1
|
Álvarez-López AI, Álvarez-Sánchez N, Cruz-Chamorro I, Santos-Sánchez G, Ponce-España E, Bejarano I, Lardone PJ, Carrillo-Vico A. Melatonin synergistically potentiates the effect of methylprednisolone on reducing neuroinflammation in the experimental autoimmune encephalomyelitis mouse model of multiple sclerosis. J Autoimmun 2024; 148:103298. [PMID: 39067314 DOI: 10.1016/j.jaut.2024.103298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune neurodegenerative disease of unknown etiology characterized by infiltration of encephalitogenic cells in the central nervous system (CNS) resulting in the presence of multifocal areas of demyelination leading to neurodegeneration. The infiltrated immune cells population is composed mainly of effector CD4+ and CD8+ T lymphocytes, B cells, macrophages, and dendritic cells that secrete pro-inflammatory factors that eventually damage myelin leading to axonal damage. The most common clinical form of MS is relapsing-remitting (RR), characterized by neuroinflammatory episodes followed by partial or total recovery of neurological deficits. The first-line treatment for RRMS relapses is a high dose of glucocorticoids, especially methylprednisolone, for three to five consecutive days. Several studies have reported the beneficial effects of melatonin in the context of neuroinflammation associated with MS or experimental autoimmune encephalomyelitis (EAE), the preclinical model for MS. Therefore, the objective of this study was to evaluate the effect of the combined treatment of melatonin and methylprednisolone on the neuroinflammatory response associated with the EAE development. This study shows for the first time the protective synergistic effect of co-treatment with melatonin and methylprednisolone on reducing the severity of EAE by decreasing CD4 lymphocytes, B cells, macrophages and dendritic cells in the CNS, as well as modulating the population of infiltrated T and B cells toward regulatory phenotypes to the detriment of pro-inflammatory effector functions. In addition to the potentiation of the protective role of methylprednisolone, treatment with melatonin from the clinical onset of EAE improves the natural course of the EAE and the response to a subsequent treatment with methylprednisolone in a later relapse of the disease, pointing melatonin as potential therapeutic tool in combination with methylprednisolone for the treatment of relapses in MS.
Collapse
Affiliation(s)
- Ana Isabel Álvarez-López
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, 41009, Spain
| | - Nuria Álvarez-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain
| | - Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, 41009, Spain
| | - Guillermo Santos-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, 41009, Spain
| | - Eduardo Ponce-España
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, 41009, Spain
| | - Ignacio Bejarano
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, 41009, Spain
| | - Patricia Judith Lardone
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, 41009, Spain.
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, 41009, Spain.
| |
Collapse
|
2
|
Greeck VB, Williams SK, Haas J, Wildemann B, Fairless R. Alterations in Lymphocytic Metabolism-An Emerging Hallmark of MS Pathophysiology? Int J Mol Sci 2023; 24:ijms24032094. [PMID: 36768415 PMCID: PMC9917089 DOI: 10.3390/ijms24032094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterised by acute inflammation and subsequent neuro-axonal degeneration resulting in progressive neurological impairment. Aberrant immune system activation in the periphery and subsequent lymphocyte migration to the CNS contribute to the pathophysiology. Recent research has identified metabolic dysfunction as an additional feature of MS. It is already well known that energy deficiency in neurons caused by impaired mitochondrial oxidative phosphorylation results in ionic imbalances that trigger degenerative pathways contributing to white and grey matter atrophy. However, metabolic dysfunction in MS appears to be more widespread than the CNS. This review focuses on recent research assessing the metabolism and mitochondrial function in peripheral immune cells of MS patients and lymphocytes isolated from murine models of MS. Emerging evidence suggests that pharmacological modulation of lymphocytic metabolism may regulate their subtype differentiation and rebalance pro- and anti-inflammatory functions. As such, further understanding of MS immunometabolism may aid the identification of novel treatments to specifically target proinflammatory immune responses.
Collapse
Affiliation(s)
- Viktoria B. Greeck
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sarah K. Williams
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jürgen Haas
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Brigitte Wildemann
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
3
|
Oud L, Garza J. Association of multiple sclerosis with mortality in sepsis: a population-level analysis. J Intensive Care 2022; 10:36. [PMID: 35879778 PMCID: PMC9310428 DOI: 10.1186/s40560-022-00628-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/17/2022] [Indexed: 11/21/2022] Open
Abstract
Background Multiple sclerosis (MS) is associated with increased risk of sepsis and higher sepsis-related mortality, compared to the general population. However, the evidence on the prognostic impact of MS in sepsis has been scarce. We aimed to evaluate the population-level association of MS with short-term mortality in sepsis. Methods We performed a retrospective population-based cohort study using a statewide data set to identify hospitalizations aged ≥ 18 years in Texas with sepsis, with and without MS during 2010–2017. Multilevel logistic models were fit to estimate the association of MS with short-term mortality among all sepsis hospitalizations, and for sensitivity analyses among hospitalizations with septic shock and those admitted to ICU. Results Among 283,025 sepsis hospitalizations, 1687 (0.6%) had MS. Compared to sepsis hospitalizations without MS, those with MS were younger (aged ≥ 65 years 35.0% vs 56.8%), less commonly racial/ethnic minority (36.2% vs 48.1%), and had lower mean Deyo comorbidity index (1.6 vs 2.7). The rates of septic shock and ICU admission were similar for sepsis hospitalizations with and without MS (58.7% vs 59.6% and 46.7% vs 46.0%, respectively). The unadjusted short-term mortality among sepsis hospitalizations with and without MS for the whole cohort, among those with septic shock, and among ICU admissions were 20.2% vs 31.3%, 25.6% vs 40.0%, and 24.0% vs 34.8%, respectively. On adjusted analyses, MS was associated with 17% lower odds of short-term mortality (adjusted odds ratio [aOR] 0.828 [95% CI 0.723–0.947]). Similar findings were observed on sensitivity analyses of patients with septic shock (aOR 0.764 [95% CI 0.651–0.896]), but MS was not associated with mortality among sepsis hospitalizations admitted to ICU (aOR 0.914 [95% CI 0.759–1.101]). Conclusions MS was associated with lower short-term mortality among septic patients, with findings consistent among the subset with septic shock. Among septic patients admitted to ICU, MS was not associated with mortality. Supplementary Information The online version contains supplementary material available at 10.1186/s40560-022-00628-1.
Collapse
|
4
|
Borziak K, Finkelstein J. X-linked genetic risk factors that promote autoimmunity and dampen remyelination are associated with multiple sclerosis susceptibility. Mult Scler Relat Disord 2022; 66:104065. [PMID: 35905688 DOI: 10.1016/j.msard.2022.104065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/14/2022] [Accepted: 07/17/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic neurodegenerative disease, which has a strong genetic component and is more prevalent in women. MS is caused by an autoimmunity initiated inflammatory response which leads to axon demyelination, followed by axon loss, plaque formation and neurodegeneration. The goal of this article was to explore X-linked genetic factors that are associated with MS susceptibility. METHODS Using UK Biobank microarray, we analyzed the prevalence of alleles on the X chromosome to identify variants potentially involved in MS. Overall, 488,225 patients across 18,857 markers were analyzed using PLINK. RESULTS Our results identify 20 SNPs that are significantly more abundant in persons with MS. The genes associated with these SNPs belong to immunity (LAMP2, AVPR2, MTMR8, F8, BCOR, PORCN, and ELF4) and remyelination (NSDHL, HS6ST2, RBM10, TAZ, and AR) pathways that are potentially of great significance for understanding the onset and progression of multiple sclerosis. We further identified a significant 20-fold increase in incidence of MS cases in women with co-occurrences of SNPs associated with myelination and immunity functions. CONCLUSIONS Our analysis provides novel insights into the roles of X-linked genes in the onset and presentation of multiple sclerosis, identifying 20 SNPs in 14 genes involved primarily in immunity and myelination functions that are significantly more abundant in persons with MS. Our co-occurrence analysis suggests that concurrent disruption of both myelination and immune systems significantly increases the risk of MS onset in women.
Collapse
Affiliation(s)
- Kirill Borziak
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 United States.
| | - Joseph Finkelstein
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 United States
| |
Collapse
|
5
|
Serin M, Kara P. Biosensing strategies (approaches) for diagnosis and monitoring of multiple sclerosis. Talanta 2022; 252:123794. [DOI: 10.1016/j.talanta.2022.123794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022]
|
6
|
Ansari MA, Nadeem A, Attia SM, Bakheet SA, Shahid M, Rehman MU, Alanazi MM, Alhamed AS, Ibrahim KE, Albekairi NA, Ahmad SF. CCR1 antagonist J-113863 corrects the imbalance of pro- and anti-inflammatory cytokines in a SJL/J mouse model of relapsing-remitting multiple sclerosis. Immunobiology 2022; 227:152245. [PMID: 35868215 DOI: 10.1016/j.imbio.2022.152245] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 06/20/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022]
Abstract
Multiple sclerosis (MS), an immune-mediated and neurodegenerative disorder of the central nervous system (CNS), is characterized by infiltrating myelin-reactive T lymphocytes and demyelinating lesions. Experimental autoimmune encephalomyelitis (EAE) is a well-established animal model used to study MS. To explore the impact of chemokine receptor CCR1 blockade in EAE and the underlying mechanisms, we used CCR1 antagonist J-113863 in PLP139-151-induced EAE in SJL/J mice. Following EAE induction, mice were treated with J-113863 (10 mg/kg) daily from day 14 until day 25. We investigated the effect of J-113863 on expression levels of GM-CSF, IL-6, IL-10, IL-27 in CD4+ spleen cells, using flow cytometry. We also analyzed the effect of J-113863 on GM-CSF, IL-6, IL-10, IL-27 mRNA and protein expression levels using RT-PCR and Western blot analysis in brain tissues. J-113863 treatment decreased the populations of CD4+GM-CSF+ and CD4+IL-6+ cells and increased CD4+IL-27+ and CD4+IL-10+ cells in the spleen. J-113863 had a suppressive effect on the mRNA and protein expression levels of GM-CSF, and IL-6 in the brain tissue. On the other hand, J-113863 treatment increased the mRNA and protein expression of IL-10 and IL-27 in the brain tissue. Our results highlighted J-113863's potential role in suppressing pro-inflammatory expression and up-regulating anti-inflammatory mediators, which could represent a beneficial alternative approach to MS treatment.
