1
|
Rosiewicz KS, Muinjonov B, Kunz S, Radbruch H, Chen J, Jüttner R, Kerkering J, Ucar J, Crowley T, Wielockx B, Paul F, Alisch M, Siffrin V. HIF prolyl hydroxylase 2/3 deletion disrupts astrocytic integrity and exacerbates neuroinflammation. Glia 2023. [PMID: 37140003 DOI: 10.1002/glia.24380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023]
Abstract
Astrocytes constitute the parenchymal border of the blood-brain barrier (BBB), modulate the exchange of soluble and cellular elements, and are essential for neuronal metabolic support. Thus, astrocytes critically influence neuronal network integrity. In hypoxia, astrocytes upregulate a transcriptional program that has been shown to boost neuroprotection in several models of neurological diseases. We investigated transgenic mice with astrocyte-specific activation of the hypoxia-response program by deleting the oxygen sensors, HIF prolyl-hydroxylase domains 2 and 3 (Phd2/3). We induced astrocytic Phd2/3 deletion after onset of clinical signs in experimental autoimmune encephalomyelitis (EAE) that led to an exacerbation of the disease mediated by massive immune cell infiltration. We found that Phd2/3-ko astrocytes, though expressing a neuroprotective signature, exhibited a gradual loss of gap-junctional Connexin-43 (Cx43), which was induced by vascular endothelial growth factor-alpha (Vegf-a) expression. These results provide mechanistic insights into astrocyte biology, their critical role in hypoxic states, and in chronic inflammatory CNS diseases.
Collapse
Affiliation(s)
- Kamil Sebastian Rosiewicz
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Bakhrom Muinjonov
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Séverine Kunz
- Technology Platform for Electron Microscopy, Max Delbrück Centre for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Helena Radbruch
- Department of Neuropathology, Charité-Universitätsmedizin Berlin., Berlin, Germany
| | - Jessy Chen
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Neurology, Charité Universitätsmedizin Berlin., Berlin, Germany
| | - René Jüttner
- Neuromuscular and Cardiovascular Cell Biology Group, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Janis Kerkering
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Julia Ucar
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Tadhg Crowley
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden., Dresden, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Marlen Alisch
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Volker Siffrin
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin und Max Delbrück Center or Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Neurology, Charité Universitätsmedizin Berlin., Berlin, Germany
| |
Collapse
|
2
|
Manjili MH. The adaptation model of immunity: A new insight into aetiology and treatment of multiple sclerosis. Scand J Immunol 2023; 97:e13255. [PMID: 36680379 DOI: 10.1111/sji.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/04/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Current research and drug development for multiple sclerosis (MS) is fully influenced by the self-nonself (SNS) model of immunity, suggesting that breakage of immunological tolerance towards self-antigens expressed in the central nervous system (CNS) is responsible for pathogenesis of MS; thus, immune suppressive drugs are recommended for the management of the disease. However, this model provides incomplete understanding of the causes and pathways involved in the onset and progression of MS and limits our ability to effectively treat this neurological disease. Some pre-clinical and clinical reports have been misunderstood; some others have been underappreciated because of the lack of a theoretical model that can explain them. Also, current immunotherapies are guided according to the models that are not designed to explain the functional outcomes of an immune response. The adaptation model of immunity is proposed to offer a new understanding of the existing data and galvanize a new direction for the treatment of MS. According to this model, the immune system continuously communicates with the CNS through the adaptation receptors (AdRs) and adaptation ligands (AdLs) or co-receptors, signal IV, to support cell growth and neuroplasticity. Alterations in the expression of the neuronal AdRs results in MS by shifting the cerebral inflammatory immune responses from remyelination to demyelination. Therefore, novel therapeutics for MS should be focused on the discovery and targeting of the AdR/AdL axis in the CNS rather than carrying on with immune suppressive interventions.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Massey Cancer Center, Richmond, Virginia, USA
| |
Collapse
|
3
|
Tang H, Gu Y, Jiang L, Zheng G, Pan Z, Jiang X. The role of immune cells and associated immunological factors in the immune response to spinal cord injury. Front Immunol 2023; 13:1070540. [PMID: 36685599 PMCID: PMC9849245 DOI: 10.3389/fimmu.2022.1070540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological condition prevalent worldwide. Where the pathological mechanisms underlying SCI are concerned, we can distinguish between primary injury caused by initial mechanical damage and secondary injury characterized by a series of biological responses, such as vascular dysfunction, oxidative stress, neurotransmitter toxicity, lipid peroxidation, and immune-inflammatory response. Secondary injury causes further tissue loss and dysfunction, and the immune response appears to be the key molecular mechanism affecting injured tissue regeneration and functional recovery from SCI. Immune response after SCI involves the activation of different immune cells and the production of immunity-associated chemicals. With the development of new biological technologies, such as transcriptomics, the heterogeneity of immune cells and chemicals can be classified with greater precision. In this review, we focus on the current understanding of the heterogeneity of these immune components and the roles they play in SCI, including reactive astrogliosis and glial scar formation, neutrophil migration, macrophage transformation, resident microglia activation and proliferation, and the humoral immunity mediated by T and B cells. We also summarize findings from clinical trials of immunomodulatory therapies for SCI and briefly review promising therapeutic drugs currently being researched.
Collapse
Affiliation(s)
- Huaguo Tang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yuanjie Gu
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Lei Jiang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Gang Zheng
- Department of Neurosurgery, The Central Hospital Affiliated to Shaoxing University, Jiaxing, China
| | - Zhuoer Pan
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xiugui Jiang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
4
|
7,8-Dihydroxyflavone accelerates recovery of Brown-Sequard syndrome in adult female rats with spinal cord lateral hemisection. Biomed Pharmacother 2022; 153:113397. [DOI: 10.1016/j.biopha.2022.113397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
|
5
|
Moriya S, Hanazono M, Fukuhara T, Iwase K, Hattori N, Takiguchi M. A53T mutant α-synuclein fibrils formed in macrophage are spread to neurons. Cell Mol Life Sci 2022; 79:234. [PMID: 35397671 PMCID: PMC11073293 DOI: 10.1007/s00018-022-04263-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 11/03/2022]
Abstract
Lewy body (LB), which mainly consists of abnormal α-synuclein (αS) aggregates, is a histological hallmark of Parkinson's disease (PD). αS aggregation and LB inclusions are induced by spreading αS fibrils to neurons; therefore, the formation and transmission of αS fibrils to neurons may play an essential role in initiating LB formation in neurons. αS expressed in neurons is released into the extracellular space and taken up by macrophages and microglia; therefore, we hypothesized that macrophages/microglia play a role in the formation and spread of αS fibrils. In this study, we aimed to investigate the involvement of macrophages/microglia in the formation and spread of αS fibrils using transgenic animals that express human αS in macrophages/microglia. Transgenic zebrafish expressing A53T mutated αS (αS_A53T) in macrophages/microglia revealed αS accumulation in neurons. Transcriptome analysis by RNA-seq of human αS and αS_A53T expressing zebrafish revealed that kinase genes and E3 ubiquitin protein ligase genes were significantly high, and neuronal activity and transport-related Gene Ontology terms were also isolated. Meanwhile, αS_A53T monomers were taken up by A-THP-1 cells; processed to larger molecules, which could be αS fibrils; and released from macrophage cells. Furthermore, the ubiquitin-proteasome system modulated αS fibrils in A-THP-1 cells. αS fibrils suggest being formed from monomers in macrophages and spread to neurons to induce αS aggregates. Therefore, macrophages may play an essential role in the formation of αS aggregates and the pathogenesis of PD.
Collapse
Affiliation(s)
- Shogo Moriya
- Department of Biochemistry and Genetics, Chiba University Graduate School of Medicine, Chiba, Japan.
| | - Michiko Hanazono
- Department of Biochemistry and Genetics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takeshi Fukuhara
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| | - Katsuro Iwase
- Department of Biochemistry and Genetics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| | - Masaki Takiguchi
- Department of Biochemistry and Genetics, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
6
|
New Paradigm in Cell Therapy Using Sperm Head to Restore Brain Function and Structure in Animal Model of Alzheimer’s Disease: Support for Boosting Constructive Inflammation vs. Anti-Inflammatory Approach. J Immunol Res 2022; 2022:8343763. [PMID: 35571563 PMCID: PMC9095412 DOI: 10.1155/2022/8343763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/25/2022] [Accepted: 03/10/2022] [Indexed: 11/18/2022] Open
Abstract
Alzheimer’s is characterized by accumulation of amyloid-β (Aβ) associated with insufficient clearance of toxicants from the brain establishing a chronic inflammation and other abnormalities in the brain. Inflammatory microglia and astrocytes along with abnormal lymphatics associated with insufficient clearance of Aβ and other toxicants from the brain establish a chronic inflammation. This causes abnormal choroid plexus, leukocyte trafficking, and hypoxic condition along with high levels of regulatory T cells (Tregs). There is no consensus among researchers regarding decreasing or increasing Tregs to achieve therapeutic effects. Different opposing studies tried to suppress or boost inflammation to treat AD. Based on reproductive immunology, sperm induces constructive inflammatory response and seminal-vesicle-fluid (SVF) suppresses inflammation leading to uterus remodeling. It prompted us to compare therapeutic efficiency of inflammatory or anti-inflammatory approaches in AD model based on reproductive immunology. To do so, SVF, sperm, or sperm head (from Wistar rat) was administered via intra-cerebro-ventricular route to Sprague Dawley rat AD model. Behavioral and histological examination were made and treatment groups were compared with control AD model and normal groups. Therapeutic efficacy was in the order of sperm head>sperm>SVF. Sperm head returned learning memory, Aβ, lymphatics, neural growth factors, choroid plexus function, Iba-1/GFAP, MHC II/CD86/CD40, CD38/IL-10, and hypoxia levels back to normal level. However, SVF just partially ameliorated the disease. Immunologic properties of sperm/sperm head to elicit constructive inflammation can be extended to organs other than reproductive. This nature-based approach overcomes genetic difference as an important obstacle and limitation in cell therapy, and is expected to be safe or with least side effects.
Collapse
|
7
|
Cantuti-Castelvetri L, Gokce O, Simons M. Reparative inflammation in multiple sclerosis. Semin Immunol 2022; 59:101630. [PMID: 35750551 DOI: 10.1016/j.smim.2022.101630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Affiliation(s)
- Ludovico Cantuti-Castelvetri
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
8
|
Huang CT, Wen YT, Desai TD, Tsai RK. Intravitreal Injection of Long-Acting Pegylated Granulocyte Colony-Stimulating Factor Provides Neuroprotective Effects via Antioxidant Response in a Rat Model of Traumatic Optic Neuropathy. Antioxidants (Basel) 2021; 10:1934. [PMID: 34943037 PMCID: PMC8750325 DOI: 10.3390/antiox10121934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022] Open
Abstract
Traumatic optic neuropathy (TON) may cause severe visual loss following direct or indirect head trauma which may result in optic nerve injuries and therefore contribute to the subsequent loss of retinal ganglion cells by inflammatory mediators and reactive oxygen species (ROS). Granulocyte colony-stimulating factor (G-CSF) provides the anti-inflammatory and anti-oxidative actions but has a short half-life and also induces leukocytosis upon typical systemic administration. The purpose of the present study was to investigate the relationship between the anti-oxidative response and neuroprotective effects of long-acting pegylated human G-CSF (PEG-G-CSF) in a rat model of optic nerve crush (ONC). Adult male Wistar rats (150-180 g) were chosen to have a sham operation in one eye and have ONC in the other. PEG-G-CSF or phosphate-buffered saline (PBS control) was immediately administered after ONC by intravitreal injection (IVI). We found the IVI of PEG-G-CSF does not induce systemic leukocytosis, but increases survival of RGCs and preserves the visual function after ONC. TUNEL assays showed fewer apoptotic cells in the retina in the PEG-G-CSF-treated eyes. The number of sorely ED1-positive cells was attenuated at the lesion site in the PEG-G-CSF-treated eyes. Immunoblotting showed up-regulation of p-Akt1, Nrf2, Sirt3, and HO-1 in the ON of the PEG-G-CSF-treated eyes. Our results demonstrated that one IVI of long-acting PEG-G-CSF is neuroprotective in the rONC. PEG-G-CSF activates the p-Akt1/Nrf2/Sirt3 and the p-Akt1/Nrf2/HO-1 axes to provide the antioxidative action and further attenuated RGC apoptosis and neuroinflammation. This provides crucial preclinical information for the development of alternative therapy with IVI of PEG-G-CSF in TON.
