant decrease in spinal cord lesion volume and an increase in spared myelin and behavioral recovery were observed in Olig2-NSC- and NSC-transplanted MBP-T cell groups. Collectively, these results suggest that MBP-T cell adoptive immunotherapy combined with NSC transplantation has a synergistic effect on histological and behavioral improvement after traumatic SCI. Although Olig2 overexpression enhances OL differentiation and myelination, the effect on functional recovery may be surpassed by MBP-T cells.
Collapse
Affiliation(s)
- Jian-Guo Hu
- />Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Anhui, 233004 People’s Republic of China
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - Lin Shen
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - Rui Wang
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - Qi-Yi Wang
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - Chen Zhang
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - Jin Xi
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - Shan-Feng Ma
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - Jian-Sheng Zhou
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| | - He-Zuo Lü
- />Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Anhui, 233004 People’s Republic of China
- />Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Anhui, 233004 People’s Republic of China
| |
Collapse
|
42
|
Palumbo ML, Trinchero MF, Zorrilla-Zubilete MA, Schinder AF, Genaro AM. Glatiramer acetate reverts stress-induced alterations on adult neurogenesis and behavior. Involvement of Th1/Th2 balance. Brain Behav Immun 2012; 26:429-38. [PMID: 22200600 DOI: 10.1016/j.bbi.2011.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 12/11/2011] [Accepted: 12/12/2011] [Indexed: 01/15/2023] Open
Abstract
Long-term exposure to stressful situations has deleterious effects on adult neurogenesis, behavior, and the immune system. We have previously shown that stressed BALB/c mice show poor learning performance, which correlates with an increase in the T helper 1/T helper 2 (Th1/Th2) cytokine balance. Glatiramer acetate (GA) can stimulate autoreactive T cells. In this work we investigated the effects of GA treatment on BALB/c mice exposed to chronic mild stress (CMS). Stressed mice exhibited a significant decline in their performance in the open field and Y-maze tasks, which was accompanied by a reduction in dentate gyrus neurogenesis and an altered Th1/Th2 balance. Interestingly, after 6 weeks of CMS exposure administration of GA reestablished normal levels of adult neurogenesis, restored the Th1/Th2 balance, and improved learning performance. These results demonstrate that GA treatment can reverse the learning impairment induced by stress through a mechanism that likely involves the regulation of the cytokine balance and adult neurogenesis.
Collapse
Affiliation(s)
- María Laura Palumbo
- CEFYBO-CONICET, 1ª, Cátedra de Farmacología, Facultad de Medicina, UBA, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
43
|
García E, Silva-García R, Mestre H, Flores N, Martiñón S, Calderón-Aranda ES, Ibarra A. Immunization with A91 peptide or copolymer-1 reduces the production of nitric oxide and inducible nitric oxide synthase gene expression after spinal cord injury. J Neurosci Res 2011; 90:656-63. [PMID: 22002544 DOI: 10.1002/jnr.22771] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 07/01/2011] [Accepted: 07/22/2011] [Indexed: 11/11/2022]
Abstract
Immunization with neurally derived peptides (INDP) boosts the action of an autoreactive immune response that has been shown to induce neuroprotection in several neurodegenerative diseases, especially after spinal cord (SC) injury. This strategy provides an environment that promotes neuronal survival and tissue preservation. The mechanisms by which this autoreactive response exerts its protective effects is not totally understood at the moment. A recent study showed that INDP reduces lipid peroxidation. Lipid peroxidation is a neurodegenerative phenomenon caused by the increased production of reactive nitrogen species such as nitric oxide (NO). It is possible that INDP could be interfering with NO production. To test this hypothesis, we examined the effect of INDP on the amount of NO produced by glial cells when cocultured with autoreactive T cells. We also evaluated the amount of NO and the expression of the inducible form of nitric oxide synthase (iNOS) at the injury site of SC-injured animals. The neural-derived peptides A91 and Cop-1 were used to immunize mice and rats with SC injury. In vitro studies showed that INDP significantly reduces the production of NO by glial cells. This observation was substantiated by in vivo experiments demonstrating that INDP decreases the amount of NO and iNOS gene expression at the site of injury. The present study provides substantial evidence on the inhibitory effect of INDP on NO production, helpingour understanding of the mechanisms through which protective autoimmunity promotes neuroprotection.
