1
|
Cominelli G, Sulas F, Pinto D, Rinaldi F, Favero G, Rezzani R. Neuro-Nutritional Approach to Neuropathic Pain Management: A Critical Review. Nutrients 2025; 17:1502. [PMID: 40362812 PMCID: PMC12073121 DOI: 10.3390/nu17091502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Pain is a significant global public health issue that can interfere with daily activities, sleep, and interpersonal relationships when it becomes chronic or worsens, ultimately impairing quality of life. Despite ongoing efforts, the efficacy of pain treatments in improving outcomes for patients remains limited. At present, the challenge lies in developing a personalized care and management plan that helps to maintain patient activity levels and effectively manages pain. Neuropathic pain is a chronic condition resulting from damage to the somatosensory nervous system, significantly impacting quality of life. It is partly thought to be caused by inflammation and oxidative stress, and clinical research has suggested a link between this condition and diet. However, these links are not yet well understood and require further investigation to evaluate the pathways involved in neuropathic pain. Specifically, the question remains whether supplementation with dietary antioxidants, such as melatonin, could serve as a potential adjunctive treatment for neuropathic pain modulation. Melatonin, primarily secreted by the pineal gland but also produced by other systems such as the digestive system, is known for its anti-inflammatory, antioxidant, and anti-aging properties. It is found in various fruits and vegetables, and its presence alongside other polyphenols in these foods may enhance melatonin intake and contribute to improved health. The aim of this review is to provide an overview of neuropathic pain and examine the potential role of melatonin as an adjunctive treatment in a neuro-nutritional approach to pain management.
Collapse
Affiliation(s)
- Giorgia Cominelli
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
| | - Francesca Sulas
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy; (D.P.); (F.R.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy; (D.P.); (F.R.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale–SISDO), 25123 Brescia, Italy
| |
Collapse
|
2
|
Bernier RA, Sundermann EE, Edland SD, Deters KD, Shepherd AL, Clark AL, Shiroma EJ, Banks SJ. Exercise: Just What the Doctor Ordered, But Why? Elucidating Mechanisms for Women's Increased High-Density Lipoprotein Benefit From Exercise and for the Health ABC Study. J Appl Gerontol 2024; 43:1939-1949. [PMID: 38835249 DOI: 10.1177/07334648241257995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
High-density lipoprotein (HDL) is protective against cardiovascular disease. Exercise can increase HDL concentration, and some evidence suggests that this effect occurs more strongly in women than in men. Both HDL and exercise are associated with inflammation. We hypothesized a sex-by-exercise interaction on HDL level, whereby women would benefit from exercise more strongly than men, and tumor necrosis factor alpha and serum soluble tumor necrosis factor receptor-2 would mediate this relationship. This study included 2,957 older adult participants (1,520 women; 41% Black, 59% White; 73.6-years-old) from the Health, Aging, and Body Composition study. Regression models revealed a positive exercise-HDL relationship in women only (sex-by-exercise interaction: β = 0.09, p = .013; exercise on HDL in women: β = 0.07, p = .015), mediated by TNFα (axb = 0.15; CI: 0.01, 0.30), suggesting that exercise may increase HDL levels in women through reduced inflammation. Given that vascular risk contributes to Alzheimer's disease risk, findings have implications for sex differences in AD risk factors.
Collapse
Affiliation(s)
- Rachel A Bernier
- University of California, San Diego, San Diego, CA, USA
- Wentworth-Douglass Hospital Mass General Brigham, Dover, NH, USA
| | | | | | - Kacie D Deters
- University of California Los Angeles, Los Angeles, CA, USA
| | | | - Alexandra L Clark
- VA San Diego Healthcare System, La Jolla, CA, USA
- University of Texas at Austin, Austin, TX, USA
| | - Eric J Shiroma
- National Institute on Aging, National Institutes of Health, Washington, DC, USA
| | - Sarah J Banks
- University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
3
|
Pegoretti V, Bauer J, Fischer R, Paro I, Douwenga W, Kontermann RE, Pfizenmaier K, Houben E, Broux B, Hellings N, Baron W, Laman JD, Eisel ULM. Sequential treatment with a TNFR2 agonist and a TNFR1 antagonist improves outcomes in a humanized mouse model for MS. J Neuroinflammation 2023; 20:106. [PMID: 37138340 PMCID: PMC10157968 DOI: 10.1186/s12974-023-02785-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
TNF signaling is an essential regulator of cellular homeostasis. Through its two receptors TNFR1 and TNFR2, soluble versus membrane-bound TNF enable cell death or survival in a variety of cell types. TNF-TNFRs signaling orchestrates important biological functions such as inflammation, neuronal activity as well as tissue de- and regeneration. TNF-TNFRs signaling is a therapeutic target for neurodegenerative diseases such as multiple sclerosis (MS) and Alzheimer's disease (AD), but animal and clinical studies yielded conflicting findings. Here, we ask whether a sequential modulation of TNFR1 and TNFR2 signaling is beneficial in experimental autoimmune encephalomyelitis (EAE), an experimental mouse model that recapitulates inflammatory and demyelinating aspects of MS. To this end, human TNFR1 antagonist and TNFR2 agonist were administered peripherally at different stages of disease development in TNFR-humanized mice. We found that stimulating TNFR2 before onset of symptoms leads to improved response to anti-TNFR1 therapeutic treatment. This sequential treatment was more effective in decreasing paralysis symptoms and demyelination, when compared to single treatments. Interestingly, the frequency of the different immune cell subsets is unaffected by TNFR modulation. Nevertheless, treatment with only a TNFR1 antagonist increases T-cell infiltration in the central nervous system (CNS) and B-cell cuffing at the perivascular sites, whereas a TNFR2 agonist promotes Treg CNS accumulation. Our findings highlight the complicated nature of TNF signaling which requires a timely balance of selective activation and inhibition of TNFRs in order to exert therapeutic effects in the context of CNS autoimmunity.
Collapse
Affiliation(s)
- Valentina Pegoretti
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Science (GELIFES), University of Groningen, 9747 AG, Groningen, The Netherlands
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Centre Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Jan Bauer
- Division of Neuroimmunology, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Roman Fischer
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Centre Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Iskra Paro
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Science (GELIFES), University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Wanda Douwenga
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Science (GELIFES), University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Centre Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Centre Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Evelien Houben
- Neuroimmune Connections and Repair (NIC&R) Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Hasselt, Belgium
- University MS Centre, 3590, Hasselt/Pelt, Belgium
| | - Bieke Broux
- Neuroimmune Connections and Repair (NIC&R) Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Hasselt, Belgium
- University MS Centre, 3590, Hasselt/Pelt, Belgium
| | - Niels Hellings
- Neuroimmune Connections and Repair (NIC&R) Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Hasselt, Belgium
- University MS Centre, 3590, Hasselt/Pelt, Belgium
| | - Wia Baron
- Department Biomedical Sciences of Cells and Systems (BSCS), Section Molecular Neurobiology, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
| | - Jon D Laman
- Department Pathology and Medical Biology, University Medical Centre Groningen (UMCG), University of Groningen, 9713 GZ, Groningen, The Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Science (GELIFES), University of Groningen, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
4
|
Oliveira CA, Mercês ÉAB, Portela FS, De Benedictis JM, De Benedictis LM, da Silva AVB, Campanati JDAG, de Melo FF, Oliveira MV, de Magalhães ACM, Soares TDJ, Amaral LSDB. Benefits of high-intensity interval training compared to continuous training to reduce apoptotic markers in female rats with cisplatin nephrotoxicity - possible modulatory role of IL-11. Apoptosis 2023; 28:566-575. [PMID: 36653732 DOI: 10.1007/s10495-023-01816-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2023] [Indexed: 01/20/2023]
Abstract
Apoptotic signaling pathways are involved in acute kidney injury (AKI) induced by the antineoplastic drug cisplatin (Cis). Mechanical stress is known to increase interleukin (IL) -11, a pleiotropic cytokine with antiapoptotic and antinecrotic effects. We compared the impact of high-intensity interval training (HIIT) with low-intensity continuous training (LICT) and moderate-intensity continuous training (MICT) on renal levels of IL-11 and the expression of apoptotic markers in female rats with nephrotoxicity induced by Cis. For that, the animals were divided into five groups (n = 7): control and sedentary (C + S); Cis and sedentary (Cis + S); Cis and LICT (Cis + LICT); Cis and MICT (Cis + MICT) and Cis and HIIT (Cis + HIIT). At the end of 8 weeks of treadmill running, the rats received a single injection of Cis (5 mg/kg), and 7 days later they were euthanized. Serum and kidney samples were collected to assess the blood urea nitrogen (BUN), gene expression of TNF receptor 1 (TNFR1) and 2 (TNFR2), caspase-3, (p38) MAPK (MAPK14), p53, Bax, Bak, Bcl-2, and Bcl-xL, renal levels of IL-11, IL-8, and p53, and immunolocalization of cleaved caspase-3, Bax, Bcl-2, and (p38) MAPK in renal tissue. Our data indicate that all trained groups showed a significant intensity-dependent increase in renal levels of IL-11 associated with reduced local expression of proapoptotic and increased antiapoptotic markers, but these effects were more pronounced with HIIT. So, HIIT appears to provide superior renoprotection than traditional continuous training by modulating apoptotic signaling pathways, and this effect can be related to the increase in renal levels of IL-11.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Júlia Mafra De Benedictis
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Laís Mafra De Benedictis
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Antônio Victor Brito da Silva
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - João de Assis Gonçalves Campanati
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Márcio Vasconcelos Oliveira
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Amélia Cristina Mendes de Magalhães
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil.
| |
Collapse
|
5
|
Ren B, Tan L, Song Y, Li D, Xue B, Lai X, Gao Y. Cerebral Small Vessel Disease: Neuroimaging Features, Biochemical Markers, Influencing Factors, Pathological Mechanism and Treatment. Front Neurol 2022; 13:843953. [PMID: 35775047 PMCID: PMC9237477 DOI: 10.3389/fneur.2022.843953] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/12/2022] [Indexed: 01/15/2023] Open
Abstract
Cerebral small vessel disease (CSVD) is the most common chronic vascular disease involving the whole brain. Great progress has been made in clinical imaging, pathological mechanism, and treatment of CSVD, but many problems remain. Clarifying the current research dilemmas and future development direction of CSVD can provide new ideas for both basic and clinical research. In this review, the risk factors, biological markers, pathological mechanisms, and the treatment of CSVD will be systematically illustrated to provide the current research status of CSVD. The future development direction of CSVD will be elucidated by summarizing the research difficulties.