Collapse
Affiliation(s)
- Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Norah A Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
7
|
Fujiwara M, Raheja R, Garo LP, Ajay AK, Kadowaki-Saga R, Karandikar SH, Gabriely G, Krishnan R, Beynon V, Paul A, Patel A, Saxena S, Hu D, Healy BC, Chitnis T, Gandhi R, Weiner HL, Murugaiyan G. microRNA-92a promotes CNS autoimmunity by modulating the regulatory and inflammatory T cell balance. J Clin Invest 2022; 132:e155693. [PMID: 35298438 PMCID: PMC9106347 DOI: 10.1172/jci155693] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/16/2022] [Indexed: 01/11/2023] Open
Abstract
A disequilibrium between immunosuppressive Tregs and inflammatory IL-17-producing Th17 cells is a hallmark of autoimmune diseases, including multiple sclerosis (MS). However, the molecular mechanisms underlying the Treg and Th17 imbalance in CNS autoimmunity remain largely unclear. Identifying the factors that drive this imbalance is of high clinical interest. Here, we report a major disease-promoting role for microRNA-92a (miR-92a) in CNS autoimmunity. miR-92a was elevated in experimental autoimmune encephalomyelitis (EAE), and its loss attenuated EAE. Mechanistically, miR-92a mediated EAE susceptibility in a T cell-intrinsic manner by restricting Treg induction and suppressive capacity, while supporting Th17 responses, by directly repressing the transcription factor Foxo1. Although miR-92a did not directly alter Th1 differentiation, it appeared to indirectly promote Th1 cells by inhibiting Treg responses. Correspondingly, miR-92a inhibitor therapy ameliorated EAE by concomitantly boosting Treg responses and dampening inflammatory T cell responses. Analogous to our findings in mice, miR-92a was elevated in CD4+ T cells from patients with MS, and miR-92a silencing in patients' T cells promoted Treg development but limited Th17 differentiation. Together, our results demonstrate that miR-92a drives CNS autoimmunity by sustaining the Treg/Th17 imbalance and implicate miR-92a as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Mai Fujiwara
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Radhika Raheja
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lucien P. Garo
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Amrendra K. Ajay
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ryoko Kadowaki-Saga
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sukrut H. Karandikar
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Galina Gabriely
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rajesh Krishnan
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Vanessa Beynon
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anu Paul
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Amee Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shrishti Saxena
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dan Hu
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brian C. Healy
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tanuja Chitnis
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Roopali Gandhi
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gopal Murugaiyan
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Jiang Z, Zhu H, Wang P, Que W, Zhong L, Li X, Du F. Different subpopulations of regulatory T cells in human autoimmune disease, transplantation, and tumor immunity. MedComm (Beijing) 2022; 3:e137. [PMID: 35474948 PMCID: PMC9023873 DOI: 10.1002/mco2.137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/11/2022] Open
Abstract
CD4+CD25+ regulatory T cells (Tregs), a subpopulation of naturally CD4+ T cells that characteristically express transcription factor Forkhead box P3 (FOXP3), play a pivotal role in the maintenance of immune homeostasis and the prevention of autoimmunity. With the development of biological technology, the understanding of plasticity and stability of Tregs has been further developed. Recent studies have suggested that human Tregs are functionally and phenotypically diverse. The functions and mechanisms of different phenotypes of Tregs in different disease settings, such as tumor microenvironment, autoimmune diseases, and transplantation, have gradually become hot spots of immunology research that arouse extensive attention. Among the complex functions, CD4+CD25+FOXP3+ Tregs possess a potent immunosuppressive capacity and can produce various cytokines, such as IL‐2, IL‐10, and TGF‐β, to regulate immune homeostasis. They can alleviate the progression of diseases by resisting inflammatory immune responses, whereas promoting the poor prognosis of diseases by helping cells evade immune surveillance or suppressing effector T cells activity. Therefore, methods for targeting Tregs to regulate their functions in the immune microenvironment, such as depleting them to strengthen tumor immunity or expanding them to treat immunological diseases, need to be developed. Here, we discuss that different subpopulations of Tregs are essential for the development of immunotherapeutic strategies involving Tregs in human diseases.
Collapse
Affiliation(s)
- Zhongyi Jiang
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Haitao Zhu
- Department of Hepatobiliary Surgery The Affiliated Hospital of Guizhou Medical University Guizhou P. R. China
| | - Pusen Wang
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Weitao Que
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Lin Zhong
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Xiao‐Kang Li
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
- Division of Transplantation Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Futian Du
- Department of Hepatobiliary Surgery Weifang People's Hospital Shandong P. R. China
| |
Collapse
|
9
|
Mayrhofer F, Dariychuk Z, Zhen A, Daugherty DJ, Bannerman P, Hanson AM, Pleasure D, Soulika A, Deng W, Chechneva OV. Reduction in CD11c + microglia correlates with clinical progression in chronic experimental autoimmune demyelination. Neurobiol Dis 2021; 161:105556. [PMID: 34752925 DOI: 10.1016/j.nbd.2021.105556] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 12/25/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease with high variability of clinical symptoms. In most cases MS appears as a relapsing-remitting disease course that at a later stage transitions into irreversible progressive decline of neurologic function. The mechanisms underlying MS progression remain poorly understood. Experimental autoimmune encephalomyelitis (EAE) is an animal model of MS. Here we demonstrate that mice that develop mild EAE after immunization with myelin oligodendrocyte glycoprotein 35-55 are prone to undergo clinical progression around 30 days after EAE induction. EAE progression was associated with reduction in CD11c+ microglia and dispersed coalescent parenchymal infiltration. We found sex-dependent differences mediated by p38α signaling, a key regulator of inflammation. Selective reduction of CD11c+ microglia in female mice with CD11c-promoter driven p38α knockout correlated with increased rate of EAE progression. In protected animals, we found CD11c+ microglia forming contacts with astrocyte processes at the glia limitans and immune cells retained within perivascular spaces. Together, our study identified pathological hallmarks of chronic EAE progression and suggests that CD11c+ microglia may regulate immune cell parenchymal infiltration in autoimmune demyelination.
Collapse
Affiliation(s)
- Florian Mayrhofer
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Zhanna Dariychuk
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Anthony Zhen
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Daniel J Daugherty
- Department of Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, United States of America; IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Peter Bannerman
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Angela M Hanson
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - David Pleasure
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Athena Soulika
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, United States of America; IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Olga V Chechneva
- Department of Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, United States of America; IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America.
| |
Collapse
|
10
|
Circulating miRNAs as Potential Biomarkers Distinguishing Relapsing-Remitting from Secondary Progressive Multiple Sclerosis. A Review. Int J Mol Sci 2021; 22:ijms222111887. [PMID: 34769314 PMCID: PMC8584709 DOI: 10.3390/ijms222111887] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/30/2021] [Accepted: 10/31/2021] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating neurodegenerative, highly heterogeneous disease with a variable course. The most common MS subtype is relapsing–remitting (RR), having interchanging periods of worsening and relative stabilization. After a decade, in most RR patients, it alters into the secondary progressive (SP) phase, the most debilitating one with no clear remissions, leading to progressive disability deterioration. Among the greatest challenges for clinicians is understanding disease progression molecular mechanisms, since RR is mainly characterized by inflammatory processes, while in SP, the neurodegeneration prevails. This is especially important because distinguishing RR from the SP subtype early will enable faster implementation of appropriate treatment. Currently, the MS course is not well-correlated with the biomarkers routinely used in clinical practice. Despite many studies, there are still no reliable indicators correlating with the disease stage and its activity degree. Circulating microRNAs (miRNAs) may be considered valuable molecules for the MS diagnosis and, presumably, helpful in predicting disease subtype. MiRNA expression dysregulation is commonly observed in the MS course. Moreover, knowledge of diverse miRNA panel expression between RRMS and SPMS may allow for deterring disability progression through successful treatment. Therefore, in this review, we address the current state of research on differences in miRNA panel expression between the phases.
Collapse
|
11
|
Fadul CE, Mao-Draayer Y, Ryan KA, Noelle RJ, Wishart HA, Channon JY, Kasper IR, Oliver B, Mielcarz DW, Kasper LH. Safety and Immune Effects of Blocking CD40 Ligand in Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e1096. [PMID: 34654708 PMCID: PMC8527364 DOI: 10.1212/nxi.0000000000001096] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/11/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVES Costimulation by CD40 and its ligand CD40L (CD154) is important for the functional differentiation of T cells. Preclinical studies have recognized the importance of this costimulatory interaction in the pathogenesis of experimental models of multiple sclerosis (MS). To determine safety, pharmacokinetics, and immune effect of a humanized monoclonal antibody (mAb) against CD40 ligand (toralizumab/IDEC-131) in patients with relapsing-remitting MS (RRMS). METHODS This single-institution open-label dose-escalation study (phase I) enrolled 12 patients with RRMS to receive 4 doses of 1, 5, 10, or 15 mg/kg of humanized αCD40L (toralizumab) IV infusion every other week. Patients were followed up to 18 weeks, annually, and finally at 5 years. In addition to safety and pharmacokinetics, other secondary and exploratory measurements are immune effects, clinical, MRI, laboratory, and neuropsychological evaluations. RESULTS Fifteen adverse events, all of mild to moderate severity, were considered to be of possible or of unknown relationship to treatment. No serious adverse events, including thromboembolic events, occurred during the 18-week defined study period. Annual and long-term follow-up at 5 years revealed no delayed toxicity. Pharmacokinetics were nonlinear between the 5 and 10 mg/kg dose groups. The serum half-life of toralizumab was consistent between the dose groups with a mean of 15.3 days (SD = 1.9). Flow cytometry revealed no depletion of lymphocyte subsets. An increase in the CD25+/CD3+ and CD25+/CD4+ ratio and a shift toward an anti-inflammatory cytokine response were seen after treatment. DISCUSSION Our study suggests that blocking CD40L is safe and well tolerated in patients with RRMS while increasing CD25 + T cells and anti-inflammatory cytokine profile. These findings support further studies to assess the efficacy of blocking CD40L as a potential treatment of RRMS. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence on the safety, pharmacokinetics, and immune effects of an mAb to CD40L in patients with RRMS.
Collapse
MESH Headings
- Adult
- Antibodies, Blocking/administration & dosage
- Antibodies, Blocking/adverse effects
- Antibodies, Blocking/pharmacology
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- CD40 Ligand
- Female
- Follow-Up Studies
- Humans
- Immunologic Factors/administration & dosage
- Immunologic Factors/adverse effects
- Immunologic Factors/pharmacokinetics
- Immunologic Factors/pharmacology
- Male
- Middle Aged
- Multiple Sclerosis, Relapsing-Remitting/drug therapy
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Outcome Assessment, Health Care
Collapse
Affiliation(s)
| | | | - Kathleen A. Ryan
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| | - Randolph J. Noelle
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| | - Heather A. Wishart
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| | - Jacqueline Y. Channon
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| | - Isaac R. Kasper
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| | - Brant Oliver
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| | - Daniel W. Mielcarz
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| | - Lloyd H. Kasper
- From the Department of Neurology (C.E.F.), University of Virginia School of Medicine, Charlottesville; Department of Medicine, Microbiology/Immunology and Psychiatry (K.A.R., R.J.N., H.A.W., J.Y.C., J.R.K., B.O., D.W.M., L.H.K.), Dartmouth Medical School, Lebanon, NH; and Department of Neurology (Y.M.-D.), Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor
| |
Collapse
|
12
|
Guerrero JM, Aguirre FS, Mota ML, Carrillo A. Advances for the Development of In Vitro Immunosensors for Multiple Sclerosis Diagnosis. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00018-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Ullrich KAM, Schulze LL, Paap EM, Müller TM, Neurath MF, Zundler S. Immunology of IL-12: An update on functional activities and implications for disease. EXCLI JOURNAL 2020; 19:1563-1589. [PMID: 33408595 PMCID: PMC7783470 DOI: 10.17179/excli2020-3104] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022]
Abstract
As its first identified member, Interleukin-12 (IL-12) named a whole family of cytokines. In response to pathogens, the heterodimeric protein, consisting of the two subunits p35 and p40, is secreted by phagocytic cells. Binding of IL-12 to the IL-12 receptor (IL-12R) on T and natural killer (NK) cells leads to signaling via signal transducer and activator of transcription 4 (STAT4) and subsequent interferon gamma (IFN-γ) production and secretion. Signaling downstream of IFN-γ includes activation of T-box transcription factor TBX21 (Tbet) and induces pro-inflammatory functions of T helper 1 (TH1) cells, thereby linking innate and adaptive immune responses. Initial views on the role of IL-12 and clinical efforts to translate them into therapeutic approaches had to be re-interpreted following the discovery of other members of the IL-12 family, such as IL-23, sharing a subunit with IL-12. However, the importance of IL-12 with regard to immune processes in the context of infection and (auto-) inflammation is still beyond doubt. In this review, we will provide an update on functional activities of IL-12 and their implications for disease. We will begin with a summary on structure and function of the cytokine itself as well as its receptor and outline the signal transduction and the transcriptional regulation of IL-12 secretion. In the second part of the review, we will depict the involvement of IL-12 in immune-mediated diseases and relevant experimental disease models, while also providing an outlook on potential translational approaches.