Collapse
Affiliation(s)
- Elisa García
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Huixquilucan Edo. de México, México
| | | | | | | | | | | | | |
Collapse
|
44
|
Wang YT, Lu XM, Zhu F, Huang P, Yu Y, Zeng L, Long ZY, Wu YM. The use of a gold nanoparticle-based adjuvant to improve the therapeutic efficacy of hNgR-Fc protein immunization in spinal cord-injured rats. Biomaterials 2011; 32:7988-98. [PMID: 21784510 DOI: 10.1016/j.biomaterials.2011.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/04/2011] [Indexed: 10/18/2022]
Abstract
As a common receptor for three myelin associated inhibitors, Nogo-66 receptor (NgR) mediates their inhibitory activities on neurite outgrowth in the adult mammalian central nervous system (CNS). Therapeutic vaccination protocol targeting NgR emulsified with Freund's adjuvant (FA) has been used in spinal cord injury (SCI) models. However, the vaccine emulsified with FA may induce some side effects, which are not suitable for further clinical application. As an adjuvant, gold nanoparticles (GNPs) could stimulate a stronger immune response without producing detectable toxicity and physiological damage than FA. There is, however, uncertainty regarding the efficacy of axon regeneration and neuroprotection in vaccines with GNPs as an adjuvant. In this investigation, a recombinant protein vaccine targeting NgR, human NgR-Fc (hNgR-Fc) fusion protein conjugated with 15 nm GNPs was prepared and its effects on axonal regeneration and functional recovery in spinal cord-injured rats were investigated. The results showed that adult rats immunized with the protein vaccine produced higher titers of anti-NgR antibody than that with FA, and the antisera promoted neurite outgrowth in presence of MAG in vitro. In a spinal cord dorsal hemisection model, vaccine immunized with GNPs promoted axonal regeneration more effectively than FA, resulted in significant protection from neuronal loss, and improved functional recovery. Thus, as an adjuvant, 15 nm GNPs can effectively boost the immunogenicity of hNgR-Fc protein vaccine, and promote the repair of spinal cord-injured rats. The utilization of GNPs, for clinical considerations, may be a more beneficial supplement than FA to the promising therapeutic vaccination strategy for promoting SCI repair.
Collapse
Affiliation(s)
- Yong-Tang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Al Nimer F, Beyeen AD, Lindblom R, Ström M, Aeinehband S, Lidman O, Piehl F. Both MHC and non-MHC genes regulate inflammation and T-cell response after traumatic brain injury. Brain Behav Immun 2011; 25:981-90. [PMID: 20974248 DOI: 10.1016/j.bbi.2010.10.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 10/06/2010] [Accepted: 10/18/2010] [Indexed: 02/06/2023] Open
Abstract
Genetic regulation of autoimmune neuroinflammation is a well known phenomenon, but genetic influences on inflammation following traumatic nerve injuries have received little attention. In this study we examined the inflammatory response in a rat traumatic brain injury (TBI) model, with a particular focus on major histocompatibility class II (MHC II) presentation, in two inbred rat strains that have been extensively characterized in experimental autoimmune encephalomyelitis (EAE); DA and PVG. In addition, MHC and Vra4 congenic strains on these backgrounds were studied to give information on MHC and non-MHC gene contribution. Thus, allelic differences in Vra4, harboring the Ciita gene, was found to regulate expression of the invariant chain at the mRNA level, with a much smaller effect exerted by the MHC locus itself. Notably, however, at the protein level the MHC congenic PVG-RT1(av1) strain displayed much stronger MHCII(+) presentation, as shown both by immunolabeling and flow cytometry, than the PVG strain, dwarfing the effect of Ciita. The PVG-RT1(av1) strain had significantly more T-cell influx than both DA and PVG, suggesting regulation both by MHC and non-MHC genes. Finally, in terms of outcome, the EAE susceptible DA strain displayed a significantly smaller resulting lesion volume than the resistant PVG-RT1(av1) strain. These results provide additional support for a role of adaptive immune response after neurotrauma and demonstrate that outcome is significantly affected by host genetic factors.