Collapse
Affiliation(s)
- Beida Ren
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditonal Chinese Medicine, Beijing, China
| | - Ling Tan
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuebo Song
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Danxi Li
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditonal Chinese Medicine, Beijing, China
| | - Bingjie Xue
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditonal Chinese Medicine, Beijing, China
| | - Xinxing Lai
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Gao
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Bernier RA, Banks SJ, Panizzon MS, Andrews MJ, Jacobs EG, Galasko DR, Shepherd AL, Akassoglou K, Sundermann EE, for the Alzheimer's Disease Neuroimaging Initiative. The neuroinflammatory marker sTNFR2 relates to worse cognition and tau in women across the Alzheimer's disease spectrum. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12284. [PMID: 35386474 PMCID: PMC8973901 DOI: 10.1002/dad2.12284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 11/27/2022]
Abstract
Introduction Despite women showing greater Alzheimer's disease (AD) prevalence, tau burden, and immune/neuroinflammatory response, whether neuroinflammation impacts cognition differently in women versus men and the biological basis of this impact remain unknown. We examined sex differences in how cerebrospinal fluid (CSF) neuroinflammation relates to cognition across the aging-mild cognitive impairment (MCI)-AD continuum and the mediating role of phosphorylated tau (p-tau) versus other AD biomarkers. Methods Participants included 284 individuals from the Alzheimer's Disease Neuroimaging Initiative study. CSF neuroinflammatory markers included interleukin-6, tumor necrosis factor α, soluble tumor necrosis factor receptor 2 (sTNFR2), and chitinase-3-like protein 1. AD biomarkers were CSF p-tau181 and amyloid beta1-42 levels and magnetic resonance imaging measures of hippocampal and white matter hyperintensity volumes. Results We found a sex-by-sTNFR2 interaction on Mini-Mental State Examination and Clinical Dementia Rating-Sum of Boxes. Higher levels of sTNFR2 related to poorer cognition in women only. Among biomarkers, only p-tau181 eliminated the female-specific relationships between neuroinflammation and cognition. Discussion Women may be more susceptible than men to the adverse effects of sTNFR2 on cognition with a potential etiological link with tau to these effects.
Collapse
Affiliation(s)
- Rachel A. Bernier
- Department of NeuroscienceUniversity of California, San DiegoSan DiegoCaliforniaUSA
| | - Sarah J. Banks
- Department of NeuroscienceUniversity of California, San DiegoSan DiegoCaliforniaUSA
| | - Matthew S. Panizzon
- Department of PsychiatryUniversity of California, San DiegoSan DiegoCaliforniaUSA
- Center for Behavior Genetics of AgingUniversity of California, San DiegoSan DiegoCaliforniaUSA
| | - Murray J. Andrews
- Department of NeuroscienceUniversity of California, San DiegoSan DiegoCaliforniaUSA
| | - Emily G. Jacobs
- Department of Psychological and Brain SciencesUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Douglas R. Galasko
- Department of NeuroscienceUniversity of California, San DiegoSan DiegoCaliforniaUSA
| | - Alyx L. Shepherd
- Department of NeuroscienceUniversity of California, San DiegoSan DiegoCaliforniaUSA
| | - Katerina Akassoglou
- Gladstone UCSF Center for Neurovascular Brain ImmunologySan FranciscoCaliforniaUSA
- Gladstone Institute of Neurological DiseaseSan FranciscoCaliforniaUSA
- Department of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Erin E. Sundermann
- Department of PsychiatryUniversity of California, San DiegoSan DiegoCaliforniaUSA
| | | |
Collapse
|
7
|
Quach TD, Huang W, Sahu R, Diadhiou CM, Raparia C, Johnson R, Leung TM, Malkiel S, Ricketts PG, Gallucci S, Tükel Ç, Jacob CO, Lesser ML, Zou YR, Davidson A. Context dependent induction of autoimmunity by TNF signaling deficiency. JCI Insight 2022; 7:149094. [PMID: 35104241 PMCID: PMC8983147 DOI: 10.1172/jci.insight.149094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
TNF inhibitors are widely used to treat inflammatory diseases; however, 30%–50% of treated patients develop new autoantibodies, and 0.5%–1% develop secondary autoimmune diseases, including lupus. TNF is required for formation of germinal centers (GCs), the site where high-affinity autoantibodies are often made. We found that TNF deficiency in Sle1 mice induced TH17 T cells and enhanced the production of germline encoded, T-dependent IgG anti-cardiolipin antibodies but did not induce GC formation or precipitate clinical disease. We then asked whether a second hit could restore GC formation or induce pathogenic autoimmunity in TNF-deficient mice. By using a range of immune stimuli, we found that somatically mutated autoantibodies and clinical disease can arise in the setting of TNF deficiency via extrafollicular pathways or via atypical GC-like pathways. This breach of tolerance may be due to defects in regulatory signals that modulate the negative selection of pathogenic autoreactive B cells.
Collapse
Affiliation(s)
- Tam D Quach
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Weiqing Huang
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Ranjit Sahu
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Catherine Mm Diadhiou
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Chirag Raparia
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Roshawn Johnson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Tung Ming Leung
- Biostatistics Unit, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Susan Malkiel
- Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Peta-Gay Ricketts
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Stefania Gallucci
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, United States of America
| | - Çagla Tükel
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, United States of America
| | - Chaim O Jacob
- Department of Medicine, University of Southern California, Los Angeles, United States of America
| | - Martin L Lesser
- Biostatistics Unit, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Yong-Rui Zou
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Anne Davidson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, United States of America
| |
Collapse
|
8
|
Litak J, Mazurek M, Kulesza B, Szmygin P, Litak J, Kamieniak P, Grochowski C. Cerebral Small Vessel Disease. Int J Mol Sci 2020; 21:ijms21249729. [PMID: 33419271 PMCID: PMC7766314 DOI: 10.3390/ijms21249729] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 01/18/2023] Open
Abstract
Cerebral small vessel disease (CSVD) represents a cluster of various vascular disorders with different pathological backgrounds. The advanced vasculature net of cerebral vessels, including small arteries, capillaries, arterioles and venules, is usually affected. Processes of oxidation underlie the pathology of CSVD, promoting the degenerative status of the epithelial layer. There are several classifications of cerebral small vessel diseases; some of them include diseases such as Binswanger’s disease, leukoaraiosis, cerebral microbleeds (CMBs) and lacunar strokes. This paper presents the characteristics of CSVD and the impact of the current knowledge of this topic on the diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Jakub Litak
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (B.K.); (P.S.); (P.K.)
- Department of Immunology, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| | - Marek Mazurek
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (B.K.); (P.S.); (P.K.)
| | - Bartłomiej Kulesza
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (B.K.); (P.S.); (P.K.)
| | - Paweł Szmygin
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (B.K.); (P.S.); (P.K.)
| | - Joanna Litak
- St. John’s Cancer Center in Lublin, 20-090 Lublin, Poland;
| | - Piotr Kamieniak
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (B.K.); (P.S.); (P.K.)
| | - Cezary Grochowski
- Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland;
- Laboratory of Virtual Man, Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
9
|
Atretkhany KSN, Gogoleva VS, Drutskaya MS, Nedospasov SA. Distinct modes of TNF signaling through its two receptors in health and disease. J Leukoc Biol 2020; 107:893-905. [PMID: 32083339 DOI: 10.1002/jlb.2mr0120-510r] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/03/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022] Open
Abstract
TNF is a key proinflammatory and immunoregulatory cytokine whose deregulation is associated with the development of autoimmune diseases and other pathologies. Recent studies suggest that distinct functions of TNF may be associated with differential engagement of its two receptors: TNFR1 or TNFR2. In this review, we discuss the relative contributions of these receptors to pathogenesis of several diseases, with the focus on autoimmunity and neuroinflammation. In particular, we discuss the role of TNFRs in the development of regulatory T cells during neuroinflammation and recent findings concerning targeting TNFR2 with agonistic and antagonistic reagents in various murine models of autoimmune and neuroinflammatory disorders and cancer.
Collapse
Affiliation(s)
- Kamar-Sulu N Atretkhany
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | - Violetta S Gogoleva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia.,Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
10
|
Santinon F, Batignes M, Mebrek ML, Biton J, Clavel G, Hervé R, Lemeiter D, Breckler M, Busato F, Tost J, Ziol M, Boissier MC, Decker P, Semerano L, Bessis N. Involvement of Tumor Necrosis Factor Receptor Type II in FoxP3 Stability and as a Marker of Treg Cells Specifically Expanded by Anti-Tumor Necrosis Factor Treatments in Rheumatoid Arthritis. Arthritis Rheumatol 2020; 72:576-587. [PMID: 31609517 DOI: 10.1002/art.41134] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 10/08/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To study the involvement of Treg cells expressing tumor necrosis factor receptor type II (TNFRII) in exerting control of inflammation in experimental models and in the response to anti-TNF treatments in patients with rheumatoid arthritis (RA) or spondyloarthritis (SpA). METHODS The role of TNFRII in Treg cells was explored using a multilevel translational approach. Treg cell stability was evaluated by analyzing the methylation status of the Foxp3 locus using bisulfite sequencing. Two models of inflammation (imiquimod-induced skin inflammation and delayed-type hypersensitivity arthritis [DTHA]) were induced in TNFRII-/- mice, with or without transfer of purified CD4+CD25+ cells from wild-type (WT) mice. In patients with RA and those with SpA, the evolution of the TNFRII+ Treg cell population before and after targeted treatment was monitored. RESULTS Foxp3 gene methylation in Treg cells was greater in TNFRII-/- mice than in WT mice (50% versus 36.7%). In cultured Treg cells, TNF enhanced the expression, maintenance, and proliferation of Foxp3 through TNFRII signaling. Imiquimod-induced skin inflammation and DTHA were aggravated in TNFRII-/- mice (P < 0.05 for mice with skin inflammation and P < 0.0001 for mice with ankle swelling during DTHA compared to WT mice). Adoptive transfer of WT mouse Treg cells into TNFRII-/- mice prevented aggravation of arthritis. In patients with RA receiving anti-TNF treatments, but not those receiving tocilizumab, the frequency of TNFRII+ Treg cells was increased at 3 months of treatment compared to baseline (mean ± SEM 65.2 ± 3.1% versus 49.1 ± 5.5%; P < 0.01). In contrast, in anti-TNF-treated patients with SpA, the frequency of TNFRII+ Treg cells was not modified. CONCLUSION TNFRII expression identifies a subset of Treg cells that are characterized by stable expression of Foxp3 via gene hypomethylation, and adoptive transfer of TNFRII-expressing Treg cells ameliorates inflammation in experimental models. Expansion and activation of TNFRII+ Treg cells may be one of the mechanisms by which anti-TNF agents control inflammation in RA, but not in SpA.