Collapse
Affiliation(s)
- Chin-Te Huang
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
- Department of Ophthalmology, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
| | - Tushar Dnyaneshwar Desai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
| | - Rong-Kung Tsai
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien 970, Taiwan
| |
Collapse
|
9
|
Palumbo ML, Moroni AD, Quiroga S, Castro MM, Burgueño AL, Genaro AM. Immunomodulation induced by central nervous system-related peptides as a therapeutic strategy for neurodegenerative disorders. Pharmacol Res Perspect 2021; 9:e00795. [PMID: 34609083 PMCID: PMC8491457 DOI: 10.1002/prp2.795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/23/2021] [Indexed: 12/29/2022] Open
Abstract
Neurodegenerative diseases (NDD) are disorders characterized by the progressive loss of neurons affecting motor, sensory, and/or cognitive functions. The incidence of these diseases is increasing and has a great impact due to their high morbidity and mortality. Unfortunately, current therapeutic strategies only temporarily improve the patients' quality of life but are insufficient for completely alleviating the symptoms. An interaction between the immune system and the central nervous system (CNS) is widely associated with neuronal damage in NDD. Usually, immune cell infiltration has been identified with inflammation and is considered harmful to the injured CNS. However, the immune system has a crucial role in the protection and regeneration of the injured CNS. Nowadays, there is a consensus that deregulation of immune homeostasis may represent one of the key initial steps in NDD. Dr. Michal Schwartz originally conceived the concept of "protective autoimmunity" (PA) as a well-controlled peripheral inflammatory reaction after injury, essential for neuroprotection and regeneration. Several studies suggested that immunizing with a weaker version of the neural self-antigen would generate PA without degenerative autoimmunity. The development of CNS-related peptides with immunomodulatory neuroprotective effect led to important research to evaluate their use in chronic and acute NDD. In this review, we refer to the role of PA and the potential applications of active immunization as a therapeutic option for NDD treatment. In particular, we focus on the experimental and clinical promissory findings for CNS-related peptides with beneficial immunomodulatory effects.
Collapse
Affiliation(s)
- María Laura Palumbo
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA)‐UNNOBA‐UNsADA‐CONICETJunínArgentina
| | - Alejandro David Moroni
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA)‐UNNOBA‐UNsADA‐CONICETJunínArgentina
| | - Sofía Quiroga
- Instituto de Investigaciones BiomédicasConsejo Nacional de Investigaciones Científicas y Técnicas (CONICETPontificia Universidad Católica ArgentinaBuenos AiresArgentina
| | - María Micaela Castro
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA)‐UNNOBA‐UNsADA‐CONICETJunínArgentina
| | - Adriana Laura Burgueño
- Instituto de Investigaciones BiomédicasConsejo Nacional de Investigaciones Científicas y Técnicas (CONICETPontificia Universidad Católica ArgentinaBuenos AiresArgentina
| | - Ana María Genaro
- Instituto de Investigaciones BiomédicasConsejo Nacional de Investigaciones Científicas y Técnicas (CONICETPontificia Universidad Católica ArgentinaBuenos AiresArgentina
| |
Collapse
|
10
|
Geng Y, Lu Z, Guan J, van Rooijen N, Zhi Y. Microglia/Macrophages and CD4 +CD25 + T Cells Enhance the Ability of Injury-Activated Lymphocytes to Reduce Traumatic Optic Neuropathy In Vitro. Front Immunol 2021; 12:687898. [PMID: 34484185 PMCID: PMC8414969 DOI: 10.3389/fimmu.2021.687898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/29/2021] [Indexed: 02/05/2023] Open
Abstract
Inflammation after acute CNS injury plays a dual role. The interplay between immune cells and inflammatory mediators is critical to the outcome of injured neurons. Microglia/macrophages are the first sensors and regulators of the immune response. We previously found that the enhancement of macrophages on neuron survival does not persist in thymectomized rats. How T lymphocytes and macrophages interact and benefit neuron survival is not fully elucidated. To this point, we introduce and characterize a cell-retina co-culture model that mimics the recruitment of peripheral lymphocytes at the injury site. Three-day post-optic nerve transection (ONT) in Fischer 344 rats, transected retinas were co-cultured with either peripheral lymph node-derived lymphocytes (injury-activated) or from intact rats as the control. The injury-activated lymphocytes preserved retinal ganglion cells (RGCs) and caused extensive retina microglial/macrophage infiltration. CD4+CD25+ T cells were upregulated in the injury-activated lymphocytes and increased RGC survival, suggesting that CD4+CD25+ T cells suppressed the cytotoxicity of control lymphocytes. When microglia/macrophages were depleted by clodronate, neuron loss was more extensive, the cytotoxicity of control lymphocytes on RGCs was alleviated, and the neuroprotective effect of injury-activated lymphocytes remain unchanged Cytokine detection showed an increase in IL-6 and TNF-α levels that were reduced with microglia/macrophage depletion. Our results suggest that microglial/macrophage infiltration into axotomized retinas promotes RGC survival by secreting cytokines to induce CD4+CD25+ T cells and suppress T cell-mediated RGC toxicity. These findings reveal a specific role for microglia/macrophage and CD4+CD25+ T cells in inflammation after CNS injury, thereby adding to the mechanistic basis for the development of microglial/macrophage modulation therapy for traumatic CNS injury.
Collapse
Affiliation(s)
- Yiqun Geng
- Laboratory of Molecular Pathology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou University Medical College, Shantou, China
| | - Zhihao Lu
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou University Medical College, Shantou, China
| | - Jitian Guan
- Department of MRI, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center, Amsterdam, Netherlands
| | - Ye Zhi
- Department of Anatomy, Shantou University Medical College, Shantou, China
| |
Collapse
|
11
|
Schmidt E, Raposo P, Vavrek R, Fouad K. Inducing inflammation following subacute spinal cord injury in female rats: A double-edged sword to promote motor recovery. Brain Behav Immun 2021; 93:55-65. [PMID: 33358981 DOI: 10.1016/j.bbi.2020.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/20/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022] Open
Abstract
The inflammatory response following spinal cord injury is associated with increased tissue damage and impaired functional recovery. However, inflammation can also promote plasticity and the secretion of growth-promoting substances. Previously we have shown that inducing inflammation with a systemic injection of lipopolysaccharide in the chronic (8 weeks) stage of spinal cord injury enhances neuronal sprouting and the efficacy of rehabilitative training in rats. Here, we tested whether administration of lipopolysaccharide in female rats in the subacute (10 days) stage of spinal cord injury would have a similar effect. Since the lesioned environment is already in a pro-inflammatory state at this earlier time after injury, we hypothesized that triggering a second immune response may not be beneficial for recovery. Contrary to our hypothesis, we found that eliciting an inflammatory response 10 days after spinal cord injury enhanced the recovery of the ipsilesional forelimb in rehabilitative training. Compared to rats that received rehabilitative training without treatment, rats that received systemic lipopolysaccharide showed restored motor function without the use of compensatory strategies that translated beyond the trained task. Furthermore, lipopolysaccharide treatment paradoxically promoted the resolution of chronic neuroinflammation around the lesion site. Unfortunately, re-triggering a systemic immune response after spinal cord injury also resulted in a long-term increase in anxiety-like behaviour.
Collapse
Affiliation(s)
- Emma Schmidt
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Pamela Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada.
| |
Collapse
|
12
|
Golan M, Krivitsky A, Mausner-Fainberg K, Benhamou M, Vigiser I, Regev K, Kolb H, Karni A. Increased Expression of Ephrins on Immune Cells of Patients with Relapsing Remitting Multiple Sclerosis Affects Oligodendrocyte Differentiation. Int J Mol Sci 2021; 22:ijms22042182. [PMID: 33671716 PMCID: PMC7927032 DOI: 10.3390/ijms22042182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/02/2022] Open
Abstract
The effect of the inflammatory response on regenerative processes in the brain is complex. This complexity is even greater when the cause of the tissue damage is an autoimmune response. Multiple sclerosis (MS) is an immune-mediated disease in which demyelination foci are formed in the central nervous system. The degree of repair through oligodendrocyte regeneration and remyelination is insufficient. Ephrins are membrane-bound ligands activating tyrosine kinase signaling proteins that are known to have an inhibitory effect on oligodendrocyte regeneration. In this study, we examined the expression of ephrins on immune cells of 43 patients with relapsing-remitting (RR) MS compared to 27 matched healthy controls (HC). We found an increased expression of ephrin-A2, -A3 and -B3, especially on T cell subpopulations. We also showed overexpression of ephrins on immune cells of patients with RR-MS that increases the forward signaling pathway and that expression of ephrins on immune cells has an inhibitory effect on the differentiation of oligodendrocyte precursor cells (OPCs) in vitro. Our study findings support the concept that the immune activity of T cells in patients with RR-MS has an inhibitory effect on the differentiation capacity of OPCs through the expression and forward signaling of ephrins.
Collapse
Affiliation(s)
- Maya Golan
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Avivit Krivitsky
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Karin Mausner-Fainberg
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Moshe Benhamou
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ifat Vigiser
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Keren Regev
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Hadar Kolb
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Arnon Karni
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
13
|
Macrophage migration inhibitory factor as a therapeutic target after traumatic spinal cord injury: a systematic review. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2021; 30:1474-1494. [PMID: 33486594 DOI: 10.1007/s00586-021-06718-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/25/2020] [Accepted: 01/01/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Macrophages play an important role in mediating damage after Spinal cord injury (SCI) by secreting macrophage migration inhibitory factor (MMIF) as a secondary injury mediator. We aimed to systematically review the role of MMIF as a therapeutic target after traumatic SCI. METHODS Our systematic review has been performed according to the PRISMA 2009 Checklist. A systematic search in the scientific databases was carried out for studies published before 20 February 2019 from major databases. Two researchers independently screened titles. The risk of bias of eligible articles was assessed, and data were extracted. Finally, we systematically analyzed and interpreted related data. RESULTS 785 papers were selected for the title and abstract screening. 12 papers were included for data extraction. Eight animal studies were of high quality and the remaining two were of medium quality. One of the two human studies was of poor quality and the other was of fair quality. MMIF as a pro-inflammatory mediator can cause increased susceptibility to glutamate-related neurotoxicity, increased nitrite production, increased ERK activation, and increased COX2/PGE2 signaling pathway activation and subsequent stimulation of CCL5-related chemotaxis. Two human studies and six animal studies demonstrated that MMIF level increases after SCI. MMIF inhibition might be a potential therapeutic target in SCI by multiple different mechanisms (6/12 studies). CONCLUSION Most animal studies demonstrate significant neurologic improvement after administration of MMIF inhibitors, but these inhibitors have not been studied in humans yet. Further clinical trials are need to further understand MMIF inhibitor utility in acute or chronic SCI. LEVEL OF EVIDENCE I Diagnostic: individual cross-sectional studies with the consistently applied reference standard and blinding.
Collapse
|
14
|
Chalons P, Courtaut F, Limagne E, Chalmin F, Cantos-Villar E, Richard T, Auger C, Chabert P, Schini-Kerth V, Ghiringhelli F, Aires V, Delmas D. Red Wine Extract Disrupts Th17 Lymphocyte Differentiation in a Colorectal Cancer Context. Mol Nutr Food Res 2020; 64:e1901286. [PMID: 32306526 DOI: 10.1002/mnfr.201901286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/18/2020] [Indexed: 12/13/2022]
Abstract
SCOPE Scope: It is well established that immune response and inflammation promote tumoral progression. Immune cells communicate through direct contact or through cytokine secretion, and it is the pro-inflammatory status that will tip the balance toward tumor progression or anti-tumor immunity. It is demonstrated here that a red wine extract (RWE) can decrease inflammation through its action on the inflammasome complex. This study determines whether an RWE could impact other key actors of inflammation, including T helper 17 (Th17) immune cells in particular. METHODS AND RESULTS Methods and results: Using an RWE containing 4.16 g of polyphenols/liter of wine, it is shown that RWE decreases colorectal cancer cells in vitro and induces a reduction in colorectal tumor growth associated with a decrease in tumor-infiltrating lymphocytes in vivo. The process of T-lymphocyte differentiation in Th17 cells is altered by RWE, as revealed by the decrease in the expression of key actors controlling this process, such as signal transducer and activator of transcription 3 and retinoid acid-related orphan receptor γt. This disruption is associated with an inhibition of inflammatory interleukin 17 secretion. CONCLUSION The data highlights the major involvement of Th17 immune cells in the biological effects of an RWE.