Collapse
Affiliation(s)
- Faiez Al Nimer
- Department of Clinical Neuroscience, Karolinska University Hospital, S171 76 Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
46
|
Magharious M, D'Onofrio PM, Hollander A, Zhu P, Chen J, Koeberle PD. Quantitative iTRAQ analysis of retinal ganglion cell degeneration after optic nerve crush. J Proteome Res 2011; 10:3344-62. [PMID: 21627321 DOI: 10.1021/pr2004055] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Retinal ganglion cells (RGCs) are central nervous system (CNS) neurons that transmit visual information from the retina to the brain. Apoptotic RGC degeneration causes visual impairment that can be modeled by optic nerve crush. Neuronal apoptosis is also a salient feature of CNS trauma, ischemia (stroke), and diseases of the CNS such as Alzheimer's, Parkinson's, multiple sclerosis, and amyotrophic lateral sclerosis. Optic nerve crush induces the apoptotic cell death of ∼ 70% of RGCs within the first 14 days after injury. This model is particularly attractive for studying adult neuron apoptosis because the time-course of RGC death is well established and axon regeneration within the myelinated optic nerve can be concurrently evaluated. Here, we performed a large scale iTRAQ proteomic study to identify and quantify proteins of the rat retina at 1, 3, 4, 7, 14, and 21 days after optic nerve crush. In total, 337 proteins were identified, and 110 were differentially regulated after injury. Of these, 58 proteins were upregulated (>1.3 ×), 46 were downregulated (<0.7 ×), and 6 showed both positive and negative regulation over 21 days, relative to normal retinas. Among the differentially expressed proteins, Thymosin-β4 showed an early upregulation at 3 days, the time-point that immediately precedes the induction of RGC apoptosis after injury. We examined the effect of exogenous Thymosin-β4 administration on RGC death after optic nerve injury. Intraocular injections of Thymosin-β4 significantly increased RGC survival by ∼ 3-fold compared to controls and enhanced axon regeneration after crush, demonstrating therapeutic potential for CNS insults. Overall, our study identified numerous proteins that are differentially regulated at key time-points after optic nerve crush, and how the temporal profiles of their expression parallel RGC death. This data will aid in the future development of novel therapeutics to promote neuronal survival and regeneration in the adult CNS.
Collapse
Affiliation(s)
- Mark Magharious
- Graduate Department of Rehabilitation Science, University of Toronto, Canada
| | | | | | | | | | | |
Collapse
|
47
|
Preti A, Wilson DR. Schizophrenia, cancer and obstetric complications in an evolutionary perspective-an empirically based hypothesis. Psychiatry Investig 2011; 8:77-88. [PMID: 21852982 PMCID: PMC3149115 DOI: 10.4306/pi.2011.8.2.77] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 10/12/2010] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Patients diagnosed with schizophrenia have reduced fecundity and premature mortality (both accidental and violent) with no obvious compensatory advantages among kin. The prevalence of the disorder is around 0.7/1%, higher than the expected prevalence of spontaneous mutations. Genes favoring schizophrenia may have been positively selected in the environment of evolutionary adaptation. Literature on potential adaptive genes is reviewed within an evolutionary framework. METHODS Literature search on major scientific search engine (PubMed/Medline, Ovid/PsychInfo) on papers aimed at investigating potential pathways justifying a mutation-selection balanced model. Findings are presented with a narrative touch to favor readability and understanding. RESULTS Reduced incidence of cancer in both patients diagnosed with schizophrenia and their siblings was reported worldwide. Such findings are notable given higher cancer risk factors in schizophrenia, i.e., smoking, alcohol abuse, obesity, poor diet, and poor adherence to therapy. Some genes involved in cancer proliferation might as well confer protective advantage in immune-surveillance, inflammation, vascular proliferation or apoptosis that otherwise will adversely affect early neurodevelopment. CONCLUSION Evidence that reduced risk of certain somatic diseases is associated with schizophrenia is quite significant to progress in the evolutionary epidemiological analysis of psychopathology.
Collapse
Affiliation(s)
- Antonio Preti
- Centro Medico Genneruxi, via Costantinopoli 42, Cagliari, Italy
| | | |
Collapse
|
48
|
Huehnchen P, Prozorovski T, Klaissle P, Lesemann A, Ingwersen J, Wolf SA, Kupsch A, Aktas O, Steiner B. Modulation of adult hippocampal neurogenesis during myelin-directed autoimmune neuroinflammation. Glia 2010; 59:132-42. [DOI: 10.1002/glia.21082] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 08/31/2010] [Indexed: 01/04/2023]
|
49
|
Seksenyan A, Ron-Harel N, Azoulay D, Cahalon L, Cardon M, Rogeri P, Ko MK, Weil M, Bulvik S, Rechavi G, Amariglio N, Konen E, Koronyo-Hamaoui M, Somech R, Schwartz M. Thymic involution, a co-morbidity factor in amyotrophic lateral sclerosis. J Cell Mol Med 2010; 14:2470-82. [PMID: 19650830 PMCID: PMC3823164 DOI: 10.1111/j.1582-4934.2009.00863.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Accepted: 07/10/2009] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating disease, characterized by extremely rapid loss of motor neurons. Our studies over the last decade have established CD4(+) T cells as important players in central nervous system maintenance and repair. Those results, together with recent findings that CD4(+) T cells play a protective role in mouse models of ALS, led us to the current hypothesis that in ALS, a rapid T-cell malfunction may develop in parallel to the motor neuron dysfunction. Here, we tested this hypothesis by assessing thymic function, which serves as a measure of peripheral T-cell availability, in an animal model of ALS (mSOD1 [superoxide dismutase] mice; G93A) and in human patients. We found a significant reduction in thymic progenitor-cell content, and abnormal thymic histology in 3-4-month-old mSOD1 mice. In ALS patients, we found a decline in thymic output, manifested in the reduction in blood levels of T-cell receptor rearrangement excision circles, a non-invasive measure of thymic function, and demonstrated a restricted T-cell repertoire. The morbidity of the peripheral immune cells was also manifested in the increase of pro-apoptotic BAX/BCXL2 expression ratio in peripheral blood mononuclear cells (PBMCs) of these patients. In addition, gene expression screening in the same PBMCs, revealed in the ALS patients a reduction in key genes known to be associated with T-cell activity, including: CD80, CD86, IFNG and IL18. In light of the reported beneficial role of T cells in animal models of ALS, the present observation of thymic dysfunction, both in human patients and in an animal model, might be a co-pathological factor in ALS, regardless of the disease aetiology. These findings may lead to the development of novel therapeutic approaches directed at overcoming the thymic defect and T-cell deficiency.