Collapse
Affiliation(s)
- François Santinon
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Maxime Batignes
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Majda Lyna Mebrek
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Jerôme Biton
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Gaëlle Clavel
- INSERM UMR 1125, Université Sorbonne Paris Cité, Université Paris 13, and Fondation Adolphe De Rothschild, Paris, France
| | - Roxane Hervé
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Delphine Lemeiter
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Magali Breckler
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Florence Busato
- Commissariat à l'énergie atomique et aux énergies alternatives, Paris, France
| | - Jorg Tost
- Commissariat à l'énergie atomique et aux énergies alternatives, Paris, France
| | - Marianne Ziol
- Hôpital Jean-Verdier, AP-HP, INSERM UMR 1162, Université Paris Descartes, and Université Paris Diderot, Paris, France
| | - Marie-Christophe Boissier
- INSERM UMR 1125, Université Sorbonne Paris Cité, Université Paris 13, Hôpital Avicenne, Hôpital Jean-Verdier, Hôpital René-Muret, and AP-HP, Paris, France
| | - Patrice Decker
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| | - Luca Semerano
- INSERM UMR 1125, Université Sorbonne Paris Cité, Université Paris 13, Hôpital Avicenne, Hôpital Jean-Verdier, Hôpital René-Muret, and AP-HP, Paris, France
| | - Natacha Bessis
- INSERM UMR 1125, Université Sorbonne Paris Cité, and Université Paris 13, Paris, France
| |
Collapse
|
11
|
TNFR1 inhibition with a Nanobody protects against EAE development in mice. Sci Rep 2017; 7:13646. [PMID: 29057962 PMCID: PMC5651799 DOI: 10.1038/s41598-017-13984-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/04/2017] [Indexed: 01/09/2023] Open
Abstract
TNF has as detrimental role in multiple sclerosis (MS), however, anti-TNF medication is not working. Selective TNF/TNFR1 inhibition whilst sparing TNFR2 signaling reduces the pro-inflammatory effects of TNF but preserves the important neuroprotective signals via TNFR2. We previously reported the generation of a Nanobody-based selective inhibitor of human TNFR1, TROS that will be tested in experimental autoimmune encephalomyelitis (EAE). We specifically antagonized TNF/TNFR1 signaling using TROS in a murine model of MS, namely MOG35-55-induced EAE. Because TROS does not cross-react with mouse TNFR1, we generated mice expressing human TNFR1 in a mouse TNFR1-knockout background (hTNFR1 Tg), and we determined biodistribution of 99mTc-TROS and effectiveness of TROS in EAE in those mice. Biodistribution analysis demonstrated that intraperitoneally injected TROS is retained more in organs of hTNFR1 Tg mice compared to wild type mice. TROS was also detected in the cerebrospinal fluid (CSF) of hTNFR1 Tg mice. Prophylactic TROS administration significantly delayed disease onset and ameliorated its symptoms. Moreover, treatment initiated early after disease onset prevented further disease development. TROS reduced spinal cord inflammation and neuroinflammation, and preserved myelin and neurons. Collectively, our data illustrate that TNFR1 is a promising therapeutic target in MS.
Collapse
|
12
|
Deora A, Hegde S, Lee J, Choi CH, Chang Q, Lee C, Eaton L, Tang H, Wang D, Lee D, Michalak M, Tomlinson M, Tao Q, Gaur N, Harvey B, McLoughlin S, Labkovsky B, Ghayur T. Transmembrane TNF-dependent uptake of anti-TNF antibodies. MAbs 2017; 9:680-695. [PMID: 28323513 PMCID: PMC5419086 DOI: 10.1080/19420862.2017.1304869] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
TNF-α (TNF), a pro-inflammatory cytokine is synthesized as a 26 kDa protein, anchors in the plasma membrane as transmembrane TNF (TmTNF), and is subjected to proteolysis by the TNF-α converting enzyme (TACE) to release the 15 kDa form of soluble TNF (sTNF). TmTNF and sTNF interact with 2 distinct receptors, TNF-R1 (p55) and TNF-R2 (p75), to mediate the multiple biologic effects of TNF described to date. Several anti-TNF biologics that bind to both forms of TNF and block their interactions with the TNF receptors are now approved for the treatment of a variety of immune-mediated diseases. Several reports suggest that binding of anti-TNFs to TmTNF delivers an outside-to-inside ‘reverse’ signal that may also contribute to the efficacy of anti-TNFs. Some patients, however, develop anti-TNF drug antibody responses (ADA or immunogenicity). Here, we demonstrate biochemically that TmTNF is transiently expressed on the surface of lipopolysaccharide-stimulated primary human monocytes, macrophages, and monocyte-derived dendritic cells (DCs) and expression of TmTNF on the cell surface is enhanced following treatment of cells with TAPI-2, a TACE inhibitor. Importantly, binding of anti-TNFs to TmTNF on DCs results in rapid internalization of the anti-TNF/TmTNF complex first into early endosomes and then lysosomes. The internalized anti-TNF is processed and anti-TNF peptides can be eluted from the surface of DCs. Finally, tetanus toxin peptides fused to anti-TNFs are presented by DCs to initiate T cell recall proliferation response. Collectively, these observations may provide new insights into understanding the biology of TmTNF, mode of action of anti-TNFs, biology of ADA response to anti-TNFs, and may help with the design of the next generation of anti-TNFs.
Collapse
Affiliation(s)
- Arun Deora
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | | | | | - Chee-Ho Choi
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | - Qing Chang
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | - Cheryl Lee
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | - Lucia Eaton
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | - Hua Tang
- b AbbVie Inc , North Chicago , IL , USA
| | | | - David Lee
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | | | | | - Qingfeng Tao
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | - Nidhi Gaur
- a AbbVie Bioresearch Center , Worcester , MA , USA
| | | | | | | | - Tariq Ghayur
- a AbbVie Bioresearch Center , Worcester , MA , USA
| |
Collapse
|
13
|
Targeting of Tumor Necrosis Factor Alpha Receptors as a Therapeutic Strategy for Neurodegenerative Disorders. Antibodies (Basel) 2015. [DOI: 10.3390/antib4040369] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
14
|
TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience 2015; 302:2-22. [DOI: 10.1016/j.neuroscience.2015.06.038] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/17/2015] [Accepted: 06/19/2015] [Indexed: 12/15/2022]
|
15
|
Arango-Dávila CA, Vera A, Londoño AC, Echeverri AF, Cañas F, Cardozo CF, Orozco JL, Rengifo J, Cañas CA. Soluble or soluble/membrane TNF-α inhibitors protect the brain from focal ischemic injury in rats. Int J Neurosci 2014; 125:936-40. [PMID: 25350870 DOI: 10.3109/00207454.2014.980906] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Tumor Necrosis Factor-alpha (TNF-α) is an immunomodulatory and proinflammatory cytokine implicated in neuro-inflammation and neuronal damage in response to cerebral ischemia. The present study tested the hypothesis that anti-TNF-α agents may be protective against cerebral infarction. Transient focal ischemia was artificially induced in anesthetized adult male Wistar rats (300-350 g) by middle cerebral artery occlusion (MCAO) with an intraluminal suture. TNF-α function was interfered with either a chimeric monoclonal antibody against TNF-α (infliximab-7 mg/kg) aiming to TNF-α soluble and membrane-attached form; or a chimeric fusion protein of TNF-α receptor-2 with a fragment crystallizable (Fc) region of IgG1 (etanercept-5 mg/kg) aiming for the TNF-α soluble form. Both agents were administered intraperitoneally 0 or 6 h after inducing ischemia. Infarct volume was measured by 2,3,5-triphenyltetrazolium chloride staining. Cerebral infarct volume was significantly reduced in either etanercept or infliximab-treated group compared with non-treated MCAO rats 24 h after reperfusion. These results suggest that anti-TNF-α agents may reduce focal ischemic injury in rats.
Collapse
Affiliation(s)
- C A Arango-Dávila
- a Unit of Psychiatry, Fundación Valle del Lili, Icesi University , Cali , Colombia
| | - A Vera
- b Department of Basic Sciences, School of Medicine, Caldas University , Manizales , Colombia
| | - A C Londoño
- c Department of Clinical Research, Unit of Clinical Pharmacology, Hospital General Universitario de Alicante, Miguel Hernández University , Alicante , Spain
| | - A F Echeverri
- d Unit of Rheumatology, Fundación Valle del Lili, Icesi University , Cali , Colombia
| | - F Cañas
- e Departament of Internal Medicine, Fundación Valle del Lili, CES University , Cali , Colombia
| | - C F Cardozo
- f Faculty of Natural Sciences, Icesi University , Cali , Colombia
| | - J L Orozco
- g Unit of Neurology. Fundación Valle del Lili, Icesi University , Cali , Colombia, and
| | - J Rengifo
- h Faculty of Natural Sciences, Icesi University , Cali , Colombia
| | - C A Cañas
- d Unit of Rheumatology, Fundación Valle del Lili, Icesi University , Cali , Colombia
| |
Collapse
|
16
|
Yang S, Wang Y, Mei K, Zhang S, Sun X, Ren F, Liu S, Yang Z, Wang X, Qin Z, Chang Z. Tumor necrosis factor receptor 2 (TNFR2)·interleukin-17 receptor D (IL-17RD) heteromerization reveals a novel mechanism for NF-κB activation. J Biol Chem 2014; 290:861-71. [PMID: 25378394 DOI: 10.1074/jbc.m114.586560] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
TNF receptor 2 (TNFR2) exerts diverse roles in the pathogenesis of inflammatory and autoimmune diseases. Here, we report that TNFR2 but not TNFR1 forms a heteromer with interleukin-17 receptor D (IL-17RD), also named Sef, to activate NF-κB signaling. TNFR2 associates with IL-17RD, leading to mutual receptor aggregation and TRAF2 recruitment, which further activate the downstream cascade of NF-κB signaling. Depletion of IL-17RD impaired TNFR2-mediated activation of NF-κB signaling. Importantly, IL-17RD was markedly increased in renal tubular epithelial cells in nephritis rats, and a strong interaction of TNFR2 and IL-17RD was observed in the renal epithelia. The IL-17RD·TNFR2 complex in activation of NF-κB may explain the role of TNFR2 in inflammatory diseases including nephritis.