Collapse
Affiliation(s)
- Pauline Chalons
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Flavie Courtaut
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Emeric Limagne
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Plateforme de Transfert en Biologie du Cancer (PTBC) Centre Georges François Leclerc, Dijon, F-21000, France
| | - Fanny Chalmin
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Emma Cantos-Villar
- Instituto de Investigación y Formación Agraria y Pesquera (IFAPA) Rancho de La Merced, Ctra. Trebujena, 11.471 Jerez de la Frontera, Cadiz, Spain
| | - Tristan Richard
- Université de Bordeaux, Unité de Recherche Œnologie, EA 4577, USC 1366 INRA, Equipe Molécules d'Intérêt Biologique - ISVV, Villenave-d'Ornon, F-33882, France
| | - Cyril Auger
- UMR 1260 INSERM Nanomédecine Régénérative, Université de Strasbourg, Illkirch, F-67401, France
| | - Philippe Chabert
- UMR CNRS 7021 - Laboratoire de Bioimagerie et Pathologies, Université de Strasbourg, Illkirch-Graffenstaden, F-67401, France
| | - Valérie Schini-Kerth
- UMR 1260 INSERM Nanomédecine Régénérative, Université de Strasbourg, Illkirch, F-67401, France
| | - François Ghiringhelli
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Plateforme de Transfert en Biologie du Cancer (PTBC) Centre Georges François Leclerc, Dijon, F-21000, France.,Department of Medical Oncology, Centre Georges François Leclerc, Dijon, F-21000, France
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Department of Medical Oncology, Centre Georges François Leclerc, Dijon, F-21000, France
| |
Collapse
|
15
|
Yip PK, Bowes AL, Hall JCE, Burguillos MA, Ip THR, Baskerville T, Liu ZH, Mohamed MAEK, Getachew F, Lindsay AD, Najeeb SUR, Popovich PG, Priestley JV, Michael-Titus AT. Docosahexaenoic acid reduces microglia phagocytic activity via miR-124 and induces neuroprotection in rodent models of spinal cord contusion injury. Hum Mol Genet 2020; 28:2427-2448. [PMID: 30972415 DOI: 10.1093/hmg/ddz073] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/31/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022] Open
Abstract
Microglia are activated after spinal cord injury (SCI), but their phagocytic mechanisms and link to neuroprotection remain incompletely characterized. Docosahexaenoic acid (DHA) has been shown to have significant neuroprotective effects after hemisection and compression SCI and can directly affect microglia in these injury models. In rodent contusion SCI, we demonstrate that DHA (500 nmol/kg) administered acutely post-injury confers neuroprotection and enhances locomotor recovery, and also exerts a complex modulation of the microglial response to injury. In rodents, at 7 days after SCI, the level of phagocytosed myelin within Iba1-positive or P2Y12-positive cells was significantly lower after DHA treatment, and this occurred in parallel with an increase in intracellular miR-124 expression. Furthermore, intraspinal administration of a miR-124 inhibitor significantly reduced the DHA-induced decrease in myelin phagocytosis in mice at 7 days post-SCI. In rat spinal primary microglia cultures, DHA reduced the phagocytic response to myelin, which was associated with an increase in miR-124, but not miR-155. A similar response was observed in a microglia cell line (BV2) treated with DHA, and the effect was blocked by a miR-124 inhibitor. Furthermore, the phagocytic response of BV2 cells to stressed neurones was also reduced in the presence of DHA. In peripheral monocyte-derived macrophages, the expression of the M1, but not the M0 or M2 phenotype, was reduced by DHA, but the phagocytic activation was not altered. These findings show that DHA induces neuroprotection in contusion injury. Furthermore, the improved outcome is via a miR-124-dependent reduction in the phagocytic response of microglia.
Collapse
Affiliation(s)
- Ping K Yip
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Amy L Bowes
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jodie C E Hall
- Centre for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - Miguel A Burguillos
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla and, Sevilla, Spain
| | - T H Richard Ip
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tracey Baskerville
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Zhuo-Hao Liu
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Chang Gung Medical College and University, Chang Gung Memorial Hospital, Department of Neurosurgery, 5 Fu-Shin Street, Linkou, Taiwan
| | - Moumin A E K Mohamed
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Fanuelle Getachew
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anna D Lindsay
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Saif-Ur-Rehman Najeeb
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Phillip G Popovich
- Centre for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - John V Priestley
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
16
|
Grossman Z. Immunological Paradigms, Mechanisms, and Models: Conceptual Understanding Is a Prerequisite to Effective Modeling. Front Immunol 2019; 10:2522. [PMID: 31749803 PMCID: PMC6848063 DOI: 10.3389/fimmu.2019.02522] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022] Open
Abstract
Most mathematical models that describe the individual or collective actions of cells aim at creating faithful representations of limited sets of data in a self-consistent manner. Consistency with relevant physiological rules pertaining to the greater picture is rarely imposed. By themselves, such models have limited predictive or even explanatory value, contrary to standard claims. Here I try to show that a more critical examination of currently held paradigms is necessary and could potentially lead to models that pass the test of time. In considering the evolution of paradigms over the past decades I focus on the “smart surveillance” theory of how T cells can respond differentially, individually and collectively, to both self- and foreign antigens depending on various “contextual” parameters. The overall perspective is that physiological messages to cells are encoded not only in the biochemical connections of signaling molecules to the cellular machinery but also in the magnitude, kinetics, and in the time- and space-contingencies, of sets of stimuli. By rationalizing the feasibility of subthreshold interactions, the “dynamic tuning hypothesis,” a central component of the theory, set the ground for further theoretical and experimental explorations of dynamically regulated immune tolerance, homeostasis and diversity, and of the notion that lymphocytes participate in nonclassical physiological functions. Some of these efforts are reviewed. Another focus of this review is the concomitant regulation of immune activation and homeostasis through the operation of a feedback mechanism controlling the balance between renewal and differentiation of activated cells. Different perspectives on the nature and regulation of chronic immune activation in HIV infection have led to conflicting models of HIV pathogenesis—a major area of research for theoretical immunologists over almost three decades—and can have profound impact on ongoing HIV cure strategies. Altogether, this critical review is intended to constructively influence the outlook of prospective model builders and of interested immunologists on the state of the art and to encourage conceptual work.
Collapse
Affiliation(s)
- Zvi Grossman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Galloway DA, Gowing E, Setayeshgar S, Kothary R. Inhibitory milieu at the multiple sclerosis lesion site and the challenges for remyelination. Glia 2019; 68:859-877. [PMID: 31441132 DOI: 10.1002/glia.23711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/26/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
Regeneration of myelin, following injury, can occur within the central nervous system to reinstate proper axonal conductance and provide trophic support. Failure to do so renders the axons vulnerable, leading to eventual degeneration, and neuronal loss. Thus, it is essential to understand the mechanisms by which remyelination or failure to remyelinate occur, particularly in the context of demyelinating and neurodegenerative disorders. In multiple sclerosis, oligodendrocyte progenitor cells (OPCs) migrate to lesion sites to repair myelin. However, during disease progression, the ability of OPCs to participate in remyelination diminishes coincident with worsening of the symptoms. Remyelination is affected by a broad range of cues from intrinsic programming of OPCs and extrinsic local factors to the immune system and other systemic elements including diet and exercise. Here we review the literature on these diverse inhibitory factors and the challenges they pose to remyelination. Results spanning several disciplines from fundamental preclinical studies to knowledge gained in the clinic will be discussed.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Elizabeth Gowing
- Neurosciences Department, Faculty of Medicine, Centre de recherche du CHUM, Université de Montreal, Montreal, Quebec, Canada
| | - Solmaz Setayeshgar
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Department of Biochemistry, Microbiology and Immunology, and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Rieckmann M, Delgobo M, Gaal C, Büchner L, Steinau P, Reshef D, Gil-Cruz C, Horst ENT, Kircher M, Reiter T, Heinze KG, Niessen HW, Krijnen PA, van der Laan AM, Piek JJ, Koch C, Wester HJ, Lapa C, Bauer WR, Ludewig B, Friedman N, Frantz S, Hofmann U, Ramos GC. Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses. J Clin Invest 2019; 129:4922-4936. [PMID: 31408441 DOI: 10.1172/jci123859] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
T cell autoreactivity is a hallmark of autoimmune diseases but can also benefit self-maintenance and foster tissue repair. Herein, we investigated whether heart-specific T cells exert salutary or detrimental effects in the context of myocardial infarction (MI), the leading cause of death worldwide. After screening more than 150 class-II-restricted epitopes, we found that myosin heavy chain alpha (MYHCA) was a dominant cardiac antigen triggering post-MI CD4+ T cell activation in mice. Transferred MYHCA614-629-specific CD4+ T (TCR-M) cells selectively accumulated in the myocardium and mediastinal lymph nodes (med-LN) of infarcted mice, acquired a Treg phenotype with a distinct pro-healing gene expression profile, and mediated cardioprotection. Myocardial Treg cells were also detected in autopsies from patients who suffered a MI. Noninvasive PET/CT imaging using a CXCR4 radioligand revealed enlarged med-LNs with increased cellularity in MI-patients. Notably, the med-LN alterations observed in MI patients correlated with the infarct size and cardiac function. Taken together, the results obtained in our study provide evidence showing that MI-context induces pro-healing T cell autoimmunity in mice and confirms the existence of an analogous heart/med-LN/T cell axis in MI patients.
Collapse
Affiliation(s)
- Max Rieckmann
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany
| | - Murilo Delgobo
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Chiara Gaal
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Lotte Büchner
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Philipp Steinau
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany
| | - Dan Reshef
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Ellis N Ter Horst
- Heart Center, Amsterdam UMC, location AMC, Amsterdam, Netherlands.,Department of Pathology and Cardiac Surgery, Amsterdam UMC, location VUmc, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| | - Malte Kircher
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Theresa Reiter
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Hans Wm Niessen
- Department of Pathology and Cardiac Surgery, Amsterdam UMC, location VUmc, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands
| | - Paul Aj Krijnen
- Department of Pathology and Cardiac Surgery, Amsterdam UMC, location VUmc, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands
| | | | - Jan J Piek
- Heart Center, Amsterdam UMC, location AMC, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands
| | - Charlotte Koch
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technical University Munich, Munich, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Wolfgang R Bauer
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nir Friedman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Stefan Frantz
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany.,Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Ulrich Hofmann
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany.,Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Gustavo Campos Ramos
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany.,Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
Tian B, Yang C, Wang J, Hou X, Zhao S, Li Y, Yang P. Peripheral blood brain-derived neurotrophic factor level and tyrosine kinase B expression on T lymphocytes in systemic lupus erythematosus: Implications for systemic involvement. Cytokine 2019; 123:154764. [PMID: 31255912 DOI: 10.1016/j.cyto.2019.154764] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 06/05/2019] [Accepted: 06/17/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Brain-derived neurotrophic factor (BDNF) has been reported to be involved in the pathogenesis of autoimmune diseases and tyrosine kinase B (TrkB) is the specific receptor for BDNF. Our aim in this study was to investigate serum BDNF level and TrkB expression on peripheral blood T cell surface in patients with systemic lupus erythematosus (SLE) and explore potential relationship between serum BDNF and SLE. METHODS Samples from fifty SLE patients and thirty healthy controls were evaluated. Serum BDNF level was measured by enzyme-linked immunosorbent assay (ELISA) and the percentages of TrkB expression on the surface of CD3 + CD4 + and CD3 + CD8 + T lymphocytes were measured by flow cytometry. The SLE patients were divided into subgroups according to whether they exhibited brain, kidney or lung involvement, and whether the disease was active or inactive. RESULTS Serum BDNF levels in SLE patients were decreased when compared to the controls (p < 0.001). Comparing with the SLE individuals without systemic involvement, the BDNF levels were decreased in SLE patients with lupus nephritis (p = 0.042) and in SLE patients with neuropsychiatric manifestations (p = 0.04). On the other hand, the BDNF level was significantly increased in the inactive SLE group (p < 0.001) compared to the active SLE group. In addition, the percentages of TrkB expression on CD3 + CD4 + and CD3 + CD8 + T cell surface in SLE were significantly higher (p < 0.001; p < 0.001, respectively) than that in the controls. CONCLUSIONS Serum BDNF level combined with TrkB expression on T cell surface can reflect SLE activity. It is possible that BDNF may be used as a potential serological biomarker for disease activity of SLE. In addition, the significant decrease in serum BDNF level may imply systemic involvement of SLE, as well as, possibly, differentiate neuropsychiatric SLE from hormone-induced mental disorders.