Collapse
Affiliation(s)
- Karen A.-M. Ullrich
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Lisa Lou Schulze
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Eva-Maria Paap
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Tanja M. Müller
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Markus F. Neurath
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Sebastian Zundler
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| |
Collapse
|
14
|
Renavikar PS, Sinha S, Brate AA, Borcherding N, Crawford MP, Steward-Tharp SM, Karandikar NJ. IL-12-Induced Immune Suppressive Deficit During CD8+ T-Cell Differentiation. Front Immunol 2020; 11:568630. [PMID: 33193343 PMCID: PMC7657266 DOI: 10.3389/fimmu.2020.568630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/02/2020] [Indexed: 11/13/2022] Open
Abstract
Autoimmune diseases are characterized by regulatory deficit in both the CD4+ and CD8+ T-cell compartments. We have shown that CD8+ T-cells associated with acute relapse of multiple sclerosis are significantly deficient in their immune suppressive ability. We hypothesized that distinct CD8+ cytotoxic T-cell (Tc) lineages, determined by cytokine milieu during naïve T-cell differentiation, may harbor differential ability to suppress effector CD4+ T-cells. We differentiated purified human naïve CD8+ T-cells in vitro toward Tc0 (media control), Tc1 and Tc17 lineages. Using in vitro flow cytometric suppression assays, we observed that Tc0 and Tc17 cells had similar suppressive ability. In contrast, Tc1 cells showed significant loss of suppressive ability against ex vivo CD4+ T-cells and in vitro-differentiated Th0, Th1 and Th17 cells. Of note, Tc1 cells were also suboptimal in suppressing CD4-induced acute xenogeneic graft versus host disease (xGVHD) in vivo. Tc subtypes derived under various cytokine combinations revealed that IL-12-containing conditions resulted in less suppressive cells exhibiting dysregulated cytotoxic degranulation. RNA sequencing transcriptome analyses indicated differential regulation of inflammatory genes and enrichment in GM-CSF-associated pathways. These studies provide insights into the role of T-cell differentiation in CD8 suppressive biology and may reveal therapeutically targetable pathways to reverse suppressive deficit during immune-mediated disease.
Collapse
Affiliation(s)
- Pranav S Renavikar
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Sushmita Sinha
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Ashley A Brate
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Nicholas Borcherding
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Michael P Crawford
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Scott M Steward-Tharp
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Nitin J Karandikar
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| |
Collapse
|
15
|
Nali LH, Olival GS, Sousa FTG, de Oliveira ACS, Montenegro H, da Silva IT, Dias-Neto E, Naya H, Spangenberg L, Penalva-de-Oliveira AC, Romano CM. Whole transcriptome analysis of multiple Sclerosis patients reveals active inflammatory profile in relapsing patients and downregulation of neurological repair pathways in secondary progressive cases. Mult Scler Relat Disord 2020; 44:102243. [PMID: 32559700 DOI: 10.1016/j.msard.2020.102243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/21/2020] [Accepted: 05/25/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is an inflammatory autoimmune neurologic disease that causes progressive destruction of myelin sheath and axons. Affecting more than 2 million people worldwide, MS may presents distinct clinical courses. However, information regarding key gene expression and genic pathways related to each clinical form is still limited. OBJECTIVE To assess the whole transcriptome of blood leukocytes from patients with remittent-recurrent (RRMS) and secondary-progressive (SPMS) forms to explore the gene expression profile of each form. METHODS Total RNA was obtained and sequenced in Illumina HiSeq platform. Reads were aligned to human genome (GRCh38/hg38), BAM files were mapped and differential expression was obtained with DeSeq2. Up or downregulated pathways were obtained through Ingenuity IPA. Pro-inflammatory cytokines levels were also assessed. RESULTS The transcriptome was generated for nine patients (6 SPMS and 3 RRMS) and 5 healthy controls. A total of 731 and 435 differentially expressed genes were identified in SPMS and RRMS, respectively. RERE, IRS2, SIPA1L1, TANC2 and PLAGL1 were upregulated in both forms, whereas PAD2 and PAD4 were upregulated in RRMS and downregulated in SPMS. Inflammatory and neuronal repair pathways were upregulated in RRMS, which was also observed in cytokine analysis. Conversely, SPMS patients presented IL-8, IL-1, Neurothrophin and Neuregulin pathways down regulated. CONCLUSIONS Overall, the transcriptome of RRMS and SPMS clearly indicated distinct inflammatory profiles, where RRMS presented marked pro-inflammatory profile but SPMS did not. SPMS individuals also presented a decrease on expression of neuronal repair pathways.
Collapse
Affiliation(s)
- Luiz H Nali
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, LIM-52 (LIMHC) Universidade de São Paulo, Rua Dr. Enéas de Carvalho Aguiar, 470, São Paulo, 05403-000, Brazil.; Post-graduation Program in Health Sciences, Santo Amaro University, Rua Prof. Enéas de Siqueira Neto, 340, São Paulo, 04829-300, Brazil
| | - Guilherme S Olival
- Departamento de Neurologia Santa Casa de Misericórdia de São Paulo, R. Dr. Cesário Mota Júnior, 112, São Paulo, 01221-020 Brazil
| | - Francielle T G Sousa
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, LIM-52 (LIMHC) Universidade de São Paulo, Rua Dr. Enéas de Carvalho Aguiar, 470, São Paulo, 05403-000, Brazil
| | - Ana Carolina S de Oliveira
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, LIM-52 (LIMHC) Universidade de São Paulo, Rua Dr. Enéas de Carvalho Aguiar, 470, São Paulo, 05403-000, Brazil
| | | | - Israel T da Silva
- Laboratory of Medical Genomics, A.C.Camargo Cancer Center, São Paulo, 01525-001, Brazil
| | - Emamnuel Dias-Neto
- Laboratory of Medical Genomics, A.C.Camargo Cancer Center, São Paulo, 01525-001, Brazil; Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, São Paulo Medical School, University of São Paulo, São Paulo, Brazil
| | - Hugo Naya
- Unidad de Bioinformática Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Lucia Spangenberg
- Unidad de Bioinformática Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Augusto C Penalva-de-Oliveira
- Departamento de Neurologia Santa Casa de Misericórdia de São Paulo, R. Dr. Cesário Mota Júnior, 112, São Paulo, 01221-020 Brazil; Departamento de Neurologia, Instituto de Infectologia Emilio Ribas, Avenida Doutor Arnaldo, 165, São Paulo, 01246-900, Brazil
| | - Camila M Romano
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, LIM-52 (LIMHC) Universidade de São Paulo, Rua Dr. Enéas de Carvalho Aguiar, 470, São Paulo, 05403-000, Brazil.; Hospital das Clinicas HCFMUSP (LIM52), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
16
|
Glik A, Mazar J, Rogachev B, Zlotnik M, Douvdevani A. CD40 Ligand Expression Correlates with Resolution of Peritonitis and Mononuclear Cell Recruitment. Perit Dial Int 2020. [DOI: 10.1177/089686080502500307] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background CD40 belongs to the tumor necrosis factor receptor family and its ligation is a central event in major inflammatory and immune reactions. We have previously demonstrated that CD40 ligation upregulates the secretion of mononuclear chemokines from peritoneal mesothelial cells (PMC), and that blocking the CD40 ligand (CD154) reduced the mononuclear infiltrate in a model of peritonitis. Objective To characterize the kinetics of CD154 expression on peritoneal leukocytes and examine the correlation of this occurrence with the mononuclear transition at the resolution phase of peritonitis. Methods Leukocytes were collected from the effluent of 11 patients during episodes of peritonitis while undergoing peritoneal dialysis (PD). The effluent was then analyzed by flow cytometry to characterize CD154 expression. Results CD154 expression on peritoneal mononuclear cells gradually increased during the resolution phase of peritonitis, peaking first on T cells (CD4+ and CD8+ cells at 20 – 45 hours) and then on macrophages (CD14+ at 20 – 50 hours). The maximal expression of CD154 on macrophages, CD4+ cells, and CD8+ cells during peak hours reached values of 33% ± 23%, 4% ± 3%, and 24% ± 17%, respectively. The increase in CD154 expression was in negative correlation ( r=–0.44, p = 0.032) with total leukocyte numbers and in positive correlation ( r = 0.52, p = 0.009) with the increase of mononuclear cells. Deterioration of peritonitis was associated with a decrease in CD154 levels, while recurrence of peritonitis was related to high CD154 levels. Conclusion Our data, which show a positive correlation between CD154 levels and mononuclear dominance, suggest that CD40–CD154 ligation plays an important role in the transition to mononuclear predominance in the late phase of peritonitis.
Collapse
Affiliation(s)
- Amir Glik
- Department of Nephrology, Soroka Medical Center, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer Sheva, Israel
| | - Julia Mazar
- Department of Nephrology, Soroka Medical Center, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer Sheva, Israel
| | - Boris Rogachev
- Department of Nephrology, Soroka Medical Center, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer Sheva, Israel
| | - Moshe Zlotnik
- Department of Nephrology, Soroka Medical Center, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer Sheva, Israel
| | - Amos Douvdevani
- Department of Nephrology, Soroka Medical Center, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer Sheva, Israel
| |
Collapse
|
17
|
Toghi M, Bitarafan S, Kasmaei HD, Ghafouri-Fard S. Bifidobacteria: A probable missing puzzle piece in the pathogenesis of multiple sclerosis. Mult Scler Relat Disord 2019; 36:101378. [PMID: 31487552 DOI: 10.1016/j.msard.2019.101378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/29/2019] [Accepted: 08/27/2019] [Indexed: 12/27/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder in which the immunopathogenesis is not fully understood. In the recent years, the role of gut microbiome in the pathogenesis of this disorder has been highlighted. Bifidobacteria as a component of gut microbiome might also be involved in MS pathogenesis. Being emerged in early days after birth, bifidobacteria have a prominent role in immune system maturation and function. Some factors like mode of delivery, breast feeding, mother's blood group and her secretory state and also environmental factors could influence its level in the early infancy, which may remain throughout lifetime. In this review, we discussed possible immunopathogenic link between the bifidobacteria and MS.