Collapse
Affiliation(s)
- Shigao Yang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China, National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinyin Wang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kunrong Mei
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing 100084, China
| | - Sen Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union medical college, Beijing, 100050, China, and
| | - Xiaojun Sun
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fangli Ren
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sihan Liu
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zi Yang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinquan Wang
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing 100084, China
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhijie Chang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China,
| |
Collapse
|
17
|
Reuss R, Mistarz M, Mirau A, Kraus J, Bödeker RH, Oschmann P. FADD is upregulated in relapsing remitting multiple sclerosis. Neuroimmunomodulation 2014; 21:221-5. [PMID: 24603611 DOI: 10.1159/000356522] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 10/18/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To elucidate the role of tumor necrosis factor (TNF) receptor signal transduction in multiple sclerosis (MS). METHODS We performed a cross-sectional analysis of the gene expression of TNF receptor-associated death domain protein (TRADD) and Fas-associated death domain protein (FADD) in peripheral blood leukocytes of 23 relapsing remitting (RR), 19 secondary progressive (SP) and 12 primary progressive (PP) MS patients, as well as of 29 healthy controls by quantitative RT-PCR. Additionally, we monitored a subgroup of 15 RR MS patients longitudinally every 3 months over the time period of 9 months. RESULTS FADD expression was significantly elevated in RR MS patients compared to the other disease courses (p < 0.048). The median of FADD expression was elevated in the RR MS patient groups compared to the healthy group, but this was not significant (p < 0.053). The median of TRADD expression was elevated in the patient groups compared to the healthy group, but this was not significant (p < 0.14). Neither variable changed significantly over the time course of 9 months. CONCLUSION FADD elevation in leukocytes might be interpreted as the molecular equivalent of an elevated general inflammatory activity in RR MS patients compared to other disease courses. FADD elevation in RR MS reinforces the concept that different pathophysiological and immunological processes sustain RR MS and SP or PP MS.
Collapse
Affiliation(s)
- Reinhard Reuss
- Department of Neurology, Bezirkskrankenhaus Bayreuth, Bayreuth, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Effects of MRP8, LPS, and lenalidomide on the expressions of TNF-α , brain-enriched, and inflammation-related microRNAs in the primary astrocyte culture. ScientificWorldJournal 2013; 2013:208309. [PMID: 24170980 PMCID: PMC3793319 DOI: 10.1155/2013/208309] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 08/22/2013] [Indexed: 01/12/2023] Open
Abstract
Astrocytes are now recognized as a heterogeneous class of cells with many important and diverse functions in healthy and diseased central nervous system (CNS). MicroRNAs (miRNAs) are small, noncoding RNAs which may have key roles in astrocytes activation in response to various stimuli. We performed quantitative real-time PCR (qPCR) to detect changes in the expressions of brain-enriched miRNAs (124, 134, 9, 132, and 138), inflammation-related miRNAs (146a, 21, 181a, 221, and 222), and tumor necrosis factor alpha (TNF- α ) in the rat primary astrocyte cultures after stimulation with myeloid-related protein 8 (MRP8) and lipopolysaccharides (LPS). Further, we inhibited the expression of TNF- α in the astrocytes by using TNF- α inhibitor (lenalidomide) and tested for the first time the effect of this inhibition on the expressions of the same tested miRNAs. Stimulation of the astrocytes with MRP8 or LPS leads to significant upregulation of miRNAs (124, 134, 9, 132, 146a, 21, 181a, 221, and 222), while miRNA-138 was downregulated. TNF- α inhibition with lenalidomide leads to opposite expressions of the tested miRNAs. These miRNAs may play an important role in activation of the astrocytes and may be a novel target for cell-specific therapeutic interventions in multiple CNS diseases.
Collapse
|
19
|
Venkatesh D, Ernandez T, Rosetti F, Batal I, Cullere X, Luscinskas FW, Zhang Y, Stavrakis G, García-Cardeña G, Horwitz BH, Mayadas TN. Endothelial TNF receptor 2 induces IRF1 transcription factor-dependent interferon-β autocrine signaling to promote monocyte recruitment. Immunity 2013; 38:1025-37. [PMID: 23623383 DOI: 10.1016/j.immuni.2013.01.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 01/23/2013] [Indexed: 02/08/2023]
Abstract
Endothelial-dependent mechanisms of mononuclear cell influx are not well understood. We showed that acute stimulation of murine microvascular endothelial cells expressing the tumor necrosis factor receptors TNFR1 and TNFR2 with the soluble cytokine TNF led to CXCR3 chemokine generation. The TNF receptors signaled through interferon regulatory factor-1 (IRF1) to induce interferon-β (IFN-β) and subsequent autocrine signaling via the type I IFN receptor and the transcription factor STAT1. Both TNFR2 and TNFR1 were required for IRF1-IFNβ signaling and, in human endothelial cells TNFR2 expression alone induced IFN-β signaling and monocyte recruitment. In vivo, TNFR1 was required for acute renal neutrophil and monocyte influx after systemic TNF treatment, whereas the TNFR2-IRF1-IFN-β autocrine loop was essential only for macrophage accumulation. In a chronic model of proliferative nephritis, IRF1 and renal-expressed TNFR2 were essential for sustained macrophage accumulation. Thus, our data identify a pathway in endothelial cells that selectively recruits monocytes during a TNF-induced inflammatory response.
Collapse
Affiliation(s)
- Deepak Venkatesh
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Reuß R, Mirau A, Mistarz M, Kraus J, Bödeker RH, Oschmann P. TRAF2 is upregulated in relapsing-remitting multiple sclerosis. Neuroimmunomodulation 2013; 20:177-83. [PMID: 23595117 DOI: 10.1159/000346794] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 12/14/2012] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To elucidate the role of tumor necrosis factor (TNF) receptor signal transduction in multiple sclerosis (MS). METHODS We performed a cross-sectional analysis of the gene expression of TRAF2 (TNF receptor-associated factor 2) and RIP (receptor-interacting protein) in peripheral blood leukocytes of 23 relapsing-remitting (RR), 19 secondary progressive (SP) and 12 primary progressive (PP) MS patients as well as of 29 healthy controls by quantitative RT-PCR. Additionally, we monitored a subgroup of 15 RRMS patients longitudinally every 3 months over a 9-month time period. RESULTS TRAF2 expression was significantly elevated in RRMS patients compared to the other disease courses (p<0.005, respectively) and the control group (p<0.009). RIP expression was significantly elevated in the patient groups compared to the healthy group (phealthy-RR<0.002; phealthy-PP<0.003; phealthy-SP<0.06). Neither variable changed over the 9-month time course. CONCLUSION TRAF2 and RIP1 elevation in leukocytes might be interpreted as the molecular equivalent of an elevated general inflammatory activity in MS patients compared to healthy control persons. TRAF2 elevation in RRMS reinforces the concept that different pathophysiological and immunological processes sustain RRMS and SPMS or PPMS.
Collapse
Affiliation(s)
- Reinhard Reuß
- Department of Neurology, Bezirkskrankenhaus Bayreuth, Bayreuth, Germany.
| | | | | | | | | | | |
Collapse
|
21
|
Tumor necrosis factor gene variation predicts hippocampus volume in healthy individuals. Biol Psychiatry 2012; 72:655-62. [PMID: 22554453 DOI: 10.1016/j.biopsych.2012.04.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 03/20/2012] [Accepted: 04/05/2012] [Indexed: 12/25/2022]
Abstract
BACKGROUND Cytokines such as tumor necrosis factor (TNF) α have been implicated in neurodegeneration relevant to various neuropsychiatric disorders. Little is known about the genetic predisposition to neurodegenerative properties of cytokine genes on brain function and on hippocampus (HC) function in particular. In this study we investigate the neurodegenerative role of TNF polymorphisms on brain morphology in healthy individuals. METHODS Voxel-based morphometry was used in a large sample of healthy individuals (n = 303) to analyze the associations between genetic variants of TNF (rs1800629; rs361525) and brain morphology (gray matter concentration). RESULTS In a region of interest analysis of the HC, for rs1800629, we observed a strong genotype effect on bilateral HC gray matter concentration. Carriers of one or two A-alleles had significantly smaller volumes compared with GG-homozygotes. For rs361525, a similar effect was observed at almost the same location, with the A-allele resulting in smaller HC volumes compared with GG homozygotes. CONCLUSIONS The findings suggest a neurodegenerative role of the A-alleles of the TNF single nucleotide polymorphisms rs1800629 (-308G/A) and rs361525 (-238G/A) on hippocampal volumes in healthy individuals. Future imaging studies on the role of these single nucleotide polymorphisms in psychiatric populations of diseases with neurodegenerative components are warranted.
Collapse
|
22
|
Alba MA, Espígol-Frigolé G, Prieto-González S, Tavera-Bahillo I, García-Martínez A, Butjosa M, Hernández-Rodríguez J, Cid MC. Central nervous system vasculitis: still more questions than answers. Curr Neuropharmacol 2012; 9:437-48. [PMID: 22379458 PMCID: PMC3151598 DOI: 10.2174/157015911796557920] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 11/30/2010] [Accepted: 11/30/2010] [Indexed: 12/19/2022] Open
Abstract
The central nervous system (CNS) may be involved by a variety of inflammatory diseases of blood vessels. These include primary angiitis of the central nervous system (PACNS), a rare disorder specifically targeting the CNS vasculature, and the systemic vasculitides which may affect the CNS among other organs and systems. Both situations are severe and convey a guarded prognosis. PACNS usually presents with headache and cognitive impairment. Focal symptoms are infrequent at disease onset but are common in more advanced stages. The diagnosis of PACNS is difficult because, although magnetic resonance imaging is almost invariably abnormal, findings are non specific. Angiography has limited sensitivity and specificity. Brain and leptomeningeal biopsy may provide a definitive diagnosis when disclosing blood vessel inflammation and are also useful to exclude other conditions presenting with similar findings. However, since lesions are segmental, a normal biopsy does not completely exclude PACNS. Secondary CNS involvement by systemic vasculitis occurs in less than one fifth of patients but may be devastating. A prompt recognition and aggressive treatment is crucial to avoid permanent damage and dysfunction. Glucocorticoids and cyclophosphamide are recommended for patients with PACNS and for patients with secondary CNS involvement by small-medium-sized systemic vasculitis. CNS involvement in large-vessel vasculitis is usually managed with high-dose glucocorticoids (giant-cell arteritis) or glucocorticoids and immunosuppressive agents (Takayasu's disease). However, in large vessel vasculitis, where CNS symptoms are usually due to involvement of extracranial arteries (Takayasu's disease) or proximal portions of intracranial arteries (giant-cell arteritis), revascularization procedures may also have an important role.