Collapse
Affiliation(s)
- Bailing Tian
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Chunshu Yang
- Department of 1st Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Jianing Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Xiaoyu Hou
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Shan Zhao
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Yujia Li
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
20
|
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
|
21
|
Yong HYF, Rawji KS, Ghorbani S, Xue M, Yong VW. The benefits of neuroinflammation for the repair of the injured central nervous system. Cell Mol Immunol 2019; 16:540-546. [PMID: 30874626 DOI: 10.1038/s41423-019-0223-3] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammation of the nervous system (neuroinflammation) is now recognized as a hallmark of virtually all neurological disorders. In neuroinflammatory conditions such as multiple sclerosis, there is prominent infiltration and a long-lasting representation of various leukocyte subsets in the central nervous system (CNS) parenchyma. Even in classic neurodegenerative disorders, where such immense inflammatory infiltrates are absent, there is still evidence of activated CNS-intrinsic microglia. The consequences of excessive and uncontrolled neuroinflammation are injury and death to neural elements, which manifest as a heterogeneous set of neurological symptoms. However, it is now readily acknowledged, due to instructive studies from the peripheral nervous system and a large body of CNS literature, that aspects of the neuroinflammatory response can be beneficial for CNS outcomes. The recognized benefits of inflammation to the CNS include the preservation of CNS constituents (neuroprotection), the proliferation and maturation of various neural precursor populations, axonal regeneration, and the reformation of myelin on denuded axons. Herein, we highlight the benefits of neuroinflammation in fostering CNS recovery after neural injury using examples from multiple sclerosis, traumatic spinal cord injury, stroke, and Alzheimer's disease. We focus on CNS regenerative responses, such as neurogenesis, axonal regeneration, and remyelination, and discuss the mechanisms by which neuroinflammation is pro-regenerative for the CNS. Finally, we highlight treatment strategies that harness the benefits of neuroinflammation for CNS regenerative responses.
Collapse
Affiliation(s)
| | | | | | - Mengzhou Xue
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | |
Collapse
|
22
|
Farahtaj F, Alizadeh L, Gholami A, Tahamtan A, Shirian S, Fazeli M, Nejad ASM, Gorji A, Niknam HM, Ghaemi A. Natural Infection with Rabies Virus: A Histopathological and Immunohistochemical Study of Human Brains. Osong Public Health Res Perspect 2019; 10:6-11. [PMID: 30847265 PMCID: PMC6396821 DOI: 10.24171/j.phrp.2019.10.1.03] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objectives Despite all the efforts and increased knowledge of rabies, the exact mechanisms of infection and mortality from the rabies virus are not well understood. To understand the mechanisms underlying the pathogenicity of rabies virus infection, it is crucial to study the tissue that the rabies virus naturally infects in humans. Methods Cerebellum brain tissue from 9 human post mortem cases from Iran, who had been infected with rabies virus, were examined histopathologically and immunohistochemically to evaluate the innate immune responses against the rabies virus. Results Histopathological examination revealed inflammation of the infected cerebellum and immunohistochemical analyses showed an increased immunoreactivity of heat shock protein 70, interleukin-6, interleukin-1, tumor necrosis factor-alpha, caspase-3, caspase-9, toll-like receptor3 and toll-like receptor4 in the infected brain tissue. Conclusion These results indicated the involvement of innate immunity in rabies infected human brain tissue, which may aggravate the progression of this deadly disease.
Collapse
Affiliation(s)
- Firouzeh Farahtaj
- Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Leila Alizadeh
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Alireza Gholami
- Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Tahamtan
- Department of Virology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Maryam Fazeli
- Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Sasan Mozaffari Nejad
- Molecular Research Center, Student Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Neurosurgery and Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | - Amir Ghaemi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
de la Vega Gallardo N, Dittmer M, Dombrowski Y, Fitzgerald DC. Regenerating CNS myelin: Emerging roles of regulatory T cells and CCN proteins. Neurochem Int 2018; 130:104349. [PMID: 30513363 DOI: 10.1016/j.neuint.2018.11.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023]
Abstract
Efficient myelin regeneration in the central nervous system (CNS) requires the migration, proliferation and differentiation of oligodendrocyte progenitor cells (OPC) into myelinating oligodendrocytes. In demyelinating diseases such as multiple sclerosis (MS), this regenerative process can fail, and therapies targeting myelin repair are currently completely lacking in the clinic. The immune system is emerging as a key regenerative player in many tissues, such as muscle and heart. We recently reported that regulatory T cells (Treg) are required for efficient CNS remyelination. Furthermore, Treg secrete CCN3, a matricellular protein from the CCN family, implicated in regeneration of other tissues. Treg-derived CCN3 promoted oligodendrocyte differentiation and myelination. In contrast, previous studies showed that CCN2 inhibited myelination. These studies highlight the need for further scrutiny of the roles that CCN proteins play in myelin development and regeneration. Collectively, these findings open up exciting avenues of research to uncover the regenerative potential of the adaptive immune system.
Collapse
Affiliation(s)
- Nira de la Vega Gallardo
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Marie Dittmer
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Yvonne Dombrowski
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Denise C Fitzgerald
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK.
| |
Collapse
|
24
|
Suner HI, Kurt G, Yildirim Z, Bulduk EB, Borcek AO, Demirci H, Kartal B, Kaplanoglu GT. Investigation of the Effect of Alemtuzumab in an Experimental Spinal Cord Trauma Model in Rats. World Neurosurg 2018; 121:e723-e730. [PMID: 30292667 DOI: 10.1016/j.wneu.2018.09.197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Spinal cord injuries generate the most negative response to medical treatment among all general body injuries. This important morbidity is thought to be caused by a complex secondary damage mechanism. In the present study, we examined the neuroprotective effects of alemtuzumab in a spinal cord trauma model. METHODS We divided 24 Long-Evans male rats into 4 groups (n = 6 per group). Laminectomy was performed at T5-T8 in all groups. Trauma was applied using the Yasargil temporary aneurysm clip for 60 seconds at these spinal cord levels in all groups, except for group 1. Next, 1 mg/kg of alemtuzumab was administered to each rat in groups 3 and 4. A functional evaluation was performed on days 1, 3, and 5 in groups 1, 2, and 4, and the rats were then sacrificed. The rats in group 3 were sacrificed on the third postoperative day to observe the early effects of alemtuzumab. The biochemical examination findings of malondialdehyde and glutathione in plasma and tissue samples and histopathological findings of the spinal cord were evaluated and compared by statistical analysis. RESULTS The inflammatory findings in the trauma group were not seen in either group treated with alemtuzumab. The clinical motor examination and inclined plane test results were also significantly better in these groups. CONCLUSION Our results have shown that alemtuzumab might prevent spinal cord injury after trauma and is a histopathologically and biochemically strong anti-inflammatory, antioxidant, and neuroprotective agent.
Collapse
Affiliation(s)
- Halil Ibrahim Suner
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Gokhan Kurt
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | | | - Erkut Baha Bulduk
- Department of Neurosurgery, Eskisehir Government Hospital, Eskisehir, Turkey
| | - Alp Ozgun Borcek
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Harun Demirci
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Bahar Kartal
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Gulnur Take Kaplanoglu
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
25
|
Torres-Espín A, Forero J, Fenrich KK, Lucas-Osma AM, Krajacic A, Schmidt E, Vavrek R, Raposo P, Bennett DJ, Popovich PG, Fouad K. Eliciting inflammation enables successful rehabilitative training in chronic spinal cord injury. Brain 2018; 141:1946-1962. [PMID: 29860396 PMCID: PMC6022560 DOI: 10.1093/brain/awy128] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/09/2018] [Accepted: 03/28/2018] [Indexed: 01/24/2023] Open
Abstract
Rehabilitative training is one of the most successful therapies to promote motor recovery after spinal cord injury, especially when applied early after injury. Polytrauma and management of other medical complications in the acute post-injury setting often preclude or complicate early rehabilitation. Therefore, interventions that reopen a window of opportunity for effective motor training after chronic injury would have significant therapeutic value. Here, we tested whether this could be achieved in rats with chronic (8 weeks) dorsolateral quadrant sections of the cervical spinal cord (C4) by inducing mild neuroinflammation. We found that systemic injection of a low dose of lipopolysaccharide improved the efficacy of rehabilitative training on forelimb function, as assessed using a single pellet reaching and grasping task. This enhanced recovery was found to be dependent on the training intensity, where a high-intensity paradigm induced the biggest improvements. Importantly, in contrast to training alone, the combination of systemic lipopolysaccharide and high-intensity training restored original function (reparative plasticity) rather than enhancing new motor strategies (compensatory plasticity). Accordingly, electrophysiological and tract-tracing studies demonstrated a recovery in the cortical drive to the affected forelimb muscles and a restructuration of the corticospinal innervation of the cervical spinal cord. Thus, we propose that techniques that can elicit mild neuroinflammation may be used to enhance the efficacy of rehabilitative training after chronic spinal cord injury.
Collapse
Affiliation(s)
- Abel Torres-Espín
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Juan Forero
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Keith K Fenrich
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Ana M Lucas-Osma
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Aleksandra Krajacic
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Emma Schmidt
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Romana Vavrek
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Pamela Raposo
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - David J Bennett
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, 460 W. 12th Ave., 694 Biomedical Research Tower, Ohio State University; Columbus, Ohio, USA
| | - Karim Fouad
- Faculty of Rehabilitation Medicine, University of Alberta; Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta; Edmonton, Alberta, Canada
| |
Collapse
|
26
|
Mango D, Nisticò R, Furlan R, Finardi A, Centonze D, Mori F. PDGF Modulates Synaptic Excitability and Short-Latency Afferent Inhibition in Multiple Sclerosis. Neurochem Res 2018; 44:726-733. [PMID: 29392518 DOI: 10.1007/s11064-018-2484-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/19/2018] [Accepted: 01/23/2018] [Indexed: 12/25/2022]
Abstract
Maintenance of synaptic plasticity reserve is crucial to contrast clinical deterioration in MS and PDGF plays a key role in this phenomenon. Indeed, higher cerebrospinal fluid PDGF concentration correlates with improved clinical recovery after a relapse, and the amplitude of LTP-like cortical plasticity in relapsing-remitting MS patients. However, LTP-like cortical plasticity varies depending on the individual level of inhibitory cortical circuits. Aim of this study was to explore whether PDGF-CSF concentration correlates with inhibitory cortical circuits explored by means of transcranial magnetic stimulation in patients affected by relapsing-remitting MS. We further performed electrophysiological experiments evaluating GABAergic transmission in the experimental autoimmune encephalomyelitis (EAE) hippocampus. Our results reveal that increased CSF PDGF concentration correlates with decreased short afferent inhibition in the motor cortex in MS patients and decreased GABAergic activity in EAE. These findings show that PDGF affects GABAergic activity both in MS patients and in EAE hippocampus.