Collapse
Affiliation(s)
- Mehdi Toghi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Bitarafan
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hosein Delavar Kasmaei
- Department of Neurology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Manoochehrabadi S, Arsang-Jang S, Mazdeh M, Inoko H, Sayad A, Taheri M. Analysis of STAT1, STAT2 and STAT3 mRNA expression levels in the blood of patients with multiple sclerosis. Hum Antibodies 2019; 27:91-98. [PMID: 30412483 DOI: 10.3233/hab-180352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is the most common chronic, inflammatory, autoimmune disease of the central nervous system (CNS) maintained by the secretion of a large number of cytokines [1]. The signal transducer and activator of transcription (STAT) family has an essential role in transmitting many of the cytokine-mediated signals and failure in the signaling process contributes to the etiopathogenesis of MS. METHODS This study aimed to assess STAT1, STAT2 and STAT3 gene expression in the blood of 50 relapsing-remitting MS (RR-MS) patients and 50 healthy controls by TaqMan Quantitative Real-Time PCR. RESULTS The results showed that STAT1 gene expression was significantly up-regulated (p= 0.023), whereas STAT2 gene expression was significantly down-regulated (p< 0.0001) in MS patients compared to controls. On the other hand, there was no significant difference between MS patients and controls for STAT3 gene expression (p= 0.837). In addition, there was no significant correlation between the expression of STAT1, STAT2, STAT3 genes and clinical findings, such as the level of physical disability in MS patients (according to the Kurtzke Expanded Disability Status Scale (EDSS) criterion) and disease duration. CONCLUSION A significant positive correlation was demonstrated between STAT1 and STAT2 and also between STAT1 and STAT3. This study shows for the first time that a comparison of the relative quantitative expression of three different STAT genes in the blood cells of MS patients compared to controls revealed marked differences in the expression of the STAT family genes that might reflect their different roles in the pathogenesis of MS. These transcripts might be useful biomarkers for evaluating the efficacy of IFN treatment of the MS patients.
Collapse
Affiliation(s)
- Saba Manoochehrabadi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Arsang-Jang
- Clinical Research Development Center, Qom University of Medical Sciences, Qom, Iran
| | - Mehrdokht Mazdeh
- Neurophysiology Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neurology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hidetoshi Inoko
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Japan.,Genodive Pharma Inc., Atsugi, Japan
| | - Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Karim H, Kim SH, Lauderdale K, Lapato AS, Atkinson K, Yasui N, Yamate-Morgan H, Sekyi M, Katzenellenbogen JA, Tiwari-Woodruff SK. Analogues of ERβ ligand chloroindazole exert immunomodulatory and remyelinating effects in a mouse model of multiple sclerosis. Sci Rep 2019; 9:503. [PMID: 30679747 PMCID: PMC6345788 DOI: 10.1038/s41598-018-37420-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/27/2018] [Indexed: 01/06/2023] Open
Abstract
Pharmaceutical agents currently approved for the treatment of multiple sclerosis reduce relapse rates, but do not reverse or prevent neurodegeneration nor initiate myelin repair. The highly selective estrogen receptor (ER) β ligand chloroindazole (IndCl) shows particular promise promoting both remyelination while reducing inflammatory cytokines in the central nervous system of mice with experimental autoimmune encephalomyelitis. To optimize these benefits, we developed and screened seven novel IndCl analogues for their efficacy in promoting primary oligodendrocyte (OL) progenitor cell survival, proliferation, and differentiation in vitro by immunohistochemistry. Two analogues, IndCl-o-chloro and IndCl-o-methyl, induced proliferation and differentiation equivalent to IndCl and were selected for subsequent in vivo evaluation for their impact on clinical disease course, white matter pathology, and inflammation. Both compounds ameliorated disease severity, increased mature OLs, and improved overall myelination in the corpus callosum and white matter tracts of the spinal cord. These effects were accompanied by reduced production of the OL toxic molecules interferon-γ and chemokine (C-X-C motif) ligand, CXCL10 by splenocytes with no discernable effect on central nervous system-infiltrating leukocyte numbers, while IndCl-o-methyl also reduced peripheral interleukin (IL)−17. In addition, expression of the chemokine CXCL1, which is associated with developmental oligodendrogenesis, was upregulated by IndCl and both analogues. Furthermore, callosal compound action potential recordings from analogue-treated mice demonstrated a larger N1 component amplitude compared to vehicle, suggesting more functionally myelinated fibers. Thus, the o-Methyl and o-Chloro IndCl analogues represent a class of ERβ ligands that offer significant remyelination and neuroprotection as well as modulation of the immune system; hence, they appear appropriate to consider further for therapeutic development in multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Hawra Karim
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Kelli Lauderdale
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Andrew S Lapato
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Kelley Atkinson
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Norio Yasui
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hana Yamate-Morgan
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Maria Sekyi
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | | | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA. .,Center for Glia Neuronal Interaction, UCR School of Medicine, Riverside, CA, 92521, USA.
| |
Collapse
|
20
|
Co-signaling Molecules in Neurological Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:233-265. [PMID: 31758537 DOI: 10.1007/978-981-32-9717-3_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
Piancone F, Saresella M, Marventano I, La Rosa F, Santangelo MA, Caputo D, Mendozzi L, Rovaris M, Clerici M. Monosodium Urate Crystals Activate the Inflammasome in Primary Progressive Multiple Sclerosis. Front Immunol 2018; 9:983. [PMID: 29780394 PMCID: PMC5945820 DOI: 10.3389/fimmu.2018.00983] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/20/2018] [Indexed: 01/04/2023] Open
Abstract
Inflammasome-driven inflammation is postulated to play a role in multiple sclerosis (MS), but there is no direct evidence that the nod-like receptor protein 3 (NLRP3) inflammasome is involved in MS pathogenesis. Uric acid was shown to be one of the “danger” signals involved in the activation of NLRP3 inflammasome; notably, the concentration of uric acid is increased in the serum and in the cerebrospinal fluid of MS individuals. To better investigate the role of the NLRP3 inflammasome in MS-associated inflammation, we primed with lipopolysaccharide and stimulated with monosodium urate crystals PBMCs of 41 MS patients with different disease phenotypes. Eleven individuals with primary progressive MS (PPMS), 10 individuals with stable relapsing–remitting MS (SMS), 10 individuals with acute relapsing–remitting MS (AMS), 10 individuals with benign MS were analyzed; 10 healthy controls were enrolled as well in the study. The expression of the NLRP3, apoptosis-associated speck-like protein containing CARD (ASC), caspase-1, caspase-8, IL-1β, and IL-18 inflammasome genes was evaluated by RT-PCR. NLRP3 and ASC-speck protein expression was analyzed by FlowSight AMNIS, whereas production of the pro-inflammatory cytokines IL-1β and IL-18 and of caspase-1 and caspase-8 was measured by ELISA in supernatants. Results showed that uric acid serum concentration was significantly increased in PPMS; in these and in AMS patients, mRNA for NLRP3, ASC, and IL-18 was upregulated as well, but caspase-8 mRNA was upregulated only in PPMS. Expression of NLRP3 and ASC-speck protein was significantly increased in PPMS, SMS, and AMS patients, but IL-18 and caspase-8 production was significantly increased only in PPMS, in whom a direct correlation between hyperuricemia and caspase-8 was detected. The NLRP3/caspase-8 inflammasome pathway is activated in PPMS, possibly as a consequence of hyperuricemia. Therapeutic strategies reducing NLRP3 activation and/or lowering hyperuricemia could be useful in the therapy of PPMS.
Collapse
Affiliation(s)
- Federica Piancone
- Laboratory of Molecular Medicine and Biotechnology, Don Gnocchi Foundation, IRCCS, Milan, Italy
| | - Marina Saresella
- Laboratory of Molecular Medicine and Biotechnology, Don Gnocchi Foundation, IRCCS, Milan, Italy
| | - Ivana Marventano
- Laboratory of Molecular Medicine and Biotechnology, Don Gnocchi Foundation, IRCCS, Milan, Italy
| | - Francesca La Rosa
- Laboratory of Molecular Medicine and Biotechnology, Don Gnocchi Foundation, IRCCS, Milan, Italy
| | | | - Domenico Caputo
- Department of Neurology, Don Gnocchi Foundation, IRCCS, Milan, Italy
| | - Laura Mendozzi
- Department of Neurology, Don Gnocchi Foundation, IRCCS, Milan, Italy
| | - Marco Rovaris
- Department of Neurology, Don Gnocchi Foundation, IRCCS, Milan, Italy
| | - Mario Clerici
- Laboratory of Molecular Medicine and Biotechnology, Don Gnocchi Foundation, IRCCS, Milan, Italy.,Department of Physiopathology and Transplants, University of Milano, Milan, Italy
| |
Collapse
|
22
|
Baecher-Allan C, Kaskow BJ, Weiner HL. Multiple Sclerosis: Mechanisms and Immunotherapy. Neuron 2018; 97:742-768. [DOI: 10.1016/j.neuron.2018.01.021] [Citation(s) in RCA: 611] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/25/2017] [Accepted: 01/09/2018] [Indexed: 12/17/2022]
|
23
|
Aarts SABM, Seijkens TTP, van Dorst KJF, Dijkstra CD, Kooij G, Lutgens E. The CD40-CD40L Dyad in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Front Immunol 2017; 8:1791. [PMID: 29312317 PMCID: PMC5732943 DOI: 10.3389/fimmu.2017.01791] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022] Open
Abstract
The CD40-CD40L dyad is an immune checkpoint regulator that promotes both innate and adaptive immune responses and has therefore an essential role in the development of inflammatory diseases, including multiple sclerosis (MS). In MS, CD40 and CD40L are expressed on immune cells present in blood and lymphoid organs, affected resident central nervous system (CNS) cells, and inflammatory cells that have infiltrated the CNS. CD40-CD40L interactions fuel the inflammatory response underlying MS, and both genetic deficiency and antibody-mediated inhibition of the CD40-CD40L dyad reduce disease severity in experimental autoimmune encephalomyelitis (EAE). Both proteins are therefore attractive therapeutic candidates to modulate aberrant inflammatory responses in MS. Here, we discuss the genetic, experimental and clinical studies on the role of CD40 and CD40L interactions in EAE and MS and we explore novel approaches to therapeutically target this dyad to combat neuroinflammatory diseases.