Collapse
Affiliation(s)
- Marco A Alba
- Vasculitis Research Unit, Department of Systemic Autoimmune Diseases, Hospital Clinic, University of Barcelona, Institut d´Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Villarroel 170, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Bae JS, Choi JK, Moon JH, Kim EC, Croft M, Lee HW. Novel transmembrane protein 126A (TMEM126A) couples with CD137L reverse signals in myeloid cells. Cell Signal 2012; 24:2227-36. [PMID: 22885069 DOI: 10.1016/j.cellsig.2012.07.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/23/2012] [Accepted: 07/25/2012] [Indexed: 11/18/2022]
Abstract
Members of the TNF family can promote signals in myeloid cells and both positively and negatively regulate the production of pro-inflammatory cytokines depending on the target myeloid cell type. Using the yeast-two hybrid system, we identified transmembrane protein 126A (TMEM126A) as a binding partner for CD137L (4-1BB ligand). We found that TMEM126A associated and co-localized with CD137L in a mouse macrophage cell line and knockdown of TMEM126A with siRNA abolished the CD137L-induced tyrosine phosphorylation as well as the up-regulation of M-CSF, IL-1β and TN-C expressions. Knockdown of TMEM126A also blocked the down-regulation of IL-1β and IL-6 expressions induced by CD137L in thioglycollate-elicited primary peritoneal macrophages. Knockdown of TMEM126A by stable retroviral TMEM126A shRNA transduction also abolished CD137L-induced tyrosine phosphorylation and cell adherence. These findings identify a novel molecule that bridges TNF family cytokines and pro-inflammatory cytokine secretion in myeloid cells.
Collapse
Affiliation(s)
- Jun-Sang Bae
- Institute of Oral Biology, School of Dentistry, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
24
|
Cabal-Hierro L, Lazo PS. Signal transduction by tumor necrosis factor receptors. Cell Signal 2012; 24:1297-305. [PMID: 22374304 DOI: 10.1016/j.cellsig.2012.02.006] [Citation(s) in RCA: 327] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/14/2012] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor (TNF) is a key mediator in the inflammatory response which is implicated in the onset of a number of diseases. Research on TNF led to the characterization of the largest family of cytokines known until now, the TNF superfamily, which exert their biological effects through the interaction with transmembrane receptors of the TNFR superfamily. TNF itself exerts its biological effects interacting with two different receptors: TNFR1 and TNFR2. TNFR1 presents a death domain on its intracellular region. In contrast to TNFR1, TNFR2 does not have a death domain. Activation of TNFR1 implies the consecutive formation of two different TNF receptor signalling complexes. Complex I controls the expression of antiapoptotic proteins that prevent the triggering of cell death processes, whereas Complex II triggers cell death processes. TNFR2 only signals for antiapoptotic reactions. However, recent evidence indicates that TNFR2 also signals to induce TRAF2 degradation. TRAF2 is a key mediator in signal transduction of both TNFR1 and TNFR2. Thus, this novel signalling pathway has two important implications: on one hand, it represents an auto regulatory loop for TNFR2; on the other hand, when this signal is triggered TNFR1 activity is modified so that antiapoptotic pathways are inhibited and apoptotic reactions are enhanced.
Collapse
Affiliation(s)
- Lucía Cabal-Hierro
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33071 Oviedo, Spain
| | | |
Collapse
|
25
|
Kim S, Steelman AJ, Zhang Y, Kinney HC, Li J. Aberrant upregulation of astroglial ceramide potentiates oligodendrocyte injury. Brain Pathol 2012; 22:41-57. [PMID: 21615590 PMCID: PMC4500118 DOI: 10.1111/j.1750-3639.2011.00501.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 05/16/2011] [Indexed: 11/30/2022] Open
Abstract
Oligodendroglial injury is a pathological hallmark of many human white matter diseases, including multiple sclerosis (MS) and periventricular leukomalacia (PVL). Critical regulatory mechanisms of oligodendroglia destruction, however, remain incompletely understood. Ceramide, a bioactive sphingolipid pivotal to sphingolipid metabolism pathways, regulates cell death in response to diverse stimuli and has been implicated in neurodegenerative disorders. We report here that ceramide accumulates in reactive astrocytes in active lesions of MS and PVL, as well as in animal models of demyelination. Serine palmitoyltransferase, the rate-limiting enzyme for ceramide de novo biosynthesis, was consistently upregulated in reactive astrocytes in the cuprizone mouse model of demyelination. Mass spectrometry confirmed the upregulation of specific ceramides during demyelination, and revealed a concomitant increase of sphingosine and a suppression of sphingosine-1-phosphate, a potent signaling molecule with key roles in cell survival and mitogenesis. Importantly, this altered sphingolipid metabolism during demyelination was restored upon active remyelination. In culture, ceramide acted synergistically with tumor necrosis factor, leading to apoptotic death of oligodendroglia in an astrocyte-dependent manner. Taken together, our findings implicate that disturbed sphingolipid pathways in reactive astrocytes may indirectly contribute to oligodendroglial injury in cerebral white matter disorders.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Cell Death/physiology
- Ceramides/adverse effects
- Ceramides/biosynthesis
- Coculture Techniques
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Humans
- Infant, Newborn
- Leukomalacia, Periventricular/metabolism
- Leukomalacia, Periventricular/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Oligodendroglia/drug effects
- Oligodendroglia/metabolism
- Oligodendroglia/pathology
- Primary Cell Culture
- Rats
- Rats, Sprague-Dawley
- Up-Regulation/genetics
- Young Adult
Collapse
Affiliation(s)
- SunJa Kim
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Andrew J. Steelman
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Hannah C. Kinney
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
| | - Jianrong Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
26
|
Shore SA, Williams ES, Chen L, Benedito LAP, Kasahara DI, Zhu M. Impact of aging on pulmonary responses to acute ozone exposure in mice: role of TNFR1. Inhal Toxicol 2011; 23:878-88. [PMID: 22066571 DOI: 10.3109/08958378.2011.622316] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CONTEXT Chamber studies in adult humans indicate reduced responses to acute ozone with increasing age. Age-related changes in TNFα have been observed. TNFα induced inflammation is predominantly mediated through TNFR1. OBJECTIVE To examine the impact of aging on inflammatory responses to acute ozone exposure in mice and determine the role of TNFR1 in age-related differences. MATERIALS AND METHODS Wildtype and TNFR1 deficient (TNFR1(-/-)) mice aged 7 or 39 weeks were exposed to ozone (2 ppm for 3 h). Four hours after exposure, bronchoalveolar lavage (BAL) was performed and BAL cells, cytokines, chemokines, and protein were examined. RESULTS Ozone-induced increases in BAL neutrophils and in neutrophil chemotactic factors were lower in 39- versus 7-week-old wildtype, but not (TNFR1(-/-)) mice. There was no effect of TNFR1 genotype in 7-week-old mice, but in 39-week-old mice, BAL neutrophils and BAL concentrations of MCP-1, KC, MIP-2, IL-6 and IP-10 were significantly greater following ozone exposure in TNFR1(-/-) versus wildtype mice. BAL concentrations of the soluble form of the TNFR1 receptor (sTNFR1) were substantially increased in 39-week-old versus 7-week-old mice, regardless of exposure. DISCUSSION AND CONCLUSION The data suggest that increased levels of sTNFR1 in the lungs of the 39-week-old mice may neutralize TNFα and protect these older mice against ozone-induced inflammation.
Collapse
Affiliation(s)
- Stephanie A Shore
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Taoufik E, Tseveleki V, Chu SY, Tselios T, Karin M, Lassmann H, Szymkowski DE, Probert L. Transmembrane tumour necrosis factor is neuroprotective and regulates experimental autoimmune encephalomyelitis via neuronal nuclear factor-kappaB. ACTA ACUST UNITED AC 2011; 134:2722-35. [PMID: 21908876 DOI: 10.1093/brain/awr203] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumour necrosis factor mediates chronic inflammatory pathologies including those affecting the central nervous system, but non-selective tumour necrosis factor inhibitors exacerbate multiple sclerosis. In addition, TNF receptor SF1A, which encodes one of the tumour necrosis factor receptors, has recently been identified as a multiple sclerosis susceptibility locus in genome-wide association studies in large patient cohorts. These clinical data have emphasized the need for a better understanding of the beneficial effects of tumour necrosis factor during central nervous system inflammation. In this study, we present evidence that the soluble and transmembrane forms of tumour necrosis factor exert opposing deleterious and beneficial effects, respectively, in a multiple sclerosis model. We compared the effects, in experimental autoimmune encephalomyelitis, of selectively inhibiting soluble tumour necrosis factor, and of both soluble and transmembrane tumour necrosis factor. Blocking the action of soluble tumour necrosis factor, but not of soluble tumour necrosis factor and transmembrane tumour necrosis factor, protected mice against the clinical symptoms of experimental autoimmune encephalomyelitis. Therapeutic benefit was independent of changes in antigen-specific immune responses and focal inflammatory spinal cord lesions, but was associated with reduced overall central nervous system immunoreactivity, increased expression of neuroprotective molecules, and was dependent upon the activity of neuronal nuclear factor-κB, a downstream mediator of neuroprotective tumour necrosis factor/tumour necrosis factor receptor signalling, because mice lacking IκB kinase β in glutamatergic neurons were not protected by soluble tumour necrosis factor blockade. Furthermore, blocking the action of soluble tumour necrosis factor, but not of soluble tumour necrosis factor and transmembrane tumour necrosis factor, protected neurons in astrocyte-neuron co-cultures against glucose deprivation, an in vitro neurodegeneration model relevant for multiple sclerosis, and this was dependent upon contact between the two cell types. Our results show that soluble tumour necrosis factor promotes central nervous system inflammation, while transmembrane tumour necrosis factor is neuroprotective, and suggest that selective inhibition of soluble tumour necrosis factor may provide a new way forward for the treatment of multiple sclerosis and possibly other inflammatory central nervous system disorders.