Collapse
Affiliation(s)
- Dalila Mango
- Neuropharmacology Unit, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| | - Robert Nisticò
- Neuropharmacology Unit, EBRI Rita Levi-Montalcini Foundation, Rome, Italy. .,Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Diego Centonze
- Neurology and Neurorehabilitation Units, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, IS, Italy. .,Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Francesco Mori
- Neurology and Neurorehabilitation Units, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, IS, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
27
|
Bajnok A, Berta L, Orbán C, Veres G, Zádori D, Barta H, Méder Ü, Vécsei L, Tulassay T, Szabó M, Toldi G. Distinct cytokine patterns may regulate the severity of neonatal asphyxia-an observational study. J Neuroinflammation 2017; 14:244. [PMID: 29233180 PMCID: PMC5727967 DOI: 10.1186/s12974-017-1023-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/05/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation and a systemic inflammatory reaction are important features of perinatal asphyxia. Neuroinflammation may have dual aspects being a hindrance, but also a significant help in the recovery of the CNS. We aimed to assess intracellular cytokine levels of T-lymphocytes and plasma cytokine levels in moderate and severe asphyxia in order to identify players of the inflammatory response that may influence patient outcome. METHODS We analyzed the data of 28 term neonates requiring moderate systemic hypothermia in a single-center observational study. Blood samples were collected between 3 and 6 h of life, at 24 h, 72 h, 1 week, and 1 month of life. Neonates were divided into a moderate (n = 17) and a severe (n = 11) group based on neuroradiological and amplitude-integrated EEG characteristics. Peripheral blood mononuclear cells were assessed with flow cytometry. Cytokine plasma levels were measured using Bioplex immunoassays. Components of the kynurenine pathway were assessed by high-performance liquid chromatography. RESULTS The prevalence and extravasation of IL-1b + CD4 cells were higher in severe than in moderate asphyxia at 6 h. Based on Receiver operator curve analysis, the assessment of the prevalence of CD4+ IL-1β+ and CD4+ IL-1β+ CD49d+ cells at 6 h appears to be able to predict the severity of the insult at an early stage in asphyxia. Intracellular levels of TNF-α in CD4 cells were increased at all time points compared to 6 h in both groups. At 1 month, intracellular levels of TNF-α were higher in the severe group. Plasma IL-6 levels were higher at 1 week in the severe group and decreased by 1 month in the moderate group. Intracellular levels of IL-6 peaked at 24 h in both groups. Intracellular TGF-β levels were increased from 24 h onwards in the moderate group. CONCLUSIONS IL-1β and IL-6 appear to play a key role in the early events of the inflammatory response, while TNF-α seems to be responsible for prolonged neuroinflammation, potentially contributing to a worse outcome. The assessment of the prevalence of CD4+ IL-1β+ and CD4+ IL-1β+ CD49d+ cells at 6 h appears to be able to predict the severity of the insult at an early stage in asphyxia.
Collapse
Affiliation(s)
- Anna Bajnok
- First Department of Obstetrics and Gynecology, Semmelweis University, Baross str. 27, Budapest, H-1088, Hungary.,First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - László Berta
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - Csaba Orbán
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - Gábor Veres
- Department of Neurology, Albert Szent-Györgyi Clinical Centre, Faculty of Medicine, University of Szeged, Semmelweis str. 6, 5th floor, Szeged, H-6725, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Dénes Zádori
- Department of Neurology, Albert Szent-Györgyi Clinical Centre, Faculty of Medicine, University of Szeged, Semmelweis str. 6, 5th floor, Szeged, H-6725, Hungary
| | - Hajnalka Barta
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - Ünőke Méder
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Clinical Centre, Faculty of Medicine, University of Szeged, Semmelweis str. 6, 5th floor, Szeged, H-6725, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Tivadar Tulassay
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Miklós Szabó
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Gergely Toldi
- First Department of Obstetrics and Gynecology, Semmelweis University, Baross str. 27, Budapest, H-1088, Hungary. .,First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary. .,Birmingham Women's and Children's Hospital, Neonatal Unit, Birmingham, UK.
| |
Collapse
|
28
|
Farrell K, Borazjani A, Damaser M, Kothapalli CR. Differential regulation of NSC phenotype and genotype by chronically activated microglia within cocultures. Integr Biol (Camb) 2017; 8:1145-1157. [PMID: 27722366 DOI: 10.1039/c6ib00126b] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Under disease or injury conditions in the central nervous system (CNS), activated microglia release cytokines and chemokines to modulate the microenvironment and influence tissue remodeling. To exploit the full potential of neural stem cell (NSC) transplantation approaches, a permissive microenvironment needs to be created for their survival, homing and differentiation. To investigate the role of chronically activated microglia in the fate of NSCs, spontaneously immortalized murine microglial cells (SIM-A9) were cocultured with embryonic murine cortical NSCs on 2D substrates or within 3D gels. Standalone NSC cultures served as controls. Cytokines and chemokines released by NSCs and SIM-A9 cells in standalone and cocultures were quantified. Coculturing with SIM-A9 cells suppressed NSC viability, neurite outgrowth, neural differentiation and TUJ1 gene expression, and promoted glia formation in both 2D and 3D cultures, over a 10-day period. The seven most-abundantly released analytes by microglia (MCP-1, MIP2, G-CSF, MIP-1α, MIP-1β, TNF-α, IL-6) were tested for their individual effects on NSCs, to investigate if the outcomes in cocultures were due to the synergistic effects of analytes or the influence of any individual analyte. All the seven analytes significantly suppressed cell survival compared to controls, but exposure to MIP-1β, IL-6, or MCP-1 enhanced neurite outgrowth and neural lineage commitment. Results attest to (i) the strong role of activated microglia in regulating NSC fate, (ii) the utility of selective analytes released by activated microglia in promoting neurogenesis and neuritogenesis, and (iii) the need to protect transplanted NSCs from the host inflammatory microenvironment to ensure their survival and functionality in treating neurological disorders.
Collapse
Affiliation(s)
- Kurt Farrell
- Department of Chemical and Biomedical Engineering, Cleveland State University, 2121 Euclid Ave., SH 460, Cleveland, OH 44141, USA.
| | - Ali Borazjani
- Department of Chemical and Biomedical Engineering, Cleveland State University, 2121 Euclid Ave., SH 460, Cleveland, OH 44141, USA. and Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Margot Damaser
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chandrasekhar R Kothapalli
- Department of Chemical and Biomedical Engineering, Cleveland State University, 2121 Euclid Ave., SH 460, Cleveland, OH 44141, USA.
| |
Collapse
|
29
|
Lin L, Du L. The role of secreted factors in stem cells-mediated immune regulation. Cell Immunol 2017; 326:24-32. [PMID: 28778535 DOI: 10.1016/j.cellimm.2017.07.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 12/14/2022]
Abstract
Stem cells are characterized by self-renew and multipotent differentiation abilities. Besides their roles in cell compensation, stem cells are also rich sources of growth factors, cytokines, chemokines, micro-RNAs and exosomes and serve as drug stores to maintain tissue homeostasis. Recent studies have revealed that the secretome of stem cells is regulated by the local inflammatory cues and highlighted the roles of these secretory factors in stem cell based therapies. Importantly, stem cell conditioned medium, in the absence of stem cell engraftment, have shown efficiency in treating diseases involves immune disorders. In this review, we summarize the recent advances in understanding the regulatory effects of stem cells secreted factors on different immune cells including macrophages, dendritic cells, neutrophils, NK cells, T cells, and B cells. We also discuss how stem cells released factors participate in the initiation, maintenance and resolution of inflammation. The in depth understanding of interaction between stem cells secreted factors and immune system would lead to new strategies to restore tissue homeostasis and improve the efficiency of stem cell therapies.
Collapse
Affiliation(s)
- Liangyu Lin
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences of Shanghai Jiao Tong University School of Medicine and Chinese Academy of Sciences, Shanghai 200025, China.
| | - Liming Du
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences of Shanghai Jiao Tong University School of Medicine and Chinese Academy of Sciences, Shanghai 200025, China.
| |
Collapse
|
30
|
Abstract
Statins are used widely in primary and secondary prevention of cardiovascular disease; a treatment effect that has long been thought to be due to their cholesterol-lowering properties. However, statins also have a wide range of anti-inflammatory effects independent of their lipid-lowering mechanisms. In depression, low-grade inflammation is a replicated finding, and several studies have shown antidepressant properties of diverse anti-inflammatory drugs. Large observational studies have suggested reduced risks of depression amongst those taking statins, an effect that is thought to be explained by the anti-inflammatory properties of this class of drugs. Also, preliminary randomized controlled trials (RCTs) have indicated that statins may have adjunctive antidepressant effects when used as add-on treatment to selective serotonin reuptake inhibitors (SSRIs). However, the RCTs were small and limited by low generalizability, and some early observational studies have pointed towards potential neuropsychiatric adverse effects of statin treatment. Nevertheless, based on the good tolerability and general safety of the statins, researchers are currently investigating the potential antidepressant properties of these agents. The present review aims to give an overview on the potential antidepressant effects of statins based on their anti-inflammatory properties, covering topics such as safety versus treatment effects, potential mechanisms of action and the possibility of targeted treatment (precision medicine).
Collapse
Affiliation(s)
- Ole Köhler-Forsberg
- Psychosis Research Unit, Aarhus University Hospital, Skovagervej 2, 8240, Risskov, Denmark. .,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark. .,Mental Health Centre Copenhagen, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Christiane Gasse
- National Centre for Register-Based Research (NCRR), Aarhus University, Aarhus, Denmark.,iPSYCH, The Lundbeck Initiative for Integrated Research in Psychiatry, Aarhus, Denmark
| | - Michael Berk
- Deakin University, School of Medicine, IMPACT Strategic Research Centre, Geelong, VIC, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia.,The Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia.,Department of Psychiatry, University of Melbourne, Parkville, VIC, Australia
| | - Søren Dinesen Østergaard
- Psychosis Research Unit, Aarhus University Hospital, Skovagervej 2, 8240, Risskov, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
31
|
El Behi M, Sanson C, Bachelin C, Guillot-Noël L, Fransson J, Stankoff B, Maillart E, Sarrazin N, Guillemot V, Abdi H, Cournu-Rebeix I, Fontaine B, Zujovic V. Adaptive human immunity drives remyelination in a mouse model of demyelination. Brain 2017; 140:967-980. [PMID: 28334918 PMCID: PMC5382952 DOI: 10.1093/brain/awx008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/08/2016] [Indexed: 02/05/2023] Open
Abstract
One major challenge in multiple sclerosis is to understand the cellular and molecular mechanisms leading to disease severity progression. The recently demonstrated correlation between disease severity and remyelination emphasizes the importance of identifying factors leading to a favourable outcome. Why remyelination fails or succeeds in multiple sclerosis patients remains largely unknown, mainly because remyelination has never been studied within a humanized pathological context that would recapitulate major events in plaque formation such as infiltration of inflammatory cells. Therefore, we developed a new paradigm by grafting healthy donor or multiple sclerosis patient lymphocytes in the demyelinated lesion of nude mice spinal cord. We show that lymphocytes play a major role in remyelination whose efficacy is significantly decreased in mice grafted with multiple sclerosis lymphocytes compared to those grafted with healthy donors lymphocytes. Mechanistically, we demonstrated in vitro that lymphocyte-derived mediators influenced differentiation of oligodendrocyte precursor cells through a crosstalk with microglial cells. Among mice grafted with lymphocytes from different patients, we observed diverse remyelination patterns reproducing for the first time the heterogeneity observed in multiple sclerosis patients. Comparing lymphocyte secretory profile from patients exhibiting high and low remyelination ability, we identified novel molecules involved in oligodendrocyte precursor cell differentiation and validated CCL19 as a target to improve remyelination. Specifically, exogenous CCL19 abolished oligodendrocyte precursor cell differentiation observed in patients with high remyelination pattern. Multiple sclerosis lymphocytes exhibit intrinsic capacities to coordinate myelin repair and further investigation on patients with high remyelination capacities will provide new pro-regenerative strategies.