Collapse
Affiliation(s)
- Suzanne A. B. M. Aarts
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Tom T. P. Seijkens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany
| | | | - Christine D. Dijkstra
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany
| |
Collapse
|
24
|
Kulu U, Tiftikcioğlu Bİ, Zorlu Y, Çetiner M, Şener U, Tuna G, Kirkali G. Efficacy of Different Durations of Intravenous Methylprednisolone Treatment in Relapses of Multiple Sclerosis. Noro Psikiyatr Ars 2017; 54:57-61. [PMID: 28566960 DOI: 10.5152/npa.2016.12382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 11/18/2015] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Relapses of multiple sclerosis (MS) are usually treated with high-dose intravenous methylprednisolone (IVMP), given over 3-10 days. There is no consensus on the optimal duration of treatment. In this study, we aimed to investigate whether longer treatment provides additional short-term clinical benefits assessed by the change in plasma cytokine levels and EDSS scores in patients with relapsing-remitting MS (RRMS). METHODS Forty RRMS patients during relapse were grouped into 3 and treated with 1 g/day of IVMP for either 5, 7, or 10 consecutive days. RESULTS Levels of IL-10 and IL-12 were analyzed, and EDSS scores were noted before treatment, after treatment (on days 6, 8, or 11) and at the 4th week. IVMP treatment significantly induced anti-inflammatory IL-10 levels but had no effect on IL-12 levels. IVMP treatment for 7 or 10 consecutive days was not significantly different than that for 5 days in terms of the change in IL-12, IL-10 levels or clinical outcome. CONCLUSION In conclusion, pulse high-dose IVMP treatment enhances functional recovery in patients with acute relapses of RRMS. In addition, IVMP treatment significantly increases the levels of IL-10 but has no effect on the levels of IL-12 in the short term.
Collapse
Affiliation(s)
- Uğur Kulu
- Clinic of Neurology, Tepecik Training and Research Hospital, İzmir, Turkey
| | | | - Yaşar Zorlu
- Clinic of Neurology, Tepecik Training and Research Hospital, İzmir, Turkey
| | - Mustafa Çetiner
- Clinic of Neurology, Tepecik Training and Research Hospital, İzmir, Turkey
| | - Ufuk Şener
- Clinic of Neurology, Tepecik Training and Research Hospital, İzmir, Turkey
| | - Gamze Tuna
- Department of Medical Biochemistry, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
| | - Güldal Kirkali
- Department of Medical Biochemistry, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
| |
Collapse
|
25
|
Lu S, Yan Y, Li Z, Chen L, Yang J, Zhang Y, Wang S, Liu L. Determination of Genes Related to Uveitis by Utilization of the Random Walk with Restart Algorithm on a Protein-Protein Interaction Network. Int J Mol Sci 2017; 18:ijms18051045. [PMID: 28505077 PMCID: PMC5454957 DOI: 10.3390/ijms18051045] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/14/2022] Open
Abstract
Uveitis, defined as inflammation of the uveal tract, may cause blindness in both young and middle-aged people. Approximately 10–15% of blindness in the West is caused by uveitis. Therefore, a comprehensive investigation to determine the disease pathogenesis is urgent, as it will thus be possible to design effective treatments. Identification of the disease genes that cause uveitis is an important requirement to achieve this goal. To begin to answer this question, in this study, a computational method was proposed to identify novel uveitis-related genes. This method was executed on a large protein–protein interaction network and employed a popular ranking algorithm, the Random Walk with Restart (RWR) algorithm. To improve the utility of the method, a permutation test and a procedure for selecting core genes were added, which helped to exclude false discoveries and select the most important candidate genes. The five-fold cross-validation was adopted to evaluate the method, yielding the average F1-measure of 0.189. In addition, we compared our method with a classic GBA-based method to further indicate its utility. Based on our method, 56 putative genes were chosen for further assessment. We have determined that several of these genes (e.g., CCL4, Jun, and MMP9) are likely to be important for the pathogenesis of uveitis.
Collapse
Affiliation(s)
- Shiheng Lu
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Yan Yan
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Zhen Li
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China.
| | - Jing Yang
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Yuhang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Shaopeng Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Lin Liu
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
26
|
Aly L, Hemmer B, Korn T. From Leflunomide to Teriflunomide: Drug Development and Immunosuppressive Oral Drugs in the Treatment of Multiple Sclerosis. Curr Neuropharmacol 2017; 15:874-891. [PMID: 27928949 PMCID: PMC5652031 DOI: 10.2174/1570159x14666161208151525] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/03/2016] [Accepted: 05/12/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Immunosuppressive drugs have been used in the treatment of multiple sclerosis (MS) for a long time. Today, orally available second generation immunosuppressive agents have been approved or are filed for licensing as MS therapeutics. Due to semi-selective targeting of cellular processes, these second-generation immunosuppressive compounds might rather be immunomodulatory. For example, Teriflunomide inhibits the de novo pyrimidine synthesis and thus only targets rapidly proliferating cells, including lymphocytes. It is used as first line disease modifying therapy (DMT) in relapsing-remitting MS (RRMS). METHODS Review of online content related to oral immunosuppressants in MS with an emphasis on Teriflunomide. RESULTS Teriflunomide and Cladribine are second-generation immunosuppressants that are efficient in the treatment of MS patients. For Teriflunomide, a daily dose of 14 mg reduces the annualized relapse rate (ARR) by more than 30% and disability progression by 30% compared to placebo. Cladribine reduces the ARR by about 50% compared to placebo but has not yet been licensed due to unresolved safety concerns. We also discuss the significance of older immunosuppressive compounds including Azathioprine, Mycophenolate mofetile, and Cyclophosphamide in current MS therapy. CONCLUSION Teriflunomide has shown a favorable safety and efficacy profile in RRMS and is a therapeutic option for a distinct group of adult patients with RRMS.
Collapse
Affiliation(s)
- Lilian Aly
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany,
- Department of Experimental Neuroimmunology, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany,
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany,
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Korn
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany,
- Department of Experimental Neuroimmunology, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany,
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
27
|
Huber AK, Giles DA, Segal BM, Irani DN. An emerging role for eotaxins in neurodegenerative disease. Clin Immunol 2016; 189:29-33. [PMID: 27664933 DOI: 10.1016/j.clim.2016.09.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/07/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022]
Abstract
Eotaxins are C-C motif chemokines first identified as potent eosinophil chemoattractants. They facilitate eosinophil recruitment to sites of inflammation in response to parasitic infections as well as allergic and autoimmune diseases such as asthma, atopic dermatitis, and inflammatory bowel disease. The eotaxin family currently includes three members: eotaxin-1 (CCL11), eotaxin-2 (CCL24), and eotaxin-3 (CCL26). Despite having only ~30% sequence homology to one another, each was identified based on its ability to bind the chemokine receptor, CCR3. Beyond their role in innate immunity, recent studies have shown that CCL11 and related molecules may directly contribute to degenerative processes in the central nervous system (CNS). CCL11 levels increase in the plasma and cerebrospinal fluid of both mice and humans as part of normal aging. In mice, these increases are associated with declining neurogenesis and impaired cognition and memory. In humans, elevated plasma levels of CCL11 have been observed in Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease, and secondary progressive multiple sclerosis when compared to age-matched, healthy controls. Since CCL11 is capable of crossing the blood-brain barrier of normal mice, it is plausible that eotaxins generated in the periphery may exert physiological and pathological actions in the CNS. Here, we briefly review known functions of eotaxin family members during innate immunity, and then focus on whether and how these molecules might participate in the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Amanda K Huber
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David A Giles
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Benjamin M Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David N Irani
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Leibowitz SM, Yan J. NF-κB Pathways in the Pathogenesis of Multiple Sclerosis and the Therapeutic Implications. Front Mol Neurosci 2016; 9:84. [PMID: 27695399 PMCID: PMC5023675 DOI: 10.3389/fnmol.2016.00084] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways are involved in cell immune responses, apoptosis and infections. In multiple sclerosis (MS), NF-κB pathways are changed, leading to increased levels of NF-κB activation in cells. This may indicate a key role for NF-κB in MS pathogenesis. NF-κB signaling is complex, with many elements involved in its activation and regulation. Interestingly, current MS treatments are found to be directly or indirectly linked to NF-κB pathways and act to adjust the innate and adaptive immune system in patients. In this review, we will first focus on the intricacies of NF-κB signaling, including the activating pathways and regulatory elements. Next, we will theorize about the role of NF-κB in MS pathogenesis, based on current research findings, and discuss some of the associated therapeutic implications. Lastly, we will review four new MS treatments which interrupt NF-κB pathways—fingolimod, teriflunomide, dimethyl fumarate (DMF) and laquinimod (LAQ)—and explain their mechanisms, and the possible strategy for MS treatments in the future.
Collapse
Affiliation(s)
- Saskia M Leibowitz
- UQ Centre for Clinical Research, The University of Queensland Brisbane, QLD, Australia
| | - Jun Yan
- UQ Centre for Clinical Research, The University of Queensland Brisbane, QLD, Australia
| |
Collapse
|
29
|
Latent virus infection upregulates CD40 expression facilitating enhanced autoimmunity in a model of multiple sclerosis. Sci Rep 2015; 5:13995. [PMID: 26356194 PMCID: PMC4564856 DOI: 10.1038/srep13995] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/13/2015] [Indexed: 02/07/2023] Open
Abstract
Epstein-Barr virus (EBV) has been identified as a putative environmental trigger of multiple sclerosis (MS) by multiple groups working worldwide. Previously, we reported that when experimental autoimmune encephalomyelitis (EAE) was induced in mice latently infected with murine γ-herpesvirus 68 (γHV-68), the murine homolog to EBV, a disease more reminiscent of MS developed. Specifically, MS-like lesions developed in the brain that included equal numbers of IFN-γ producing CD4+ and CD8+ T cells and demyelination, none of which is observed in MOG induced EAE. Herein, we demonstrate that this enhanced disease was dependent on the γHV-68 latent life cycle and was associated with STAT1 and CD40 upregulation on uninfected dendritic cells. Importantly, we also show that, during viral latency, the frequency of regulatory T cells is reduced via a CD40 dependent mechanism and this contributes towards a strong T helper 1 response that resolves in severe EAE disease pathology. Latent γ-herpesvirus infection established a long-lasting impact that enhances subsequent adaptive autoimmune responses.
Collapse
|
30
|
Estes ML, McAllister AK. Alterations in immune cells and mediators in the brain: it's not always neuroinflammation! Brain Pathol 2015; 24:623-30. [PMID: 25345893 DOI: 10.1111/bpa.12198] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/02/2023] Open
Abstract
Neuroinflammation was once a clearly defined term denoting pathological immune processes within the central nervous system (CNS). Historically, this term was used to indicate the four hallmarks of peripheral inflammaton that occur following severe CNS injuries, such as stroke, injury or infection. Recently, however, the definition of neuroinflammation has relaxed to the point that it is often now assumed to be present when even only a single classical hallmark of inflammation is measured. As a result, a wide range of disorders, from psychiatric to degenerative diseases, are now assumed to have an integral inflammatory component. Ironically, at the same time, research has revealed unexpected nonclassical immune actions of immune mediators and cells in the CNS in the absence of pathology, increasing the likelihood that homeostatic and adaptive immune processes in the CNS will be mistaken for neuroinflammation. Thus, we suggest reserving the term neuroinflammation for contexts where multiple signs of inflammation are present to avoid erroneously classifying disorders as inflammatory when they may instead be caused by nonimmune etiologies or secondary immune processes that serve adaptive roles.