Collapse
Affiliation(s)
- Era Taoufik
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 11521 Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Mor E, Cabilly Y, Goldshmit Y, Zalts H, Modai S, Edry L, Elroy-Stein O, Shomron N. Species-specific microRNA roles elucidated following astrocyte activation. Nucleic Acids Res 2011; 39:3710-23. [PMID: 21247879 PMCID: PMC3089466 DOI: 10.1093/nar/gkq1325] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 11/14/2010] [Accepted: 11/14/2010] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that play a central role in regulation of gene expression by binding to target genes. Many miRNAs were associated with the function of the central nervous system (CNS) in health and disease. Astrocytes are the CNS most abundant glia cells, providing support by maintaining homeostasis and by regulating neuronal signaling, survival and synaptic plasticity. Astrocytes play a key role in repair of brain insults, as part of local immune reactivity triggered by inflammatory or pathological conditions. Thus, astrocyte activation, or astrogliosis, is an important outcome of the innate immune response, which can be elicited by endotoxins such as lipopolysaccharide (LPS) and cytokines such as interferon-gamma (IFN-γ). The involvement of miRNAs in inflammation and stress led us to hypothesize that astrogliosis is mediated by miRNA function. In this study, we compared the miRNA regulatory layer expressed in primary cultured astrocyte derived from rodents (mice) and primates (marmosets) brains upon exposure to LPS and IFN-γ. We identified subsets of differentially expressed miRNAs some of which are shared with other immunological related systems while others, surprisingly, are mouse and rat specific. Of interest, these specific miRNAs regulate genes involved in the tumor necrosis factor-alpha (TNF-α) signaling pathway, indicating a miRNA-based species-specific regulation. Our data suggests that miRNA function is more significant in the mechanisms governing astrocyte activation in rodents compared to primates.
Collapse
Affiliation(s)
- Eyal Mor
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Yuval Cabilly
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Yona Goldshmit
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Harel Zalts
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Shira Modai
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Liat Edry
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Orna Elroy-Stein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
29
|
Bae JS, Kim HS, Park JH, Park SH, Lee HW. Cross-linking of CD137 ligand modulates immune responses of thioglycollate-elicited mouse peritoneal macrophages. Inflamm Res 2010; 60:467-73. [DOI: 10.1007/s00011-010-0289-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 11/24/2010] [Indexed: 11/29/2022] Open
|
30
|
Campbell IL, Hofer MJ, Pagenstecher A. Transgenic models for cytokine-induced neurological disease. Biochim Biophys Acta Mol Basis Dis 2009; 1802:903-17. [PMID: 19835956 DOI: 10.1016/j.bbadis.2009.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 10/01/2009] [Accepted: 10/02/2009] [Indexed: 12/22/2022]
Abstract
Considerable evidence supports the idea that cytokines are important mediators of pathophysiologic processes within the central nervous system (CNS). Numerous studies have documented the increased production of various cytokines in the human CNS in a variety of neurological and neuropsychiatric disorders. Deciphering cytokine actions in the intact CNS has important implications for our understanding of the pathogenesis and treatment of these disorders. One approach to address this problem that has been used widely employs transgenic mice with CNS-targeted production of different cytokines. Transgenic production of cytokines in the CNS of mice allows not only for the investigation of complex cellular responses at a localized level in the intact brain but also more closely recapitulates the expression of these mediators as found in disease states. As discussed in this review, the findings show that these transgenic animals exhibit wide-ranging structural and functional deficits that are linked to the development of distinct neuroinflammatory responses which are relatively specific for each cytokine. These cytokine-induced alterations often recapitulate those found in various human neurological disorders not only underscoring the relevance of these models but also reinforcing the clinicopathogenetic significance of cytokines in diseases of the CNS.
Collapse
Affiliation(s)
- Iain L Campbell
- School of Molecular and Microbial Biosciences and Bosch Institute, The University of Sydney, NSW 2006, Australia.
| | | | | |
Collapse
|
31
|
Kim DK, Lee SC, Lee HW. CD137 ligand-mediated reverse signals increase cell viability and cytokine expression in murine myeloid cells: involvement of mTOR/p70S6 kinase and Akt. Eur J Immunol 2009; 39:2617-28. [PMID: 19676073 DOI: 10.1002/eji.200939292] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cross-linking of CD137 ligand (CD137L), a member of the TNF family, with recombinant CD137-Fc (rCD137-Fc) protein enhanced adherence of bone marrow-derived macrophages, and increased the expression of ICAM-1, IL-1beta, IL-6, M-CSF and phosphotyrosine proteins. In RAW264.7 cells, a murine myeloid cell line, rCD137-Fc not only increased adherence but also cell multiplication, in a manner comparable to LPS or M-CSF. In addition, it up-regulated expression of IL-1beta, IL-1 receptor antagonist, IL-6, COX2, tenascin C, neuropeptide Y and M-CSF mRNA. Neutralization of M-CSF by incubating the RAW264.7 cells with anti-M-CSF mAb did not prevent the CD137L signal-induced viability. Viability was blocked by PP2, an Src tyrosine kinase inhibitor, rapamycin, an mTOR inhibitor and LY294002, a PI3K inhibitor, but not by Wortmannin, another PI3K inhibitor. Cross-linking of CD137L increased phosphorylation of Akt and p70S6 kinase. The latter was blocked by PP2, rapamycin or LY294002, but not by Wortmannin, whereas phosphorylation of Akt was blocked by LY294002 or Wortmannin. These findings demonstrate that reverse signals evoked by CD137L regulate immune functions in macrophages.
Collapse
Affiliation(s)
- Dong-Ku Kim
- Graduate School of Life Science and Biotechnology, Pochon CHA University, CHA Stem Cell Institute, Seoul, Korea
| | | | | |
Collapse
|
32
|
Kraft AD, McPherson CA, Harry GJ. Heterogeneity of microglia and TNF signaling as determinants for neuronal death or survival. Neurotoxicology 2009; 30:785-93. [PMID: 19596372 PMCID: PMC3329780 DOI: 10.1016/j.neuro.2009.07.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 07/01/2009] [Indexed: 12/30/2022]
Abstract
Microglia do not constitute a single, uniform cell population, but rather comprise cells with varied phenotypes, some which are beneficial and others that may require active regulatory control. Thus, gaining a better understanding of the heterogeneity of resident microglia responses will contribute to any interpretation regarding the impact of any such response in the brain. Microglia are the primary source of the pro-inflammatory cytokine, tumor necrosis factor (TNF) that can initiate various effects through the activation of membrane receptors. The TNF p55 receptor contains a death domain and activation normally leads to cellular apoptosis; however, under specific conditions, receptor activation can also lead to the activation of NF-kappaB and contribute to cell survival. These divergent outcomes have been linked to receptor localization with receptor internalization leading to cell death and membrane localization supporting cell survival. A second TNF receptor, TNF p75 receptor, is normally linked to cell growth and survival, however, it can cooperate with the p55 receptor and contribute to cell death. Thus, while an elevation in TNFalpha in the brain is often considered an indicator of microglia activation and neuroinflammation, a number of factors come into play to determine the final outcome. Data are reviewed demonstrating that heterogeneity in morphological response of microglia and the expression of TNFalpha and TNF receptors are critical in identifying and characterizing neurotoxic events as they relate to neuroinflammation, neuronal damage and in stimulating neuroprotection.
Collapse
Affiliation(s)
- Andrew D. Kraft
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| | - Christopher A McPherson
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| | - G. Jean Harry
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| |
Collapse
|
33
|
Badiola N, Malagelada C, Llecha N, Hidalgo J, Comella JX, Sabriá J, Rodríguez-Alvarez J. Activation of caspase-8 by tumour necrosis factor receptor 1 is necessary for caspase-3 activation and apoptosis in oxygen-glucose deprived cultured cortical cells. Neurobiol Dis 2009; 35:438-47. [PMID: 19555759 DOI: 10.1016/j.nbd.2009.06.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 05/22/2009] [Accepted: 06/16/2009] [Indexed: 01/08/2023] Open
Abstract
TNF-alpha has been reported to be relevant in stroke-induced neuronal death. However the precise function of TNF-alpha in brain ischemia remains controversial since there are data supporting either a detrimental or a protective effect. Here we show that TNF-alpha is released after oxygen-glucose deprivation (OGD) of cortical cultures and is a major contributor to the apoptotic death observed without affecting the OGD-mediated necrotic cell death. In this paradigm, apoptosis depends on TNF-alpha-induced activation of caspase-8 and -3 without affecting the activation of caspase-9. By using knock-out mice for TNF-alpha receptor 1, we show that the activation of both caspase-3 and -8 by TNF-alpha is mediated by TNF-alpha receptor 1. The pro-apoptotic role of TNF-alpha in OGD is restricted to neurons and microglia, since astrocytes do not express either TNF-alpha or TNF-alpha receptor 1. Altogether, these results show that apoptosis of cortical neurons after OGD is mediated by TNF-alpha/TNF-alpha receptor 1.
Collapse
Affiliation(s)
- Nahuai Badiola
- Institut de Neurociencies, Departament de Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
34
|
Gaultier A, Wu X, Le Moan N, Takimoto S, Mukandala G, Akassoglou K, Campana WM, Gonias SL. Low-density lipoprotein receptor-related protein 1 is an essential receptor for myelin phagocytosis. J Cell Sci 2009; 122:1155-62. [PMID: 19299462 DOI: 10.1242/jcs.040717] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease in which myelin is progressively degraded. Because degraded myelin may both initiate and accelerate disease progression, clearing degraded myelin from extracellular spaces may be critical. In this study, we prepared myelin vesicles (MV) from rat brains as a model of degraded myelin. Murine embryonic fibroblasts (MEFs) rapidly internalized MVs, which accumulated in lysosomes only when these cells expressed low-density lipoprotein receptor-related protein (LRP1). Receptor-associated protein (RAP), which binds LRP1 and inhibits interaction with other ligands, blocked MV uptake by LRP1-expressing MEFs. As a complementary approach, we prepared primary cultures of rat astrocytes, microglia and oligodendrocytes. All three cell types expressed LRP1 and mediated MV uptake, which was inhibited by RAP. LRP1 gene-silencing in oligodendrocytes also blocked MV uptake. Myelin basic protein (MBP), which was expressed as a recombinant protein, bound directly to LRP1. MBP-specific antibody inhibited MV uptake by oligodendrocytes. In experimental autoimmune encephalomyelitis in mice, LRP1 protein expression was substantially increased in the cerebellum and spinal cord. LRP1 colocalized with multiple CNS cell types. These studies establish LRP1 as a major receptor for phagocytosis of degraded myelin, which may function alone or in concert with co-receptors previously implicated in myelin phagocytosis.