Collapse
Affiliation(s)
- Mohamed El Behi
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Charles Sanson
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Corinne Bachelin
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Léna Guillot-Noël
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Jennifer Fransson
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Bruno Stankoff
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France.,Assistance Publique-Hôpitaux de Paris, Neurology Service, Hôpital Saint Antoine-HUEP, Paris 12, France
| | - Elisabeth Maillart
- Assistance Publique-Hôpitaux de Paris, Neurology Department Pitié Salpétrière University Hospital Paris, France
| | - Nadège Sarrazin
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Vincent Guillemot
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Hervé Abdi
- School of Brain and Behavioral Sciences, The University of Texas, Dallas, USA
| | - Isabelle Cournu-Rebeix
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| | - Bertrand Fontaine
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France.,Assistance Publique-Hôpitaux de Paris, Neurology Service, Hôpital Saint Antoine-HUEP, Paris 12, France.,Assistance Publique-Hôpitaux de Paris, Neurology Department Pitié Salpétrière University Hospital Paris, France
| | - Violetta Zujovic
- Sorbonne-Universités-UPMC 06, INSERM, CNRS, UMR ICM-75-1127-7225, 47 boulevard de l'Hôpital, 75013 Paris, France
| |
Collapse
|
32
|
Sutherland TC, Mathews KJ, Mao Y, Nguyen T, Gorrie CA. Differences in the Cellular Response to Acute Spinal Cord Injury between Developing and Mature Rats Highlights the Potential Significance of the Inflammatory Response. Front Cell Neurosci 2017; 10:310. [PMID: 28133446 PMCID: PMC5233684 DOI: 10.3389/fncel.2016.00310] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/28/2016] [Indexed: 01/11/2023] Open
Abstract
There exists a trend for a better functional recovery from spinal cord injury (SCI) in younger patients compared to adults, which is also reported for animal studies; however, the reasons for this are yet to be elucidated. The post injury tissue microenvironment is a complex milieu of cells and signals that interact on multiple levels. Inflammation has been shown to play a significant role in this post injury microenvironment. Endogenous neural progenitor cells (NPC), in the ependymal layer of the central canal, have also been shown to respond and migrate to the lesion site. This study used a mild contusion injury model to compare adult (9 week), juvenile (5 week) and infant (P7) Sprague-Dawley rats at 24 h, 1, 2, and 6 weeks post-injury (n = 108). The innate cells of the inflammatory response were examined using counts of ED1/IBA1 labeled cells. This found a decreased inflammatory response in the infants, compared to the adult and juvenile animals, demonstrated by a decreased neutrophil infiltration and macrophage and microglial activation at all 4 time points. Two other prominent cellular contributors to the post-injury microenvironment, the reactive astrocytes, which eventually form the glial scar, and the NPC were quantitated using GFAP and Nestin immunohistochemistry. After SCI in all 3 ages there was an obvious increase in Nestin staining in the ependymal layer, with long basal processes extending into the parenchyma. This was consistent between age groups early post injury then deviated at 2 weeks. The GFAP results also showed stark differences between the mature and infant animals. These results point to significant differences in the inflammatory response between infants and adults that may contribute to the better recovery indicated by other researchers, as well as differences in the overall injury progression and cellular responses. This may have important consequences if we are able to mirror and manipulate this response in patients of all ages; however much greater exploration in this area is required.
Collapse
Affiliation(s)
- Theresa C Sutherland
- Neural Injury Research Unit, School of Medical and Molecular Bioscience, University of Technology Sydney Ultimo, NSW, Australia
| | - Kathryn J Mathews
- Discipline of Biomedical Sciences and Brain and Mind Centre, Sydney Medical School, The University of Sydney Sydney, NSW, Australia
| | - Yilin Mao
- Neural Injury Research Unit, School of Medical and Molecular Bioscience, University of Technology Sydney Ultimo, NSW, Australia
| | - Tara Nguyen
- Neural Injury Research Unit, School of Medical and Molecular Bioscience, University of Technology Sydney Ultimo, NSW, Australia
| | - Catherine A Gorrie
- Neural Injury Research Unit, School of Medical and Molecular Bioscience, University of Technology Sydney Ultimo, NSW, Australia
| |
Collapse
|
33
|
Wootla B, Denic A, Watzlawik JO, Warrington AE, Zoecklein LJ, Papke-Norton LM, David C, Rodriguez M. Human class I major histocompatibility complex alleles determine central nervous system injury versus repair. J Neuroinflammation 2016; 13:293. [PMID: 27855706 PMCID: PMC5112886 DOI: 10.1186/s12974-016-0759-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 11/03/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We investigated the role of human HLA class I molecules in persistent central nervous system (CNS) injury versus repair following virus infection of the CNS. METHODS Human class I A11+ and B27+ transgenic human beta-2 microglobulin positive (Hβ2m+) mice of the H-2 b background were generated on a combined class I-deficient (mouse beta-2 microglobulin deficient, β2m0) and class II-deficient (mouse Aβ0) phenotype. Intracranial infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL mice results in acute encephalitis with prominent injury in the hippocampus, striatum, and cortex. RESULTS Following infection with TMEV, a picornavirus, the Aβ0.β2m0 mice lacking active immune responses died within 18 to 21 days post-infection. These mice showed severe encephalomyelitis due to rapid replication of the viral genome. In contrast, transgenic Hβ2m mice with insertion of a single human class I MHC gene in the absence of human or mouse class II survived the acute infection. Both A11+ and B27+ mice significantly controlled virus RNA expression by 45 days and did not develop late-onset spinal cord demyelination. By 45 days post-infection (DPI), B27+ transgenic mice showed almost complete repair of the virus-induced brain injury, but A11+ mice conversely showed persistent severe hippocampal and cortical injury. CONCLUSIONS The findings support the hypothesis that the expression of a single human class I MHC molecule, independent of persistent virus infection, influences the extent of sub frequent chronic neuronal injury or repair in the absence of a class II MHC immune response.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Aleksandar Denic
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Jens O. Watzlawik
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224 USA
| | - Arthur E. Warrington
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Laurie J. Zoecklein
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Louisa M. Papke-Norton
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Chella David
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|
34
|
Nardo G, Trolese MC, de Vito G, Cecchi R, Riva N, Dina G, Heath PR, Quattrini A, Shaw PJ, Piazza V, Bendotti C. Immune response in peripheral axons delays disease progression in SOD1 G93A mice. J Neuroinflammation 2016; 13:261. [PMID: 27717377 PMCID: PMC5055725 DOI: 10.1186/s12974-016-0732-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/23/2016] [Indexed: 11/16/2022] Open
Abstract
Background Increasing evidence suggests that the immune system has a beneficial role in the progression of amyotrophic lateral sclerosis (ALS) although the mechanism remains unclear. Recently, we demonstrated that motor neurons (MNs) of C57SOD1G93A mice with slow disease progression activate molecules classically involved in the cross-talk with the immune system. This happens a lot less in 129SvSOD1G93A mice which, while expressing the same amount of transgene, had faster disease progression and earlier axonal damage. The present study investigated whether and how the immune response is involved in the preservation of motor axons in the mouse model of familial ALS with a more benign disease course. Methods First, the extent of axonal damage, Schwann cell proliferation, and neuromuscular junction (NMJ) denervation were compared between the two ALS mouse models at the disease onset. Then, we compared the expression levels of different immune molecules, the morphology of myelin sheaths, and the presence of blood-derived immune cell infiltrates in the sciatic nerve of the two SOD1G93A mouse strains using immunohistochemical, immunoblot, quantitative reverse transcription PCR, and rotating-polarization Coherent Anti-Stokes Raman Scattering techniques. Results Muscle denervation, axonal dysregulation, and myelin disruption together with reduced Schwann cell proliferation are prominent in 129SvSOD1G93A compared to C57SOD1G93A mice at the disease onset, and this correlates with a faster disease progression in the first strain. On the contrary, a striking increase of immune molecules such as CCL2, MHCI, and C3 was seen in sciatic nerves of slow progressor C57SOD1G93A mice and this was accompanied by heavy infiltration of CD8+ T lymphocytes and macrophages. These phenomena were not detectable in the peripheral nervous system of fast-progressing mice. Conclusions These data show for the first time that damaged MNs in SOD1-related ALS actively recruit immune cells in the peripheral nervous system to delay muscle denervation and prolong the lifespan. On the contrary, the lack of this response has a negative impact on the disease course. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0732-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy.
| | - Maria Chiara Trolese
- Laboratory of Molecular Neurobiology, Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| | - Giuseppe de Vito
- NEST, Scuola Normale Superiore, Piazza San Silvestro 12, I-56127, Pisa, Italy.,Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, I-56127, Pisa, Italy
| | - Roberta Cecchi
- NEST, Scuola Normale Superiore, Piazza San Silvestro 12, I-56127, Pisa, Italy
| | - Nilo Riva
- Neuropathology Unit, Department of Neurology, INSPE, San Raffaele Scientific Institute, Dibit II, Via Olgettina 48, 20132, Milan, Italy
| | - Giorgia Dina
- Neuropathology Unit, Department of Neurology, INSPE, San Raffaele Scientific Institute, Dibit II, Via Olgettina 48, 20132, Milan, Italy
| | - Paul R Heath
- Department of Neuroscience, Academic Neurology Unit, Faculty of Medicine, Dentistry and Health, Sheffield Institute for Translational Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Angelo Quattrini
- Neuropathology Unit, Department of Neurology, INSPE, San Raffaele Scientific Institute, Dibit II, Via Olgettina 48, 20132, Milan, Italy
| | - Pamela J Shaw
- Department of Neuroscience, Academic Neurology Unit, Faculty of Medicine, Dentistry and Health, Sheffield Institute for Translational Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Vincenzo Piazza
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, I-56127, Pisa, Italy
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy.
| |
Collapse
|
35
|
DeLeo JA, Tanga FY, Tawfik VL. Neuroimmune Activation and Neuroinflammation in Chronic Pain and Opioid Tolerance/Hyperalgesia. Neuroscientist 2016; 10:40-52. [PMID: 14987447 DOI: 10.1177/1073858403259950] [Citation(s) in RCA: 297] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
One area that has emerged as a promising therapeutic target for the treatment and prevention of chronic pain and opioid tolerance/hyperalgesia is the modulation of the central nervous system (CNS) immunological response that ensues following injury or opioid administration. Broadly defined, central neuroimmune activation involves the activation of cells that interface with the peripheral nervous system and blood. Activation of these cells, as well as parenchymal microglia and astrocytes by injury, opioids, and other stressors, leads to subsequent production of cytokines, cellular adhesion molecules, chemokines, and the expression of surface antigens that enhance a CNS immune cascade. This response can lead to the production of numerous pain mediators that can sensitize and lower the threshold of neuronal firing: the pathologic correlate to central sensitization and chronic pain states. CNS innate immunity and Toll-like receptors, in particular, may be vital players in this orchestrated immune response and may hold the answers to what initiates this complex cascade. The challenge remains in the careful perturbation of injury/opioid-induced neuroimmune activation to down-regulate this process without inhibiting beneficial CNS autoimmunity that subserves neuronal protection following injury.
Collapse
Affiliation(s)
- Joyce A DeLeo
- Department of Anesthesiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA.
| | | | | |
Collapse
|
36
|
Cytokine and Growth Factor Activation In Vivo and In Vitro after Spinal Cord Injury. Mediators Inflamm 2016; 2016:9476020. [PMID: 27418745 PMCID: PMC4935915 DOI: 10.1155/2016/9476020] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/18/2016] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury results in a life-disrupting series of deleterious interconnected mechanisms encompassed by the primary and secondary injury. These events are mediated by the upregulation of genes with roles in inflammation, transcription, and signaling proteins. In particular, cytokines and growth factors are signaling proteins that have important roles in the pathophysiology of SCI. The balance between the proinflammatory and anti-inflammatory effects of these molecules plays a critical role in the progression and outcome of the lesion. The excessive inflammatory Th1 and Th17 phenotypes observed after SCI tilt the scale towards a proinflammatory environment, which exacerbates the deleterious mechanisms present after the injury. These mechanisms include the disruption of the spinal cord blood barrier, edema and ion imbalance, in particular intracellular calcium and sodium concentrations, glutamate excitotoxicity, free radicals, and the inflammatory response contributing to the neurodegenerative process which is characterized by demyelination and apoptosis of neuronal tissue.
Collapse
|
37
|
Petersen GF, Strappe PM. Generation of diverse neural cell types through direct conversion. World J Stem Cells 2016; 8:32-46. [PMID: 26981169 PMCID: PMC4766249 DOI: 10.4252/wjsc.v8.i2.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/18/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
A characteristic of neurological disorders is the loss of critical populations of cells that the body is unable to replace, thus there has been much interest in identifying methods of generating clinically relevant numbers of cells to replace those that have been damaged or lost. The process of neural direct conversion, in which cells of one lineage are converted into cells of a neural lineage without first inducing pluripotency, shows great potential, with evidence of the generation of a range of functional neural cell types both in vitro and in vivo, through viral and non-viral delivery of exogenous factors, as well as chemical induction methods. Induced neural cells have been proposed as an attractive alternative to neural cells derived from embryonic or induced pluripotent stem cells, with prospective roles in the investigation of neurological disorders, including neurodegenerative disease modelling, drug screening, and cellular replacement for regenerative medicine applications, however further investigations into improving the efficacy and safety of these methods need to be performed before neural direct conversion becomes a clinically viable option. In this review, we describe the generation of diverse neural cell types via direct conversion of somatic cells, with comparison against stem cell-based approaches, as well as discussion of their potential research and clinical applications.