Collapse
|
31
|
Up-regulation of IL-12 expression in patients with chronic hepatitis B is mediated by the PI3K/Akt pathway. Mol Cell Biochem 2015; 407:135-42. [PMID: 26062743 DOI: 10.1007/s11010-015-2463-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/29/2015] [Indexed: 12/15/2022]
Abstract
Hepatitis B virus (HBV) replicates noncytopathically in hepatocytes, but HBV or proteins encoded by HBV genome could induce cytokines, chemokines expression by hepatocytes.IL-12 is a typical proinflammatory cytokine that plays a critical role in host defense against pathogens, including the HBV. However, the role of IL-12 in chronic hepatitis B (CHB) remains unclear. The aims of this study were to detect the expression of IL-12 in CHB patients and explore the molecular mechanism of HBV-induced IL-12 expression. The results showed that serum levels and hepatic expression of IL-12 were significantly upregulated in CHB patients. HBx protein increased IL-12 expression in a dose-dependent manner. Furthermore, inhibition of PI3K/Akt significantly decreased the HBx-induced IL-12 expression and Akt activation. Taken together, these results indicate that the molecular mechanism of HBV-induced IL-12 expression involves activation of the PI3K/Akt pathway by HBx, leading to transactivation of the IL-12 p35 and p40 promoters.
Collapse
|
32
|
Tejera-Alhambra M, Casrouge A, de Andrés C, Seyfferth A, Ramos-Medina R, Alonso B, Vega J, Fernández-Paredes L, Albert ML, Sánchez-Ramón S. Plasma biomarkers discriminate clinical forms of multiple sclerosis. PLoS One 2015; 10:e0128952. [PMID: 26039252 PMCID: PMC4454618 DOI: 10.1371/journal.pone.0128952] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 04/30/2015] [Indexed: 01/23/2023] Open
Abstract
Multiple sclerosis, the most common cause of neurological disability in young population after trauma, represents a significant public health burden. Current challenges associated with management of multiple sclerosis (MS) patients stem from the lack of biomarkers that might enable stratification of the different clinical forms of MS and thus prompt treatment for those patients with progressive MS, for whom there is currently no therapy available. In the present work we analyzed a set of thirty different plasma cytokines, chemokines and growth factors present in circulation of 129 MS patients with different clinical forms (relapsing remitting, secondary progressive and primary progressive MS) and 53 healthy controls, across two independent cohorts. The set of plasma analytes was quantified with Luminex xMAP technology and their predictive power regarding clinical outcome was evaluated both individually using ROC curves and in combination using logistic regression analysis. Our results from two independent cohorts of MS patients demonstrate that the divergent clinical and histology-based MS forms are associated with distinct profiles of circulating plasma protein biomarkers, with distinct signatures being composed of chemokines and growth/angiogenic factors. With this work, we propose that an evaluation of a set of 4 circulating biomarkers (HGF, Eotaxin/CCL11, EGF and MIP-1β/CCL4) in MS patients might serve as an effective tool in the diagnosis and more personalized therapeutic targeting of MS patients.
Collapse
Affiliation(s)
- Marta Tejera-Alhambra
- Department of Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Armanda Casrouge
- Department of Immunology, Center for Human Immunology, Institut Pasteur, Paris, France
- Department of Immunology, INSERM U818, Institut Pasteur, Paris, France
| | - Clara de Andrés
- Department of Neurology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Rocío Ramos-Medina
- Department of Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Bárbara Alonso
- Department of Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Janet Vega
- Center Alicia Koplowitz for Multiple Sclerosis of the Community of Madrid, Madrid, Spain
| | | | - Matthew L. Albert
- Department of Immunology, Center for Human Immunology, Institut Pasteur, Paris, France
- Department of Immunology, INSERM U818, Institut Pasteur, Paris, France
| | - Silvia Sánchez-Ramón
- Department of Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- * E-mail:
| |
Collapse
|
33
|
Interleukin 12 (IL-12) family cytokines: Role in immune pathogenesis and treatment of CNS autoimmune disease. Cytokine 2015; 75:249-55. [PMID: 25796985 DOI: 10.1016/j.cyto.2015.01.030] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/23/2015] [Accepted: 01/23/2015] [Indexed: 12/18/2022]
Abstract
Cytokines play crucial roles in coordinating the activities of innate and adaptive immune systems. In response to pathogen recognition, innate immune cells secrete cytokines that inform the adaptive immune system about the nature of the pathogen and instruct naïve T cells to differentiate into the appropriate T cell subtypes required to clear the infection. These include Interleukins, Interferons and other immune-regulatory cytokines that exhibit remarkable functional redundancy and pleiotropic effects. The focus of this review, however, is on the enigmatic Interleukin 12 (IL-12) family of cytokines. This family of cytokines plays crucial roles in shaping immune responses during antigen presentation and influence cell-fate decisions of differentiating naïve T cells. They also play essential roles in regulating functions of a variety of effector cells, making IL-12 family cytokines important therapeutic targets or agents in a number of inflammatory diseases, such as the CNS autoimmune diseases, uveitis and multiple sclerosis.
Collapse
|
34
|
Rumble JM, Huber AK, Krishnamoorthy G, Srinivasan A, Giles DA, Zhang X, Wang L, Segal BM. Neutrophil-related factors as biomarkers in EAE and MS. ACTA ACUST UNITED AC 2015; 212:23-35. [PMID: 25559893 PMCID: PMC4291533 DOI: 10.1084/jem.20141015] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Using a mouse model of multiple sclerosis (MS), the authors show that neutrophils expand in the bone marrow and accumulate in the circulation before clinical onset of disease. Early in disease development, neutrophils infiltrate the CNS, which is suppressed by G-CSF receptor deficiency and blockade of CXCL1 to ameliorate disease. In patients with MS, systemic expression of neutrophil-related mediators correlates with new lesion formation, lesion burden, and clinical disability. A major function of T helper (Th) 17 cells is to induce the production of factors that activate and mobilize neutrophils. Although Th17 cells have been implicated in the pathogenesis of multiple sclerosis (MS) and the animal model experimental autoimmune encephalomyelitis (EAE), little attention has been focused on the role of granulocytes in those disorders. We show that neutrophils, as well as monocytes, expand in the bone marrow and accumulate in the circulation before the clinical onset of EAE, in response to systemic up-regulation of granulocyte colony-stimulating factor (G-CSF) and the ELR+ CXC chemokine CXCL1. Neutrophils comprised a relatively high percentage of leukocytes infiltrating the central nervous system (CNS) early in disease development. G-CSF receptor deficiency and CXCL1 blockade suppressed myeloid cell accumulation in the blood and ameliorated the clinical course of mice that were injected with myelin-reactive Th17 cells. In relapsing MS patients, plasma levels of CXCL5, another ELR+ CXC chemokine, were elevated during acute lesion formation. Systemic expression of CXCL1, CXCL5, and neutrophil elastase correlated with measures of MS lesion burden and clinical disability. Based on these results, we advocate that neutrophil-related molecules be further investigated as novel biomarkers and therapeutic targets in MS.
Collapse
Affiliation(s)
- Julie M Rumble
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Amanda K Huber
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | | | - Ashok Srinivasan
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - David A Giles
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Xu Zhang
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Lu Wang
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin M Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109 Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109 Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| |
Collapse
|
35
|
Kacperska MJ, Jastrzebski K, Tomasik B, Walenczak J, Konarska-Krol M, Glabinski A. Selected extracellular microRNA as potential biomarkers of multiple sclerosis activity--preliminary study. J Mol Neurosci 2014; 56:154-63. [PMID: 25487315 PMCID: PMC4382531 DOI: 10.1007/s12031-014-0476-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/26/2014] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). Four distinct disease courses are known, although approximately 90% of patients are diagnosed with the relapsing-remitting form (RRMS). The name "multiple sclerosis" pertains to the underlying pathology: the presence of demyelinating plaques in the CNS, in particular in the periventricular region, corpus callosum, cervical spine, and the cerebellum. There are ongoing efforts to discover biomarkers that would allow for an unequivocal diagnosis, assess the activity of inflammatory and neurodegenerative processes, or warn of disease progression. At present, small noncoding RNA particles-microRNA (miRNA, miR) seem to be particularly noteworthy, as they take part in posttranscriptional regulation of expression of various genes. Changes in composition as well as function of miRNA found in body fluids of MS patients are subjects of research, in the hope they prove accurate markers of MS activity. This preliminary study aims to evaluate the expression of selected extracellular microRNA particles (miRNA-let-7a, miRNA-92a, miRNA-684a) in patients experiencing MS relapse and remission, with healthy volunteers serving as a control group and to evaluate the correlation between miRNA expression and selected clinical parameters of those patients. Thirty-seven patients suffering from MS formed two examined groups: 20 patients undergoing relapse and 17 in remission. Thirty healthy volunteers formed the control group. All patients who were subjects to peripheral blood sampling had been hospitalized in the Department of Neurology and Stroke(1). Four milliliters of venous whole blood had been collected into EDTA tubes. The basis for the selection of the three particular miRNA investigated in this study (miRNA-let-7a, miRNA-92a, miRNA-684a) was a preliminary bioinformatic analysis of data compiled from several medical databases, including Ovid MEDLINE®, Embase, Cochrane Database of Systematic Reviews (CDSR), miRWalk, and miRBase. The isolation of extracellular microRNA from plasma was carried out using miRNeasy Mini Kit (Qiagen) reagents. The reverse transcription was carried out with TaqMan® MicroRNA Reverse Transcription Kit (Applied Biosystems), as per manufacturers' instructions. Standard microRNA TaqMan® tests (Applied Biosystems) were used for miRNA quantification. The qPCR were performed on a 7900 HT Fast Real-Time PCR System (Applied Biosystems) and analyzed using Sequence Detection System 2.3 software. In addition, all patients at the Department of Neurology and Stroke undergo a routine complete blood count with differential. The main objective of this study was to evaluate the expression of selected microRNA (has-miR-let-7a, miR-92a, and miR-648a) in the plasma of patients with MS during a relapse as well as in remission and attempt to correlate the acquired data with clinically relevant parameters of the disease. Finding such correlations may potentially lead to the use of miRNA as a biomarker of MS, which could help diagnose the disease and assess its severity and the efficacy of treatment. The difference in the expression of has-miR-let-7a in the remission group and the control group was statistically significant (p = 0.002). Similarly, the expression of miRNA-648a in patients in remission was significantly different from the expression in the control group (p = 0.02). Analysis of the correlation between the expression of miRNA-92a and the severity of the disease as measured by the EDSS scale in patients undergoing relapse showed significant negative linear correlation (r = -0.54, p = 0.01). Higher miR-648a expression correlated with more frequent flare-ups in the joint group of patients in remission and relapse (p = 0.03). This study is one of the few that demonstrate significantly changed expression of selected extracellular miRNA in plasma of MS patients and correlate those findings with clinical parameters. These observations may suggest that some miRNA subsets may be potential biomarkers for MS activity.