Collapse
Affiliation(s)
- Alban Gaultier
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Girard S, Kadhim H, Roy M, Lavoie K, Brochu ME, Larouche A, Sébire G. Role of perinatal inflammation in cerebral palsy. Pediatr Neurol 2009; 40:168-74. [PMID: 19218029 DOI: 10.1016/j.pediatrneurol.2008.09.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 08/07/2008] [Accepted: 09/17/2008] [Indexed: 01/19/2023]
Abstract
Inflammatory molecules are promptly upregulated in the fetal environment and postnatally in brain-damaged subjects. Intrauterine infections and inflammation are often associated with asphyxia. This double-hit effect by combined infection or inflammation and hypoxia is therefore a frequent concomitant in neonatal brain damage. Animal models combining hypoxia and infection were recently designed to explore the mechanisms underlying brain damage in such circumstances and to look for possible neuroprotective strategies. Proinflammatory cytokines are thought to be major mediators in brain injury in neonates with perinatal asphyxia, bacterial infection, or both. Cytokines, however, could also have neuroprotective properties. The critical point in the balance between neurodamaging and neuroprotective effects of cytokines has yet to be unraveled. This understanding might help to develop new therapeutic approaches to counteract the inflammatory disequilibrium observed in the pathophysiologic mechanisms associated with brain injury.
Collapse
Affiliation(s)
- Sylvie Girard
- Laboratory of Neuropediatrics-Laboratoire de Neuropédiatrie, Neurosciences Centre, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
36
|
Chadwick W, Magnus T, Martin B, Keselman A, Mattson MP, Maudsley S. Targeting TNF-alpha receptors for neurotherapeutics. Trends Neurosci 2008; 31:504-11. [PMID: 18774186 PMCID: PMC2574933 DOI: 10.1016/j.tins.2008.07.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 06/30/2008] [Accepted: 07/30/2008] [Indexed: 12/19/2022]
Affiliation(s)
- Wayne Chadwick
- Receptor Pharmacology Unit, Laboratory of Neurosciences, National Institute on Aging, Biomedical Research Center, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
37
|
Pan W, Yu C, Hsuchou H, Zhang Y, Kastin AJ. Neuroinflammation facilitates LIF entry into brain: role of TNF. Am J Physiol Cell Physiol 2008; 294:C1436-42. [PMID: 18385284 PMCID: PMC2740796 DOI: 10.1152/ajpcell.00489.2007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Leukemia inhibitory factor (LIF) is a proinflammatory cytokine mediating a variety of central nervous system (CNS) responses to inflammatory stimuli. During lipopolysaccharide (LPS)-induced inflammation, blood concentrations of LIF increase, correlating with lethality of sepsis. Circulating LIF crosses the blood-brain barrier (BBB) by a saturable transport system. Here we determine how this transport system is regulated in neuroinflammation. Using transport assays that quantify the influx rate and volume of distribution of LIF in mice, we show that LPS facilitated the permeation of LIF from the blood to the brain without compromising the paracellular permeability of the BBB as determined by coadministration of fluorescein. Concurrently, gp130 (shared by the interleukin-6 family of cytokines), but not gp190 (the specific receptor for LIF) or cilliary neutrophic factor (CNTF-Ralpha, a unique receptor for cilliary neurotrophic factor that also uses gp130 and gp190), showed increased levels of mRNA and protein expression in cerebral microvessels from the LPS-treated mice. The upregulation of gp130 by LPS was at least partially mediated by vascular tumor necrosis factor receptor (TNFR)1 and TNFR2. This was shown by elevated TNFR1 and TNFR2 mRNA and protein in cerebral microvessels after LPS and by the absence of the LPS effect on gp130 in knockout mice lacking these receptors. The results show that neuroinflammation by LPS induces endothelial signaling and enhances cytokine transport across the BBB.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/metabolism
- Capillary Permeability
- Ciliary Neurotrophic Factor Receptor alpha Subunit/genetics
- Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism
- Cytokine Receptor gp130/genetics
- Cytokine Receptor gp130/metabolism
- Disease Models, Animal
- Fluorescein/metabolism
- Fluorescent Dyes/metabolism
- Leukemia Inhibitory Factor/blood
- Leukemia Inhibitory Factor/metabolism
- Lipopolysaccharides
- Male
- Mice
- Mice, Knockout
- Microcirculation/metabolism
- RNA, Messenger/metabolism
- Receptors, OSM-LIF/genetics
- Receptors, OSM-LIF/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Sepsis/chemically induced
- Sepsis/metabolism
- Time Factors
- Tumor Necrosis Factors/deficiency
- Tumor Necrosis Factors/genetics
- Tumor Necrosis Factors/metabolism
Collapse
Affiliation(s)
- Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | | | | | | | | |
Collapse
|
38
|
Kruglov AA, Kuchmiy A, Grivennikov SI, Tumanov AV, Kuprash DV, Nedospasov SA. Physiological functions of tumor necrosis factor and the consequences of its pathologic overexpression or blockade: Mouse models. Cytokine Growth Factor Rev 2008; 19:231-44. [DOI: 10.1016/j.cytogfr.2008.04.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
39
|
Bielefeldt-Ohmann H, Tolnay AE, Reisenhauer C, Hansen T, Smirnova N, Van Campen H. Transplacental Infection with Non-Cytopathic Bovine Viral Diarrhoea Virus Types 1b and 2: Viral Spread and Molecular Neuropathology. J Comp Pathol 2008; 138:72-85. [DOI: 10.1016/j.jcpa.2007.10.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 10/27/2007] [Indexed: 11/24/2022]
|
40
|
Sarnico I, Boroni F, Benarese M, Alghisi M, Valerio A, Battistin L, Spano P, Pizzi M. Targeting IKK2 by pharmacological inhibitor AS602868 prevents excitotoxic injury to neurons and oligodendrocytes. J Neural Transm (Vienna) 2008; 115:693-701. [PMID: 18197358 DOI: 10.1007/s00702-007-0016-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 12/17/2007] [Indexed: 01/01/2023]
Abstract
Among the diverse mechanisms involved in the pathophysiology of post-ischemic and post-traumatic injuries, excitotoxicity and nuclear factor-kappaB (NF-kappaB) activation through induction of IkappaB kinase (IKK) complex have a primary role. We investigated the effects of the selective inhibitor of the IKK2 subunit, the anilinopyrimidine derivative AS602868, on excitotoxic injury produced in rat organotypic hippocampal slices and cerebellar primary neurons. Brief exposure to N-methyl-D-aspartate (NMDA) induces astrocyte reactivity, neuron cell death and oligodendrocyte degeneration in hippocampal slices. Application of AS602868 elicited a long-lasting protection of both neurons and oligodendrocytes. Maximal effect was observed with prolonged application of the compound after NMDA exposure. Neuroprotection was also evident in primary cultures of cerebellar granule cells starting from 20 nM concentration. AS602868-elicited neuroprotection correlated with inhibition of NF-kappaB activity. Our results suggest that AS602868 may prove to be a valuable approach in treating neurodegeneration and demyelination associated with cerebral trauma and ischemia.
Collapse
Affiliation(s)
- I Sarnico
- Division of Pharmacology and Experimental Therapeutics, Department of Biomedical Sciences and Biotechnologies, School of Medicine, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Taoufik E, Tseveleki V, Euagelidou M, Emmanouil M, Voulgari-Kokota A, Haralambous S, Probert L. Positive and Negative Implications of Tumor Necrosis Factor Neutralization for the Pathogenesis of Multiple Sclerosis. NEURODEGENER DIS 2007; 5:32-7. [DOI: 10.1159/000109936] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Accepted: 01/16/2007] [Indexed: 11/19/2022] Open
|
42
|
Pan W, Kastin AJ. Tumor necrosis factor and stroke: role of the blood-brain barrier. Prog Neurobiol 2007; 83:363-74. [PMID: 17913328 PMCID: PMC2190541 DOI: 10.1016/j.pneurobio.2007.07.008] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2007] [Revised: 04/30/2007] [Accepted: 07/26/2007] [Indexed: 01/22/2023]
Abstract
The progression and outcome of stroke is affected by the intricate relationship between the blood-brain barrier (BBB) and tumor necrosis factor alpha (TNFalpha). TNFalpha crosses the intact BBB by a receptor-mediated transport system that is upregulated by CNS trauma and inflammation. In this review, we discuss intracellular trafficking and transcytosis of TNFalpha, regulation of TNFalpha transport after stroke, and the effects of TNFalpha on stroke preconditioning. TNFalpha can activate cytoprotective pathways by pretreatment or persistent exposure to low doses. This explains the paradoxical observation that transport of this proinflammatory cytokine improves the survival and function of hypoxic cells and of mice with stroke. The dual effects of TNFalpha may be related to differential regulation of TNFalpha trafficking downstream to TNFR1 and TNFR2 receptors. As we better understand how peripheral TNFalpha affects its own transport and modulates neuroregeneration, we may be in a better position to pharmacologically manipulate its regulatory transport system to treat stroke.
Collapse
Affiliation(s)
- Weihong Pan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, United States.