Collapse
|
38
|
Kaka G, Yaghoobi K, Davoodi S, Hosseini SR, Sadraie SH, Mansouri K. Assessment of the Neuroprotective Effects of Lavandula angustifolia Extract on the Contusive Model of Spinal Cord Injury in Wistar Rats. Front Neurosci 2016; 10:25. [PMID: 26903793 PMCID: PMC4744928 DOI: 10.3389/fnins.2016.00025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/19/2016] [Indexed: 12/22/2022] Open
Abstract
Introduction: Spinal cord injury (SCI) involves a primary trauma and secondary cellular processes that can lead to severe damage to the nervous system, resulting in long-term spinal deficits. At the cellular level, SCI causes astrogliosis, of which glial fibrillary acidic protein (GFAP) is a major index. Objective: The aim of this study was to investigate the neuroprotective effects of Lavandula angustifolia (Lav) on the repair of spinal cord injuries in Wistar rats. Materials and Methods: Forty-five female rats were randomly divided into six groups of seven rats each: the intact, sham, control (SCI), Lav 100, Lav 200, and Lav 400 groups. Every week after SCI onset, all animals were evaluated for behavior outcomes by the Basso, Beattie, and Bresnahan (BBB) score. H&E staining was performed to examine the lesions post-injury. GFAP expression was assessed for astrogliosis. Somatosensory evoked potential (SEP) testing was performed to detect the recovery of neural conduction. Results: BBB scores were significantly increased and delayed responses on sensory tests were significantly decreased in the Lav 200 and Lav 400 groups compared to the control group. The greatest decrease of GFAP was evident in the Lav 200 and Lav 400 groups. EMG results showed significant improvement in the hindlimbs in the Lav 200 and Lav 400 groups compared to the control group. Cavity areas significantly decreased and the number of ventral motor neurons significantly increased in the Lav 200 and Lav 400 groups. Conclusion: Lav at doses of 200 and 400 mg/kg can promote structural and functional recovery after SCI. The neuroprotective effects of L. angustifolia can lead to improvement in the contusive model of SCI in Wistar rats.
Collapse
Affiliation(s)
- Gholamreza Kaka
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences Tehran, Iran
| | - Kayvan Yaghoobi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences Tehran, Iran
| | - Shaghayegh Davoodi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences Tehran, Iran
| | - Seyed R Hosseini
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences Tehran, Iran
| | - Seyed H Sadraie
- Department of Anatomy, School of Medicine, Baqiyatallah University of Medical Sciences Tehran, Iran
| | - Korosh Mansouri
- Department of Physical Medicine and Rehabilitation, Iran University of Medical Sciences Tehran, Iran
| |
Collapse
|
39
|
Wlodarczyk A, Cédile O, Jensen KN, Jasson A, Mony JT, Khorooshi R, Owens T. Pathologic and Protective Roles for Microglial Subsets and Bone Marrow- and Blood-Derived Myeloid Cells in Central Nervous System Inflammation. Front Immunol 2015; 6:463. [PMID: 26441968 PMCID: PMC4562247 DOI: 10.3389/fimmu.2015.00463] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a series of processes designed for eventual clearance of pathogens and repair of damaged tissue. In the context of autoimmune recognition, inflammatory processes are usually considered to be pathological. This is also true for inflammatory responses in the central nervous system (CNS). However, as in other tissues, neuroinflammation can have beneficial as well as pathological outcomes. The complex role of encephalitogenic T cells in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE) may derive from heterogeneity of the myeloid cells with which these T cells interact within the CNS. Myeloid cells, including resident microglia and infiltrating bone marrow-derived cells, such as dendritic cells (DC) and monocytes/macrophages [bone marrow-derived macrophages (BMDM)], are highly heterogeneous populations that may be involved in neurotoxicity and also immunoregulation and regenerative processes. Better understanding and characterization of myeloid cell heterogeneity is essential for future development of treatments controlling inflammation and inducing neuroprotection and neuroregeneration in diseased CNS. Here, we describe and compare three populations of myeloid cells: CD11c+ microglia, CD11c− microglia, and CD11c+ blood-derived cells in terms of their pathological versus protective functions in the CNS of mice with EAE. Our data show that CNS-resident microglia include functionally distinct subsets that can be distinguished by their expression of CD11c. These subsets differ in their expression of Arg-1, YM1, iNOS, IL-10, and IGF-1. Moreover, in contrast to BMDM/DC, both subsets of microglia express protective interferon-beta (IFNβ), high levels of colony-stimulating factor-1 receptor, and do not express the Th1-associated transcription factor T-bet. Taken together, our data suggest that CD11c+ microglia, CD11c− microglia, and infiltrating BMDM/DC represent separate and distinct populations and illustrate the heterogeneity of the CNS inflammatory environment.
Collapse
Affiliation(s)
- Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Oriane Cédile
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Kirstine Nolling Jensen
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Agathe Jasson
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark ; Department of Biology, École Normale Supérieure de Lyon , Lyon , France
| | - Jyothi Thyagabhavan Mony
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| |
Collapse
|
40
|
Neural Plasticity in Multiple Sclerosis: The Functional and Molecular Background. Neural Plast 2015; 2015:307175. [PMID: 26229689 PMCID: PMC4503575 DOI: 10.1155/2015/307175] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/09/2015] [Accepted: 06/21/2015] [Indexed: 01/19/2023] Open
Abstract
Multiple sclerosis is an autoimmune neurodegenerative disorder resulting in motor dysfunction and cognitive decline. The inflammatory and neurodegenerative changes seen in the brains of MS patients lead to progressive disability and increasing brain atrophy. The most common type of MS is characterized by episodes of clinical exacerbations and remissions. This suggests the presence of compensating mechanisms for accumulating damage. Apart from the widely known repair mechanisms like remyelination, another important phenomenon is neuronal plasticity. Initially, neuroplasticity was connected with the developmental stages of life; however, there is now growing evidence confirming that structural and functional reorganization occurs throughout our lifetime. Several functional studies, utilizing such techniques as fMRI, TBS, or MRS, have provided valuable data about the presence of neuronal plasticity in MS patients. CNS ability to compensate for neuronal damage is most evident in RR-MS; however it has been shown that brain plasticity is also preserved in patients with substantial brain damage. Regardless of the numerous studies, the molecular background of neuronal plasticity in MS is still not well understood. Several factors, like IL-1β, BDNF, PDGF, or CB1Rs, have been implicated in functional recovery from the acute phase of MS and are thus considered as potential therapeutic targets.
Collapse
|
41
|
Palavra F, Díaz EC, Sena A. Cardiometabolic Risk, Inflammation, and Neurodegenerative Disorders. BIOMARKERS OF CARDIOMETABOLIC RISK, INFLAMMATION AND DISEASE 2015:133-159. [DOI: 10.1007/978-3-319-16018-4_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Modulation properties of factors released by bone marrow stromal cells on activated microglia: an in vitro study. Sci Rep 2014; 4:7514. [PMID: 25524416 PMCID: PMC5378994 DOI: 10.1038/srep07514] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/24/2014] [Indexed: 12/14/2022] Open
Abstract
In the present paper we develop a new non-cell based (cell-free) therapeutic approach applied to BV2 microglial cells and spinal cord derived primary microglia (PM) using conditioned media from rat bone marrow stromal cells (BMSCs-CM). First we collected conditioned media (CM) from either naive or injured rat spinal cord tissue (SCI-CM, inflammatory stimulation agent) and from rat bone marrow stromal cells (BMSCs-CM, therapeutic immunomodulation agent). They were both subsequently checked for the presence of chemokines and growth, neurotrophic and neural migration factors using proteomics analysis. The data clearly showed that rat BMSCs-CM contain in vitro growth factors, neural migration factors, osteogenic factors, differentiating factors and immunomodulators, whereas SCI-CM contain chemokines, chemoattractant factors and neurotrophic factors. Afterwards we determined whether the BMSCs-CM affect chemotactic activity, NO production, morphological and pro-apoptotic changes of either BV2 or PM cells once activated with SCI-CM. Our results confirm the anti-migratory and NO-inhibitory effects of BMSCs-CM on SCI-CM-activated microglia with higher impact on primary microglia. The cytotoxic effect of BMSCs-CM occurred only on SCI-CM-stimulated BV2 cells and PM, not on naive BV2 cells, nor on PM. Taken together, the molecular cocktail found in BMSCs-CM is favorable for immunomodulatory properties.
Collapse
|
43
|
Ghaemi A, Sajadian A, Khodaie B, Lotfinia AA, Lotfinia M, Aghabarari A, Khaleghi Ghadiri M, Meuth S, Gorji A. Immunomodulatory Effect of Toll-Like Receptor-3 Ligand Poly I:C on Cortical Spreading Depression. Mol Neurobiol 2014; 53:143-154. [PMID: 25416860 DOI: 10.1007/s12035-014-8995-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023]
Abstract
The release of inflammatory mediators following cortical spreading depression (CSD) is suggested to play a role in pathophysiology of CSD-related neurological disorders. Toll-like receptors (TLR) are master regulators of innate immune function and involved in the activation of inflammatory responses in the brain. TLR3 agonist poly I:C exerts anti-inflammatory effect and prevents cell injury in the brain. The aim of the present study was to examine the effect of systemic administration of poly I:C on the release of cytokines (TNF-α, IFN-γ, IL-4, TGF-β1, and GM-CSF) in the brain and spleen, splenic lymphocyte proliferation, expression of GAD65, GABAAα, GABAAβ as well as Hsp70, and production of dark neurons after induction of repetitive CSD in juvenile rats. Poly I:C significantly attenuated CSD-induced production of TNF-α and IFN-γ in the brain as well as TNF-α and IL-4 in the spleen. Poly I:C did not affect enhancement of splenic lymphocyte proliferation after CSD. Administration of poly I:C increased expression of GABAAα, GABAAβ as well as Hsp70 and decreased expression of GAD65 in the entorhinal cortex compared to CSD-treated tissues. In addition, poly I:C significantly prevented production of CSD-induced dark neurons. The data indicate neuroprotective and anti-inflammatory effects of TLR3 activation on CSD-induced neuroinflammation. Targeting TLR3 may provide a novel strategy for developing new treatments for CSD-related neurological disorders.
Collapse
Affiliation(s)
- Amir Ghaemi
- Shefa Neuroscience Research Center, Tehran, Iran.,Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | | | | | | | | | | | - Maryam Khaleghi Ghadiri
- Klinik und Poliklinik für Neurochirurgie, WestfälischeWilhelms-Universität Münster, Münster, Germany
| | - Sven Meuth
- Department of Neurology, WestfälischeWilhelms-Universität Münster, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Center, Tehran, Iran. .,Institut für Physiologie I, WestfälischeWilhelms-Universität Münster, Münster, Germany. .,Epilepsy Research Center, Universität Münster, Albert-Schweitzer-Campus 1, Gebäude: A1, 48149, Münster, Germany.
| |
Collapse
|
44
|
Mazzon E, Bruscoli S, Galuppo M, Biagioli M, Sorcini D, Bereshchenko O, Fiorucci C, Migliorati G, Bramanti P, Riccardi C. Glucocorticoid-induced leucine zipper (GILZ) controls inflammation and tissue damage after spinal cord injury. CNS Neurosci Ther 2014; 20:973-81. [PMID: 25146427 DOI: 10.1111/cns.12315] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/17/2014] [Accepted: 07/28/2014] [Indexed: 01/08/2023] Open
Abstract
AIMS Spinal cord injury (SCI) occurs following damage to the spinal column. Following trauma, tissue damage is further exacerbated by a secondary damage due to a SCI-activated inflammatory process. Control of leukocytes activity is essential to therapeutic inhibition of the spinal cord damage to ameliorate the patient's conditions. The mechanisms that regulate neuroinflammation following SCI, including T-cell infiltration, have not been completely clarified. Glucocorticoids (GC) are antiinflammatory drugs widely used in therapy, including treatment of SCI. GC efficacy may be linked to many molecular mechanisms that are involved in regulation of leukocytes migration, activation, and differentiation. We have previously shown that the antiinflammatory activity of GC is in part mediated by glucocorticoid-induced leucine zipper (GILZ). Here, we investigated the role of GILZ in inflammation and spinal cord tissue damage following a spinal trauma. METHODS We address the role of GILZ in SCI-induced inflammation and tissue damage using a model of SCI in gilz knockout (gilz KO) and wild-type (WT) mice. RESULTS We found that GILZ deficiency is associated with a strong reduction of SCI-induced inflammation and a significantly reduced lesion area following SCI. CONCLUSION These results demonstrate that GILZ is involved in induction of neuroinflammation and functional outcomes of spinal cord trauma.