Collapse
|
36
|
Huber AK, Wang L, Han P, Zhang X, Ekholm S, Srinivasan A, Irani DN, Segal BM. Dysregulation of the IL-23/IL-17 axis and myeloid factors in secondary progressive MS. Neurology 2014; 83:1500-7. [PMID: 25253754 PMCID: PMC4222856 DOI: 10.1212/wnl.0000000000000908] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 07/25/2014] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE In the current exploratory study, we longitudinally measured immune parameters in the blood of individuals with relapsing-remitting multiple sclerosis (RRMS) and secondary progressive multiple sclerosis (SPMS), and investigated their relationship to disease duration and clinical and radiologic measures of CNS injury. METHODS Peripheral blood mononuclear cells (PBMCs) and plasma were obtained from subjects with RRMS, SPMS, and from healthy controls on a monthly basis over the course of 1 year. MRI and Expanded Disability Status Scale evaluations were performed serially. PBMCs were analyzed by enzyme-linked immunosorbent spot assay to enumerate myelin basic protein-specific interleukin (IL)-17- and interferon (IFN)-γ-producing cells. Plasma concentrations of proinflammatory factors were measured using customized Luminex panels. RESULTS Frequencies of myelin basic protein-specific IL-17- and IFN-γ-producing PBMCs were higher in individuals with RRMS and SPMS compared to healthy controls. Patients with SPMS expressed elevated levels of IL-17-inducible chemokines that activate and recruit myeloid cells. In the cohort of patients with SPMS without inflammatory activity, upregulation of myeloid-related factors correlated directly with MRI T2 lesion burden and inversely with brain parenchymal tissue volume. CONCLUSIONS The results of this exploratory study raise the possibility that Th17 responses and IL-17-inducible myeloid factors are elevated during SPMS compared with RRMS, and correlate with lesion burden. Our data endorse further investigation of Th17- and myeloid-related factors as candidate therapeutic targets in SPMS.
Collapse
Affiliation(s)
- Amanda K Huber
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Lu Wang
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Peisong Han
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Xu Zhang
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Sven Ekholm
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Ashok Srinivasan
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - David N Irani
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Benjamin M Segal
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY.
| |
Collapse
|
37
|
Ratzer R, Romme Christensen J, Romme Nielsen B, Sørensen PS, Börnsen L, Sellebjerg F. Immunological effects of methylprednisolone pulse treatment in progressive multiple sclerosis. J Neuroimmunol 2014; 276:195-201. [PMID: 25218212 DOI: 10.1016/j.jneuroim.2014.08.623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/08/2014] [Accepted: 08/21/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To investigate the effect of monthly oral methylprednisolone pulse treatment in progressive MS. METHODS 30 progressive MS patients were treated with oral methylprednisolone every month. Peripheral blood mononuclear cells were analyzed by flow cytometry. RESULTS Out of 102 leukocyte phenotypes investigated, 25 changed at nominal significance from baseline to week 12 (p<0.05). After correction for multiple comparisons, we found 5 subpopulations that changed compared to baseline. No pattern were suggesting modulation of Th17 or TFH cells. CONCLUSION Methylprednisolone pulse treatment has some effects on circulating immune cells but does not modulate markers of Th17 and TFH cell activity in progressive MS.
Collapse
Affiliation(s)
- R Ratzer
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Denmark.
| | - J Romme Christensen
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Denmark
| | - B Romme Nielsen
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Denmark
| | - P S Sørensen
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Denmark
| | - L Börnsen
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Denmark
| | - F Sellebjerg
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Denmark
| |
Collapse
|
38
|
Lim ET, Giovannoni G. Immunopathogenesis and immunotherapeutic approaches in multiple sclerosis. Expert Rev Neurother 2014; 5:379-90. [PMID: 15938671 DOI: 10.1586/14737175.5.3.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis is an organ-specific autoimmune disease, characterized pathologically by cell-mediated inflammation, demyelination and variable degrees of axonal loss. Although inflammation is considered central to the pathogenesis of multiple sclerosis, to date, the only licensed and hence widely used multiple sclerosis immunotherapies are interferon-beta, glatiramer acetate and mitoxantrone. This review discusses the immunopathogenesis of multiple sclerosis, focusing on a number of emerging immunotherapies. A number of new approaches likely to manipulate the immunopathogenesis of multiple sclerosis and which may ultimately allow for the development of more effective immunotherapy are also highlighted.
Collapse
Affiliation(s)
- Ee Tuan Lim
- University College London, Department of Neuroinflammation, Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | | |
Collapse
|
39
|
Okuda DT. Immunosuppressive treatments in multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:503-11. [DOI: 10.1016/b978-0-444-52001-2.00022-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
40
|
Silva GAA, Pradella F, Moraes A, Farias A, dos Santos LMB, de Oliveira ALR. Impact of pregabalin treatment on synaptic plasticity and glial reactivity during the course of experimental autoimmune encephalomyelitis. Brain Behav 2014; 4:925-35. [PMID: 25365796 PMCID: PMC4178248 DOI: 10.1002/brb3.276] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/22/2014] [Accepted: 07/29/2014] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune and neurodegenerative disease that affects young adults. It is characterized by generating a chronic demyelinating autoimmune inflammation in the central nervous system. An experimental model for studying MS is the experimental autoimmune encephalomyelitis (EAE), induced by immunization with antigenic proteins from myelin. AIMS The present study investigated the evolution of EAE in pregabalin treated animals up to the remission phase. METHODS AND RESULTS The results demonstrated a delay in the onset of the disease with statistical differences at the 10th and the 16th day after immunization. Additionally, the walking track test (CatWalk) was used to evaluate different parameters related to motor function. Although no difference between groups was obtained for the foot print pressure, the regularity index was improved post treatment, indicating a better motor coordination. The immunohistochemical analysis of putative synapse preservation and glial reactivity revealed that pregabalin treatment improved the overall morphology of the spinal cord. A preservation of circuits was depicted and the glial reaction was downregulated during the course of the disease. qRT-PCR data did not show immunomodulatory effects of pregabalin, indicating that the positive effects were restricted to the CNS environment. CONCLUSIONS Overall, the present data indicate that pregabalin is efficient for reducing the seriousness of EAE, delaying its course as well as reducing synaptic loss and astroglial reaction.
Collapse
Affiliation(s)
- Gleidy A A Silva
- Laboratory of Nerve Regeneration, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP Campinas, SP, Brazil
| | - Fernando Pradella
- Neuroimmunology Unit, Department of Genetics, Evolution and Bioagents, University of Campinas - UNICAMP Campinas, SP, Brazil ; Neuroimmunomodulation Group, Department of Genetics, Evolution and Bioagents, University of Campinas - UNICAMP Campinas, SP, Brazil
| | - Adriel Moraes
- Neuroimmunology Unit, Department of Genetics, Evolution and Bioagents, University of Campinas - UNICAMP Campinas, SP, Brazil ; Neuroimmunomodulation Group, Department of Genetics, Evolution and Bioagents, University of Campinas - UNICAMP Campinas, SP, Brazil
| | - Alessandro Farias
- Neuroimmunology Unit, Department of Genetics, Evolution and Bioagents, University of Campinas - UNICAMP Campinas, SP, Brazil ; Neuroimmunomodulation Group, Department of Genetics, Evolution and Bioagents, University of Campinas - UNICAMP Campinas, SP, Brazil
| | - Leonilda M B dos Santos
- Neuroimmunology Unit, Department of Genetics, Evolution and Bioagents, University of Campinas - UNICAMP Campinas, SP, Brazil
| | - Alexandre L R de Oliveira
- Laboratory of Nerve Regeneration, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP Campinas, SP, Brazil
| |
Collapse
|
41
|
Gandhi R, Healy B, Gholipour T, Egorova S, Musallam A, Hussain MS, Nejad P, Patel B, Hei H, Khoury S, Quintana F, Kivisakk P, Chitnis T, Weiner HL. Circulating microRNAs as biomarkers for disease staging in multiple sclerosis. Ann Neurol 2013; 73:729-40. [PMID: 23494648 DOI: 10.1002/ana.23880] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 02/19/2013] [Accepted: 03/01/2013] [Indexed: 12/16/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) are single-stranded, small noncoding RNAs that regulate gene expression. Because they are stable in serum, they are being developed as biomarkers for cancer and other diseases. In multiple sclerosis (MS), miRNAs have been studied in cell populations but not in the circulation. In MS, a major challenge is to develop immune biomarkers to monitor disease. We asked whether circulating miRNAs could be identified in MS and whether they are linked to disease stage and/or disability. METHODS A total of 368 miRNAs were measured in ethylenediaminetetraacetic acid plasma in 10 relapsing-remitting MS (RRMS) patients, 9 secondary progressive MS (SPMS) patients, and 9 healthy controls (HCs) using miRCURY LNA™ Universal RT microRNA polymerase chain reaction panels. Nineteen miRNAs from this discovery set were validated using qPCR on an independent set of 50 RRMS patients, 51 SPMS patients, and 32 HCs. RESULTS We found that circulating miRNAs are differentially expressed in RRMS and SPMS versus HCs and in RRMS versus SPMS. We also found miRNAs to be linked to Expanded Disability Status Scale (EDSS). hsa-miR-92a-1* was identified in the largest number of comparisons. It was different in RRMS versus SPMS, and RRMS versus HCs, and showed an association with EDSS and disease duration. miR-92 has target genes involved in cell cycle regulation and cell signaling. The let-7 family of miRNAs differentiated SPMS from HCs and RRMS from SPMS. let-7 miRNAs regulate stem cell differentiation and T cell activation, activate Toll-like receptor 7, and are linked to neurodegeneration. hsa-miR-454 differentiated RRMS from SPMS, and hsa-miR-145 differentiated RRMS from HCs and RRMS from SPMS. Interestingly, the same circulating miRNAs (let-7 and miR-92) that were differentially expressed in RRMS versus SPMS also differentiated amyotrophic lateral sclerosis (ALS) from RRMS subjects, but were not different between SPMS and ALS, suggesting that similar processes may occur in SPMS and ALS. INTERPRETATION Our results establish circulating miRNAs as a readily accessible blood biomarker to monitor disease in MS.
Collapse
Affiliation(s)
- Roopali Gandhi
- Partners Multiple Sclerosis Center, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Levy YA, Fainberg KM, Amidror T, Regev K, Auriel E, Karni A. High and dysregulated secretion of epidermal growth factor from immune cells of patients with relapsing-remitting multiple sclerosis. J Neuroimmunol 2013; 257:82-9. [PMID: 23466131 DOI: 10.1016/j.jneuroim.2013.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 01/19/2013] [Accepted: 01/30/2013] [Indexed: 10/27/2022]
Abstract
We studied the secretion and regulation of epidermal growth factor (EGF) from immune cells of patients with relapsing remitting multiple sclerosis (RR-MS), and the relevance of these levels to neuronal morphology and survival. Our data suggest that the immune-mediated neuronal and oligodendroglial regeneration may be defective by the increased EGF secretion from immune cells of RR-MS patients. We also suggest an increased neurotoxicity of the immune response in RR-MS via high levels of EGF secretion. This is a heretofore unreported aspect of the immune response of patients with RR-MS. Our results may support the inadequate tissue repair that has been observed in MS.