| | | |
Collapse
|
43
|
Kaur C, Dheen ST, Ling EA. From blood to brain: amoeboid microglial cell, a nascent macrophage and its functions in developing brain. Acta Pharmacol Sin 2007; 28:1087-96. [PMID: 17640468 DOI: 10.1111/j.1745-7254.2007.00625.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Amoeboid microglial cells (AMC) in the developing brain are active macrophages. The macrophagic nature of these cells has been demonstrated by many methods, such as the localization of various hydrolytic enzymes and the presence of complement type 3 surface receptors in them. More importantly is the direct visualization of these cells engaged in the phagocytosis of degenerating cells at the ultrastructural level. Further evidence of them being active macrophages is the avid internalization of tracers administered by the intravenous or intraperitoneal routes in developing rats. The potential involvement of AMC in immune functions is supported by the induced expression of major histocompatibility complex class I and II antigens on them when challenged by lipopolysaccharide or interferon-gamma. Immunosuppressive drugs, such as glucocorticoids and immune function-enhancing drugs like melatonin, affect the expression of surface receptors and antigens and the release of cytokines by AMC. Recent studies in our laboratory have shown the expression of insulin-like growth factors, endothelins, 2',3'-cyclic nucleotide 3'-phosphodiesterase, and N-methyl-D-asparate receptors. This along with the release of chemokines, such as stromal derived factor-1a and monocyte chemoattractant protein-1, suggests multiple functional roles of AMC in early brain development.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | | | |
Collapse
|
44
|
McCandless EE, Klein RS. Molecular targets for disrupting leukocyte trafficking during multiple sclerosis. Expert Rev Mol Med 2007; 9:1-19. [PMID: 17637110 DOI: 10.1017/s1462399407000397] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AbstractAutoimmune diseases of the central nervous system (CNS) involve the migration of abnormal numbers of self-directed leukocytes across the blood–brain barrier that normally separates the CNS from the immune system. The cardinal lesion associated with neuroinflammatory diseases is the perivascular infiltrate, which comprises leukocytes that have traversed the endothelium and have congregated in a subendothelial space between the endothelial-cell basement membrane and the glial limitans. The exit of mononuclear cells from this space can be beneficial, as when virus-specific lymphocytes enter the CNS for pathogen clearance, or might induce CNS damage, such as in the autoimmune disease multiple sclerosis when myelin-specific lymphocytes invade and induce demyelinating lesions. The molecular mechanisms involved in the movement of lymphocytes through these compartments involve multiple signalling pathways between these cells and the microvasculature. In this review, we discuss adhesion, costimulatory, cytokine, chemokine and signalling molecules involved in the dialogue between lymphocytes and endothelial cells that leads to inflammatory infiltrates within the CNS, and the targeting of these molecules as therapies for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Erin E McCandless
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
45
|
Sriram K, O'Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol 2007; 2:140-53. [PMID: 18040839 DOI: 10.1007/s11481-007-9070-6] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 03/01/2007] [Indexed: 01/05/2023]
Abstract
Proinflammatory cytokines and chemokines have been implicated in the pathogenesis of several neurological and neurodegenerative disorders. Prominent among such factors is the pleiotropic cytokine, tumor necrosis factor (TNF)-alpha. Under normal physiological conditions, TNF-alpha orchestrates a diverse array of functions involved in immune surveillance and defense, cellular homeostasis, and protection against certain neurological insults. However, paradoxical effects of this cytokine have been observed. TNF-alpha is elicited in the brain following injury (ischemia, trauma), infection (HIV, meningitis), neurodegeneration (Alzheimer's, Parkinson's), and chemically induced neurotoxicity. The multifarious identity for this cytokine appears to be influenced by several mechanisms. Among the most prominent are the regulation of TNFalpha-induced NF-kappaB activation by adapter proteins such as TRADD and TRAF, and second, the heterogeneity of microglia and their distribution pattern across brain regions. Here, we review the differential role of TNF-alpha in response to brain injury, with emphasis on neurodegeneration, and discuss the possible mechanisms for such diverse and region-specific effects.
Collapse
Affiliation(s)
- Krishnan Sriram
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, CDC-NIOSH, 1095 Willowdale Road, Morgantown, WV 26505, USA
| | | |
Collapse
|
46
|
Lambertsen KL, Clausen BH, Fenger C, Wulf H, Owens T, Dagnaes-Hansen F, Meldgaard M, Finsen B. Microglia and macrophages express tumor necrosis factor receptor p75 following middle cerebral artery occlusion in mice. Neuroscience 2007; 144:934-49. [PMID: 17161916 DOI: 10.1016/j.neuroscience.2006.10.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Revised: 10/24/2006] [Accepted: 10/26/2006] [Indexed: 01/01/2023]
Abstract
The proinflammatory and potential neurotoxic cytokine tumor necrosis factor (TNF) is produced by activated CNS resident microglia and infiltrating blood-borne macrophages in infarct and peri-infarct areas following induction of focal cerebral ischemia. Here, we investigated the expression of the TNF receptors, TNF-p55R and TNF-p75R, from 1 to 10 days following permanent occlusion of the middle cerebral artery in mice. Using quantitative polymerase chain reaction (PCR), we observed that the relative level of TNF-p55R mRNA was significantly increased at 1-2 days and TNF-p75R mRNA was significantly increased at 1-10 days following arterial occlusion, reaching peak values at 5 days, when microglial-macrophage CD11b mRNA expression was also increased. In comparison, the relative level of TNF mRNA was significantly increased from 1 to 5 days, with peak levels 1 day after arterial occlusion. In situ hybridization revealed mRNA expression of both receptors in predominantly microglial- and macrophage-like cells in the peri-infarct and subsequently in the infarct, and being most marked from 1 to 5 days. Using green fluorescent protein-bone marrow chimeric mice, we confirmed that TNF-p75R was expressed in resident microglia and blood-borne macrophages located in the peri-infarct and infarct 1 and 5 days after arterial occlusion, which was supported by Western blotting. The data show that increased expression of the TNF-p75 receptor following induction of focal cerebral ischemia in mice can be attributed to expression in activated microglial cells and blood-borne macrophages.
Collapse
Affiliation(s)
- K L Lambertsen
- Medical Biotechnology Center, Winsloewparken 25, University of Southern Denmark, Odense, DK-5000, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Fan YH, Lu B, Wang M, Ni GB, Chen MT, Xu Y. Expression and significance of nerve growth factor receptor p75 in rats' cathartic colonic wall. ACTA ACUST UNITED AC 2007; 7:225-9. [PMID: 17054585 DOI: 10.1111/j.1443-9573.2006.00274.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate the expression of nerve growth factor receptor p75 in a normal and cathartic colon and its significance in the formation of the cathartic colon in rats. METHODS Sixty Sprague-Dawley rats were equally divided into normal control group, rhubarb group and phenolphthalein group. A model of the cathartic colon was constructed by gastric infusion with rhubarb or phenolphthalein in rats. The first dose of rhubarb and phenolphthalein was both 200 mg/kg/d and was increased by 200 mg/kg/d with each passing day. The last dose of rhubarb and phenolphthalein was 3200 mg/kg/d and 4200 mg/kg/d, respectively. The transit function of colon was measured by the Chinese ink expulsion test; the p75 in colon wall was determined by the immunohistochemical method. RESULTS The transit speed was much slower in the cathartic colon group than that in the control group. The imprinted Chinese ink length and the ratio of imprinted length/total colon length in the rhubarb-induced cathartic colon was significantly shorter than that of the control group (77.38 +/- 8.42 vs 94.25 +/- 7.07 cm, P < 0.01). Those in the phenolphthalein-induced group (83.38 +/- 9.75 cm) were also significantly shorter than those of the control group but to a lesser degree (P < 0.05). p75 was abundantly expressed in the submucosal nerve plexus and weakly expressed in the myenteric plexus. The expression of p75 was much higher in the rhubarb-induced group. The expression was strongly positive in the submucosal nerve plexus, significantly higher than that in the controls (P < 0.01). In the myenteric plexus, p75 was also highly expressed (P < 0.05). In the case of the phenolphthalein-induced group, the expression of p75 was positive in the submucosal nerve plexus but was positive in the myenteric plexus of three rats only. The remaining rats were negative or weakly positive. This was not significantly different from that of the control group. CONCLUSIONS The abnormal expression of p75 in cathartic colon probably has some effect on the degeneration or apoptosis of neuronal cells in the enteric nerve plexus, with a subsequent pathological change of the enteric nervous system, and thus leads to abnormalities in colonic dynamics. The kind of lesion is probably associated with long-term use of irritative cathartics.
Collapse
Affiliation(s)
- Yi Hong Fan
- Department of Gastroenterology, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang Province, China.
| | | | | | | | | | | |
Collapse
|
48
|
Sébire G, Larouche A, Girard S, Roy M, Kadhim H. Effets pervers des cytokines sur le développement cérébral périnatal ? Med Sci (Paris) 2006; 22:1015-6. [PMID: 17156714 DOI: 10.1051/medsci/200622121015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
49
|
Konsman JP, Drukarch B, Van Dam AM. (Peri)vascular production and action of pro-inflammatory cytokines in brain pathology. Clin Sci (Lond) 2006; 112:1-25. [PMID: 17132137 DOI: 10.1042/cs20060043] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In response to tissue injury or infection, the peripheral tissue macrophage induces an inflammatory response through the release of IL-1β (interleukin-1β) and TNFα (tumour necrosis factor α). These cytokines stimulate macrophages and endothelial cells to express chemokines and adhesion molecules that attract leucocytes into the peripheral site of injury or infection. The aims of the present review are to (i) discuss the relevance of brain (peri)vascular cells and compartments to bacterial meningitis, HIV-1-associated dementia, multiple sclerosis, ischaemic and traumatic brain injury, and Alzheimer's disease, and (ii) to provide an overview of the production and action of pro-inflammatory cytokines by (peri)vascular cells in these pathologies of the CNS (central nervous system). The brain (peri)vascular compartments are highly relevant to pathologies affecting the CNS, as infections are almost exclusively blood-borne. Insults disrupt blood and energy flow to neurons, and active brain-to-blood transport mechanisms, which are the bottleneck in the clearance of unwanted molecules from the brain. Perivascular macrophages are the most reactive cell type and produce IL-1β and TNFα after infection or injury to the CNS. The main cellular target for IL-1β and TNFα produced in the brain (peri)vascular compartment is the endothelium, where these cytokines induce the expression of adhesion molecules and promote leucocyte infiltration. Whether this and other effects of IL-1 and TNF in the brain (peri)vascular compartments are detrimental or beneficial in neuropathology remains to be shown and requires a clear understanding of the role of these cytokines in both damaging and repair processes in the CNS.
Collapse
Affiliation(s)
- Jan P Konsman
- Laboratory of Integrative Neurobiology, CNRS FRE 2723/INRA UR 1244/University Bordeaux2, Institut François Magendie, Bordeaux, France
| | | | | |
Collapse
|
50
|
Plumb J, McQuaid S, Cross AK, Surr J, Haddock G, Bunning RAD, Woodroofe MN. Upregulation of ADAM-17 expression in active lesions in multiple sclerosis. Mult Scler 2006; 12:375-85. [PMID: 16900751 DOI: 10.1191/135248506ms1276oa] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
ADAM-17, a disintegrin and metalloproteinase, is the major proteinase responsible for the cleavage of membrane-bound tumour necrosis factor (TNF) as well as being an active sheddase of other cytokines, cytokine receptors, growth factors and adhesion molecules. TNF is a major proinflammatory cytokine that has been identified as having a pathogenic role in inflammatory diseases within the CNS including multiple sclerosis (MS). Here we report the cellular origin and distribution of ADAM-17 expression within clinically and neuropathologically confirmed MS and normal control white matter, assessed by immunohistochemistry, western blotting and PCR. ADAM-17 expression was associated with the blood vessel endothelium, activated macrophages/microglia and parenchymal astrocytes in MS white matter. Increased levels of ADAM-17 immunoreactivity were displayed in active lesions with evidence of recent myelin breakdown. Further studies into the functional role of ADAM-17 in the pathogenesis of MS and other inflammatory conditions are required.
Collapse
Affiliation(s)
- J Plumb
- Biomedical Research Centre, Sheffield Hallam University, Howard St, Sheffield S1 1WB, UK.
| | | | | | | | | | | | | |
Collapse
|