Collapse
Affiliation(s)
- Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Messina, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhang B, Gensel J. Is neuroinflammation in the injured spinal cord different than in the brain? Examining intrinsic differences between the brain and spinal cord. Exp Neurol 2014; 258:112-20. [DOI: 10.1016/j.expneurol.2014.04.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 03/28/2014] [Accepted: 04/08/2014] [Indexed: 12/17/2022]
|
46
|
Cizkova D, Le Marrec-Croq F, Franck J, Slovinska L, Grulova I, Devaux S, Lefebvre C, Fournier I, Salzet M. Alterations of protein composition along the rostro-caudal axis after spinal cord injury: proteomic, in vitro and in vivo analyses. Front Cell Neurosci 2014; 8:105. [PMID: 24860426 PMCID: PMC4028999 DOI: 10.3389/fncel.2014.00105] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/24/2014] [Indexed: 12/14/2022] Open
Abstract
Based on proteomic analyses we investigated the differences of released molecules in the conditioned media (CM) from the spinal cord central lesion and adjacent rostral and caudal segments at 3, 7, and 10 days after spinal cord injury (SCI), in order to specify the molecular environment within greater extent of tissue damage. Proteins found in CM were analyzed by shot-gun MS using nanoLC coupled to an orbitrap. The results showed some specific proteins at each site of the lesion at 3days. Among the proteins from rostral and lesion segments, some are related to chemokines, cytokines or to neurogenesis factors. In contrast, proteins from caudal segments are more related to necrosis factors. The CM from each spinal segment were used in vitro, on microglial BV2 cell lines and DRGs explants, showing a lesion site-dependent impact on microglia activation and DRGs neurite outgrowth. In addition, while naive BV2 cells exhibited insignificant staining for CX3CR1 receptor, the level of CX3CR1 was strongly enhanced in some BV2 cells after their stimulation by CM collected from SCI. The molecular data might correlate with different polarization of activated microglia and macrophages along the rostro-caudal axis following acute injury. This was partially confirmed in vivo with CX3CR1 receptor, revealing higher expression in the rostral segment, with potential neuroprotective action. In addition, the neurotrophic factors released from rostral and lesion segments enhanced outgrowth of DRGs explants. Taken together these data suggest that regionalization in terms of inflammatory and neurotrophic responses may occur between rostral and caudal segments in acute SCI.
Collapse
Affiliation(s)
- Dasa Cizkova
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, FRE CNRS 3637, Université Lille 1 Villeneuve d'Ascq, France ; Laboratory of Cell and Tissue Culture, Institute of Neurobiology, Center of Excellence for Brain Research, Slovak Academy of Sciences Košice, Slovakia
| | - Françoise Le Marrec-Croq
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, FRE CNRS 3637, Université Lille 1 Villeneuve d'Ascq, France
| | - Julien Franck
- Laboratory of Cell and Tissue Culture, Institute of Neurobiology, Center of Excellence for Brain Research, Slovak Academy of Sciences Košice, Slovakia
| | - Lucia Slovinska
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, FRE CNRS 3637, Université Lille 1 Villeneuve d'Ascq, France
| | - Ivana Grulova
- Laboratory of Cell and Tissue Culture, Institute of Neurobiology, Center of Excellence for Brain Research, Slovak Academy of Sciences Košice, Slovakia
| | - Stéphanie Devaux
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, FRE CNRS 3637, Université Lille 1 Villeneuve d'Ascq, France
| | - Christophe Lefebvre
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, FRE CNRS 3637, Université Lille 1 Villeneuve d'Ascq, France
| | - Isabelle Fournier
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, FRE CNRS 3637, Université Lille 1 Villeneuve d'Ascq, France
| | - Michel Salzet
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, EA 4550, FRE CNRS 3637, Université Lille 1 Villeneuve d'Ascq, France
| |
Collapse
|
47
|
Growth factors and synaptic plasticity in relapsing-remitting multiple sclerosis. Neuromolecular Med 2014; 16:490-8. [PMID: 24671722 DOI: 10.1007/s12017-014-8297-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/13/2014] [Indexed: 10/25/2022]
Abstract
During multiple sclerosis (MS) inflammatory attacks, and in subsequent clinical recovery phases, immune cells contribute to neuronal and oligodendroglial cell survival and tissue repair by secreting growth factors. Animal studies showed that growth factors also play a substantial role in regulating synaptic plasticity, and namely in long-term potentiation (LTP). LTP could drive clinical recovery in relapsing patients by restoring the excitability of denervated neurons. We recently reported that maintenance of synaptic plasticity reserve is crucial to contrast clinical deterioration in MS and that the platelet-derived growth factor (PDGF) may play a key role in its regulation. We also reported that a Hebbian form of LTP-like cortical plasticity, explored by paired associative stimulation (PAS), correlates with clinical recovery from a relapse in MS. Here, we explored the role of PDGF in clinical recovery and in adaptive neuroplasticity in relapsing-remitting MS (RR-MS) patients. We found a correlation between the cerebrospinal fluid (CSF) PDGF concentrations and the extent of clinical recovery after a relapse, as full recovery was more likely observed in patients with high PDGF concentrations and poor recovery in subjects with low PDGF levels. Consistently with the idea that PDGF-driven synaptic plasticity contributes to attenuate the clinical consequences of tissue damage in RR-MS, we also found a striking correlation between CSF levels of PDGF and the amplitude of LTP-like cortical plasticity explored by PAS. CSF levels of fibroblast growth factor, granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor did not correlate with clinical recovery nor with measures of synaptic transmission and plasticity.
Collapse
|
48
|
Codeluppi S, Fernandez-Zafra T, Sandor K, Kjell J, Liu Q, Abrams M, Olson L, Gray NS, Svensson CI, Uhlén P. Interleukin-6 secretion by astrocytes is dynamically regulated by PI3K-mTOR-calcium signaling. PLoS One 2014; 9:e92649. [PMID: 24667246 PMCID: PMC3965459 DOI: 10.1371/journal.pone.0092649] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 02/25/2014] [Indexed: 12/22/2022] Open
Abstract
After contusion spinal cord injury (SCI), astrocytes become reactive and form a glial scar. While this reduces spreading of the damage by containing the area of injury, it inhibits regeneration. One strategy to improve the recovery after SCI is therefore to reduce the inhibitory effect of the scar, once the acute phase of the injury has passed. The pleiotropic cytokine interleukin-6 (IL-6) is secreted immediately after injury and regulates scar formation; however, little is known about the role of IL-6 in the sub-acute phases of SCI. Interestingly, IL-6 also promotes axon regeneration, and therefore its induction in reactive astrocytes may improve regeneration after SCI. We found that IL-6 is expressed by astrocytes and neurons one week post-injury and then declines. Using primary cultures of rat astrocytes we delineated the molecular mechanisms that regulate IL-6 expression and secretion. IL-6 expression requires activation of p38 and depends on NF-κB transcriptional activity. Activation of these pathways in astrocytes occurs when the PI3K-mTOR-AKT pathway is inhibited. Furthermore, we found that an increase in cytosolic calcium concentration was necessary for IL-6 secretion. To induce IL-6 secretion in astrocytes, we used torin2 and rapamycin to block the PI3K-mTOR pathway and increase cytosolic calcium, respectively. Treating injured animals with torin2 and rapamycin for two weeks, starting two weeks after injury when the scar has been formed, lead to a modest effect on mechanical hypersensitivity, limited to the period of treatment. These data, taken together, suggest that treatment with torin2 and rapamycin induces IL-6 secretion by astrocytes and may contribute to the reduction of mechanical hypersensitivity after SCI.
Collapse
Affiliation(s)
- Simone Codeluppi
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Teresa Fernandez-Zafra
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jacob Kjell
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Qingsong Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mathew Abrams
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Per Uhlén
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
49
|
Beurel E, Harrington LE, Buchser W, Lemmon V, Jope RS. Astrocytes modulate the polarization of CD4+ T cells to Th1 cells. PLoS One 2014; 9:e86257. [PMID: 24489707 PMCID: PMC3904883 DOI: 10.1371/journal.pone.0086257] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 12/10/2013] [Indexed: 12/24/2022] Open
Abstract
T-cell characteristics are dynamic and influenced by multiple factors. To test whether cells and the environment in the central nervous system (CNS) can influence T-cells, we tested if culturing mouse CD4+ T-cells on mouse primary astrocytes, compared with standard feeder cells, modified T-cell polarization to Th1 and Treg subtypes. Astrocytes supported the production of Th1 cells and Tregs, which was diminished by inflammatory activation of astrocytes, and glutamate accumulation that may result from impaired glutamate uptake by astrocytes strongly promoted Th1 production. These results demonstrate that astrocytes and the environment in the CNS have the capacity to regulate T-cell characteristics.
Collapse
Affiliation(s)
- Eléonore Beurel
- Departments of Psychiatry and Behavioral Sciences, and Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- * E-mail:
| | - Laurie E. Harrington
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - William Buchser
- Department of Genetics, Washington University, St Louis, Missouri, United States of America
| | - Vance Lemmon
- Miami Project to Cure Paralysis, University of Miami, Miami, Florida, United States of America
| | - Richard S. Jope
- Departments of Psychiatry and Behavioral Sciences, and Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
50
|
Paterniti I, Impellizzeri D, Di Paola R, Esposito E, Gladman S, Yip P, Priestley JV, Michael-Titus AT, Cuzzocrea S. Docosahexaenoic acid attenuates the early inflammatory response following spinal cord injury in mice: in-vivo and in-vitro studies. J Neuroinflammation 2014; 11:6. [PMID: 24405628 PMCID: PMC3895696 DOI: 10.1186/1742-2094-11-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/30/2013] [Indexed: 12/30/2022] Open
Abstract
Background Two families of polyunsaturated fatty acid (PUFA), omega-3 (ω-3) and omega-6 (ω-6), are required for physiological functions. The long chain ω-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have significant biological effects. In particular, DHA is a major component of cell membranes in the brain. It is also involved in neurotransmission. Spinal cord injury (SCI) is a highly devastating pathology that can lead to catastrophic dysfunction, with a significant reduction in the quality of life. Previous studies have shown that EPA and DHA can exert neuroprotective effects in SCI in mice and rats. The aim of this study was to analyze the mechanism of action of ω-3 PUFAs, such as DHA, in a mouse model of SCI, with a focus on the early pathophysiological processes. Methods In this study, SCI was induced in mice by the application of an aneurysm clip onto the dura mater via a four-level T5 to T8 laminectomy. Thirty minutes after compression, animals received a tail vein injection of DHA at a dose of 250 nmol/kg. All animals were killed at 24 h after SCI, to evaluate various parameters implicated in the spread of the injury. Results Our results in this in-vivo study clearly demonstrate that DHA treatment reduces key factors associated with spinal cord trauma. Treatment with DHA significantly reduced: (1) the degree of spinal cord inflammation and tissue injury, (2) pro-inflammatory cytokine expression (TNF-α), (3) nitrotyrosine formation, (4) glial fibrillary acidic protein (GFAP) expression, and (5) apoptosis (Fas-L, Bax, and Bcl-2 expression). Moreover, DHA significantly improved the recovery of limb function. Furthermore, in this study we evaluated the effect of oxidative stress on dorsal root ganglion (DRG) cells using a well-characterized in-vitro model. Treatment with DHA ameliorated the effects of oxidative stress on neurite length and branching. Conclusions Our results, in vivo and in vitro, clearly demonstrate that DHA treatment reduces the development of inflammation and tissue injury associated with spinal cord trauma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 98166 Messina, Italy.
| |
Collapse
|