Collapse
Affiliation(s)
- Yifat Amir Levy
- Neuroimmunology Laboratory, Department of Neurology, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
43
|
Soulillou JP. Missing links in multiple sclerosis etiology. A working connecting hypothesis. Med Hypotheses 2013; 80:509-16. [PMID: 23466062 DOI: 10.1016/j.mehy.2013.01.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 01/29/2013] [Indexed: 12/15/2022]
Abstract
The etiology of multiple sclerosis is still elusive despite an extended patchwork of mechanistic events has been accumulated. In this article, are tentatively identified from scattered literature sources new factors that may link well known characteristic of MS such as the central alteration of BBR selectivity, its association with EBV status and its biased distribution of the globe more comprehensively. The hypothesis proposes that the concomitant important rise in some heterophilic antibodies (anti Neu5Gc) which activate BBB endothelial cells and in the frequency of anti EBV committed T cells and of memory B infected cells with EBV contemporary to EBV infection play a major role in MS etiology. In addition, the hypothesis proposes new possible explanations for the elevated risk of MS in specific geographical area.
Collapse
|
44
|
Romme Christensen J, Börnsen L, Ratzer R, Piehl F, Khademi M, Olsson T, Sørensen PS, Sellebjerg F. Systemic inflammation in progressive multiple sclerosis involves follicular T-helper, Th17- and activated B-cells and correlates with progression. PLoS One 2013; 8:e57820. [PMID: 23469245 PMCID: PMC3585852 DOI: 10.1371/journal.pone.0057820] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 01/26/2013] [Indexed: 02/06/2023] Open
Abstract
Pathology studies of progressive multiple sclerosis (MS) indicate a major role of inflammation including Th17-cells and meningeal inflammation with ectopic lymphoid follicles, B-cells and plasma cells, the latter indicating a possible role of the newly identified subset of follicular T-helper (TFH) cells. Although previous studies reported increased systemic inflammation in progressive MS it remains unclear whether systemic inflammation contributes to disease progression and intrathecal inflammation. This study aimed to investigate systemic inflammation in progressive MS and its relationship with disease progression, using flow cytometry and gene expression analysis of CD4+ and CD8+T-cells, B-cells, monocytes and dendritic cells. Furthermore, gene expression of cerebrospinal fluid cells was studied. Flow cytometry studies revealed increased frequencies of ICOS+TFH-cells in peripheral blood from relapsing-remitting (RRMS) and secondary progressive (SPMS) MS patients. All MS subtypes had decreased frequencies of Th1 TFH-cells, while primary progressive (PPMS) MS patients had increased frequency of Th17 TFH-cells. The Th17-subset, interleukin-23-receptor+CD4+T-cells, was significantly increased in PPMS and SPMS. In the analysis of B-cells, we found a significant increase of plasmablasts and DC-SIGN+ and CD83+B-cells in SPMS. ICOS+TFH-cells and DC-SIGN+B-cells correlated with disease progression in SPMS patients. Gene expression analysis of peripheral blood cell subsets substantiated the flow cytometry findings by demonstrating increased expression of IL21, IL21R and ICOS in CD4+T-cells in progressive MS. Cerebrospinal fluid cells from RRMS and progressive MS (pooled SPMS and PPMS patients) had increased expression of TFH-cell and plasmablast markers. In conclusion, this study is the first to demonstrate the potential involvement of activated TFH-cells in MS. The increased frequencies of Th17-cells, activated TFH- and B-cells parallel findings from pathology studies which, along with the correlation between activated TFH- and B-cells and disease progression, suggest a pathogenic role of systemic inflammation in progressive MS. These observations may have implications for the treatment of progressive MS.
Collapse
|
45
|
Ishikawa K, Miyamoto M, Yoshioka T, Kadoya M, Li L, Mishra R, Ichinokawa K, Shoji Y, Matsumura Y, Hida Y, Kaga K, Kato T, Kaji M, Ohbuchi T, Itoh T, Dosaka-Akita H, Matsui Y, Hirano S. Method for the validation of immunohistochemical staining using SCID mouse xenografts: expression of CD40 and CD154 in human non-small cell lung cancer. Oncol Rep 2013; 29:1315-21. [PMID: 23404288 PMCID: PMC3621727 DOI: 10.3892/or.2013.2275] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 12/21/2012] [Indexed: 11/06/2022] Open
Abstract
This report proposes a concept for the standardization of immunohistochemical evaluation. Immunohistochemical staining has several problems associated with the sensitivity of the technical process and standardization of the assessment of potent staining. We provided data focusing on this concept through immunostaining for CD154 in non-small cell lung cancer (NSCLC). We used two types of anti-CD154 antibody as primary antibodies in immunohistochemical staining, as previously reported. Western blot analysis confirmed strong CD154 expression in the cultured cell line PC10, but not in LK2. We also assessed CD154 expression in SCID mouse xenografts of these cell lines. SCID xenograft data on western blot analysis were consistent with those of cultured cell lines. These xenografts could thus be used as positive or negative tissue controls for CD154 immunostaining. Primary antibodies should therefore be confirmed as recognizing target lesions, while control tissue specimens should be objectively confirmed as having target products using another experimental method. Our method would allow results to be unified at more than one laboratory and could act as an objective control assessment method in immunohistochemistry.
Collapse
Affiliation(s)
- Keidai Ishikawa
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, and Department of Thoracic Surgery, Sapporo-Minamisanjo Hospital, Sapporo, Hokkaido 060-8638, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Onset of multiple sclerosis in childhood occurs in 3-5% of patients. There is limited, but growing knowledge about the underlying pathobiology of pediatric MS. It is crucial to better understand this area in order to address central questions in the field: 1) Can pediatric multiple sclerosis inform us about factors related to disease initiation and propagation? 2) What are the biomarkers of disease course in pediatric multiple sclerosis; 3) Does pediatric multiple sclerosis pathogenesis differ from adult-onset multiple sclerosis; 4) How can we optimize treatment in pediatric demyelinating diseases? 5) Can pediatric multiple sclerosis provide insights into the environmental risk factors for multiple sclerosis in general? Here we review the current knowledge of the pathogenesis of multiple sclerosis in children, and address the five questions raised above.
Collapse
Affiliation(s)
- David Vargas-Lowy
- Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
47
|
Abstract
Demyelinating diseases such as multiple sclerosis are chronic inflammatory autoimmune diseases with a heterogeneous clinical presentation and course. Both the adaptive and the innate immune systems have been suggested to contribute to their pathogenesis and recovery. In this review, we discuss the role of the innate immune system in mediating demyelinating diseases. In particular, we provide an overview of the anti-inflammatory or pro-inflammatory functions of dendritic cells, mast cells, natural killer (NK) cells, NK-T cells, γδ T cells, microglial cells, and astrocytes. We emphasize the interaction of astroctyes with the immune system and how this interaction relates to the demyelinating pathologies. Given the pivotal role of the innate immune system, it is possible that targeting these cells may provide an effective therapeutic approach for demyelinating diseases.
Collapse
Affiliation(s)
- Lior Mayo
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
48
|
Smith KM, Guerau-de-Arellano M, Costinean S, Williams JL, Bottoni A, Mavrikis Cox G, Satoskar AR, Croce CM, Racke MK, Lovett-Racke AE, Whitacre CC. miR-29ab1 deficiency identifies a negative feedback loop controlling Th1 bias that is dysregulated in multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2012; 189:1567-76. [PMID: 22772450 DOI: 10.4049/jimmunol.1103171] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Th cell programming and function is tightly regulated by complex biological networks to prevent excessive inflammatory responses and autoimmune disease. The importance of microRNAs (miRNAs) in this process is highlighted by the preferential Th1 polarization of Dicer-deficient T cells that lack miRNAs. Using genetic knockouts, we demonstrate that loss of endogenous miR-29, derived from the miR-29ab1 genomic cluster, results in unrestrained T-bet expression and IFN-γ production. miR-29b regulates T-bet and IFN-γ via a direct interaction with the 3' untranslated regions, and IFN-γ itself enhances miR-29b expression, establishing a novel regulatory feedback loop. miR-29b is increased in memory CD4(+) T cells from multiple sclerosis (MS) patients, which may reflect chronic Th1 inflammation. However, miR-29b levels decrease significantly upon T cell activation in MS patients, suggesting that this feedback loop is dysregulated in MS patients and may contribute to chronic inflammation. miR-29 thus serves as a novel regulator of Th1 differentiation, adding to the understanding of T cell-intrinsic regulatory mechanisms that maintain a balance between protective immunity and autoimmunity.
Collapse
Affiliation(s)
- Kristen M Smith
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
During the last decade, several defects in self-tolerance have been identified in multiple sclerosis. Dysfunction in central tolerance leads to the thymic output of antigen-specific T cells with T cell receptor alterations favouring autoimmune reactions. In addition, premature thymic involution results in a reduced export of naïve regulatory T cells, the fully suppressive clone. Alterations in peripheral tolerance concern costimulatory molecules as well as transcriptional and epigenetic mechanisms. Recent data underline the key role of regulatory T cells that suppress Th1 and Th17 effector cell responses and whose immunosuppressive activity is impaired in patients with multiple sclerosis. Those recent observations suggest that a defect in self-tolerance homeostasis might be the primary mover of multiple sclerosis leading to subsequent immune attacks, inflammation and neurodegeneration. The concept of multiple sclerosis as a consequence of the failure of central and peripheral tolerance mechanisms to maintain a self-tolerance state, particularly of regulatory T cells, may have therapeutic implications. Restoring normal thymic output and suppressive functions of regulatory T cells appears an appealing approach. Regulatory T cells suppress the general local immune response via bystander effects rather than through individual antigen-specific responses. Interestingly, the beneficial effects of currently approved immunomodulators (interferons β and glatiramer acetate) are associated with a restored regulatory T cell homeostasis. However, the feedback regulation between Th1 and Th17 effector cells and regulatory T cells is not so simple and tolerogenic mechanisms also involve other regulatory cells such as B cells, dendritic cells and CD56(bright) natural killer cells.
Collapse
Affiliation(s)
- R E Gonsette
- Fondation-Charcot-Stichting, Avenue Huart Hamoir 48, 1030 Brussels, Belgium.
| |
Collapse
|
50
|
Frisullo G, Plantone D, Marti A, Iorio R, Damato V, Nociti V, Patanella AK, Bianco A, Batocchi AP. Type 1 immune response in progressive multiple sclerosis. J Neuroimmunol 2012; 249:112-6. [PMID: 22613700 DOI: 10.1016/j.jneuroim.2012.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 04/26/2012] [Indexed: 10/28/2022]
Abstract
The aim of the study was to evaluate the type-1 immune response by analyzing T-bet expression in circulating T and B cells in Primary Progressive (PP) and Secondary Progressive (SP) Multiple Sclerosis (MS) patients. We found higher percentages of circulating CD4+T-bet+ and CD8+T-bet+ T cells in SPMS and PPMS than in remitting-relapsing MS patients and controls. Moreover, in SPMS, we observed a positive correlation between the percentages of circulating CD4+T-bet+ or CD8+T-bet+ T cells and disease severity. The increased percentages of Th1 and Tc1 cells suggest that MS progressive forms, unlike RRMS, are characterized by a permanent peripheral type-1 immune activation.
Collapse
Affiliation(s)
- Giovanni Frisullo
- Department of Neurology, Catholic University of Sacred Heart Rome, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|