Collapse
Affiliation(s)
- Akop Seksenyan
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| | - Noga Ron-Harel
- Department of Neurobiology, the Weizmann Institute of ScienceRehovot, Israel
| | - David Azoulay
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| | - Liora Cahalon
- Department of Neurobiology, the Weizmann Institute of ScienceRehovot, Israel
| | - Michal Cardon
- Department of Neurobiology, the Weizmann Institute of ScienceRehovot, Israel
| | - Patricia Rogeri
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| | - Minhee K Ko
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| | - Miguel Weil
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv UniversityTel Aviv, Israel
| | - Shlomo Bulvik
- Hematology Department, Laniado HospitalNetanya, Israel
| | - Gideon Rechavi
- Cancer Research Center, Sheba Medical Center, Tel Hashomer and Sackler School of MedicineTel Aviv, Israel
| | - Ninette Amariglio
- Cancer Research Center, Sheba Medical Center, Tel Hashomer and Sackler School of MedicineTel Aviv, Israel
| | - Eli Konen
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer and Sackler School of MedicineTel Aviv, Israel
| | - Maya Koronyo-Hamaoui
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| | - Raz Somech
- Cancer Research Center, Sheba Medical Center, Tel Hashomer and Sackler School of MedicineTel Aviv, Israel
- Pediatric Immunology Service, Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer and Sackler School of MedicineTel Aviv, Israel
| | - Michal Schwartz
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical CenterLos Angeles, CA, USA
- Department of Neurobiology, the Weizmann Institute of ScienceRehovot, Israel
| |
Collapse
|
50
|
T cells specifically targeted to amyloid plaques enhance plaque clearance in a mouse model of Alzheimer's disease. PLoS One 2010; 5:e10830. [PMID: 20520819 PMCID: PMC2877087 DOI: 10.1371/journal.pone.0010830] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Accepted: 05/03/2010] [Indexed: 01/07/2023] Open
Abstract
Patients with Alzheimer's disease (AD) exhibit substantial accumulation of amyloid-β (Aβ) plaques in the brain. Here, we examine whether Aβ vaccination can facilitate the migration of T lymphocytes to specifically target Aβ plaques and consequently enhance their removal. Using a new mouse model of AD, we show that immunization with Aβ, but not with the encephalitogenic proteolipid protein (PLP), results in the accumulation of T cells at Aβ plaques in the brain. Although both Aβ-reactive and PLP-reactive T cells have a similar phenotype of Th1 cells secreting primarily IFN-γ, the encephalitogenic T cells penetrated the spinal cord and caused experimental autoimmune encephalomyelitis (EAE), whereas Aβ T cells accumulated primarily at Aβ plaques in the brain but not the spinal cord and induced almost complete clearance of Aβ. Furthermore, while a single vaccination with Aβ resulted in upregulation of the phagocytic markers triggering receptors expressed on myeloid cells-2 (TREM2) and signal regulatory protein-β1 (SIRPβ1) in the brain, it caused downregulation of the proinflammatory cytokines TNF-α and IL-6. We thus suggest that Aβ deposits in the hippocampus area prioritize the targeting of Aβ-reactive but not PLP-reactive T cells upon vaccination. The stimulation of Aβ-reactive T cells at sites of Aβ plaques resulted in IFN-γ-induced chemotaxis of leukocytes and therapeutic clearance of Aβ.
Collapse
|