1
|
Schenck JK, Clarkson-Paredes C, Pushkarsky T, Wang Y, Miller RH, Bukrinsky MI. Nef mediates neuroimmune response, myelin impairment, and neuronal injury in EcoHIV-infected mice. Life Sci Alliance 2025; 8:e202402879. [PMID: 39532531 PMCID: PMC11557684 DOI: 10.26508/lsa.202402879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
The introduction of antiretroviral therapy has markedly improved the management of HIV-associated neurocognitive disorders (HAND). However, HAND still affects nearly half of HIV-infected individuals, presenting significant challenges to their well-being. This highlights the critical need for a deeper understanding of HAND mechanisms. Among HIV viral proteins, Nef is notable for its multifaceted role in HIV pathogenesis, though its specific involvement in HAND remains unclear. To investigate this, we used a murine model infected with Nef-expressing (EcoHIV) and Nef-deficient (EcoHIVΔNef) murine HIV. Comparative analyses revealed increased neuroinflammation and reduced myelin and neuronal integrity in EcoHIV-infected brains compared with those with EcoHIVΔNef. Both viruses induced astrogliosis, with stronger GFAP activation in Nef-deficient infections. These findings suggest that Nef contributes to neuroinflammation, primarily through microglial targeting and demyelination, although other factors may regulate astrogliosis. Our results indicate that Nef may significantly contribute to neuronal injury in EcoHIV-infected mice, offering insights into Nef-induced neuropathology in HAND and guiding future research.
Collapse
Affiliation(s)
- Jessica K Schenck
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Cheryl Clarkson-Paredes
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Tatiana Pushkarsky
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Yongsen Wang
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Robert H Miller
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Michael I Bukrinsky
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| |
Collapse
|
2
|
Anselmo F, Tatomir A, Boodhoo D, Mekala AP, Nguyen V, Rus V, Rus H. JNK and phosphorylated Bcl-2 predict multiple sclerosis clinical activity and glatiramer acetate therapeutic response. Clin Immunol 2019; 210:108297. [PMID: 31698073 DOI: 10.1016/j.clim.2019.108297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/15/2019] [Accepted: 11/03/2019] [Indexed: 01/04/2023]
Abstract
In this study, we investigated the role of JNK and phospho-Bcl-2 as possible biomarkers of multiple sclerosis (MS) relapse and of glatiramer acetate (GA) therapeutic response in relapsing-remitting MS patients. We enrolled a cohort of 15 GA-treated patients and measured the expression of JNK1, JNK2, phospho-JNK and phospho-Bcl-2 through Western blotting of lysates from peripheral blood mononuclear cells collected at 0, 3, 6, and 12 months after initiating GA therapy. We found significantly higher levels of JNK1 p54 and JNK2 p54 and significantly lower levels of p-Bcl-2 in relapse patients and in GA non-responders. By using receiver operating characteristic analysis, we found that the probability of accurately detecting relapse and response to GA was: 92% and 75.5%, respectively, for JNK1 p54 and 86% and 94.6%, respectively, for p-Bcl-2. Our data suggest that JNK1 and p-Bcl-2 could serve as potential biomarkers for MS relapse and the therapeutic response to GA.
Collapse
Affiliation(s)
- Freidrich Anselmo
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alexandru Tatomir
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Neurosciences, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Armugam P Mekala
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Horea Rus
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA.
| |
Collapse
|
3
|
Chamberlain KA, Chapey KS, Nanescu SE, Huang JK. Creatine Enhances Mitochondrial-Mediated Oligodendrocyte Survival After Demyelinating Injury. J Neurosci 2017; 37:1479-1492. [PMID: 28069926 PMCID: PMC5299567 DOI: 10.1523/jneurosci.1941-16.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 12/03/2016] [Accepted: 12/28/2016] [Indexed: 01/11/2023] Open
Abstract
Chronic oligodendrocyte loss, which occurs in the demyelinating disorder multiple sclerosis (MS), contributes to axonal dysfunction and neurodegeneration. Current therapies are able to reduce MS severity, but do not prevent transition into the progressive phase of the disease, which is characterized by chronic neurodegeneration. Therefore, pharmacological compounds that promote oligodendrocyte survival could be beneficial for neuroprotection in MS. Here, we investigated the role of creatine, an organic acid involved in adenosine triphosphate (ATP) buffering, in oligodendrocyte function. We found that creatine increased mitochondrial ATP production directly in oligodendrocyte lineage cell cultures and exerted robust protection on oligodendrocytes by preventing cell death in both naive and lipopolysaccharide-treated mixed glia. Moreover, lysolecithin-mediated demyelination in mice deficient in the creatine-synthesizing enzyme guanidinoacetate-methyltransferase (Gamt) did not affect oligodendrocyte precursor cell recruitment, but resulted in exacerbated apoptosis of regenerated oligodendrocytes in central nervous system (CNS) lesions. Remarkably, creatine administration into Gamt-deficient and wild-type mice with demyelinating injury reduced oligodendrocyte apoptosis, thereby increasing oligodendrocyte density and myelin basic protein staining in CNS lesions. We found that creatine did not affect the recruitment of macrophages/microglia into lesions, suggesting that creatine affects oligodendrocyte survival independently of inflammation. Together, our results demonstrate a novel function for creatine in promoting oligodendrocyte viability during CNS remyelination.SIGNIFICANCE STATEMENT We report that creatine enhances oligodendrocyte mitochondrial function and protects against caspase-dependent oligodendrocyte apoptosis during CNS remyelination. This work has important implications for the development of therapeutic targets for diseases characterized by oligodendrocyte death, including multiple sclerosis.
Collapse
Affiliation(s)
- Kelly A Chamberlain
- Department of Biology and
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia 20057
| | | | | | - Jeffrey K Huang
- Department of Biology and
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia 20057
| |
Collapse
|
4
|
Patergnani S, Fossati V, Bonora M, Giorgi C, Marchi S, Missiroli S, Rusielewicz T, Wieckowski MR, Pinton P. Mitochondria in Multiple Sclerosis: Molecular Mechanisms of Pathogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 328:49-103. [PMID: 28069137 DOI: 10.1016/bs.ircmb.2016.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria, the organelles that function as the powerhouse of the cell, have been increasingly linked to the pathogenesis of many neurological disorders, including multiple sclerosis (MS). MS is a chronic inflammatory demyelinating disease of the central nervous system (CNS) and a leading cause of neurological disability in young adults in the western world. Its etiology remains unknown, and while the inflammatory component of MS has been heavily investigated and targeted for therapeutic intervention, the failure of remyelination and the process of axonal degeneration are still poorly understood. Recent studies suggest a role of mitochondrial dysfunction in the neurodegenerative aspects of MS. This review is focused on mitochondrial functions under physiological conditions and the consequences of mitochondrial alterations in various CNS disorders. Moreover, we summarize recent findings linking mitochondrial dysfunction to MS and discuss novel therapeutic strategies targeting mitochondria-related pathways as well as emerging experimental approaches for modeling mitochondrial disease.
Collapse
Affiliation(s)
- S Patergnani
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - V Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - M Bonora
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - C Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - S Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - S Missiroli
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - T Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - M R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - P Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
5
|
Kruszewski AM, Rao G, Tatomir A, Hewes D, Tegla CA, Cudrici CD, Nguyen V, Royal W, Bever CT, Rus V, Rus H. RGC-32 as a potential biomarker of relapse and response to treatment with glatiramer acetate in multiple sclerosis. Exp Mol Pathol 2015; 99:498-505. [PMID: 26407760 DOI: 10.1016/j.yexmp.2015.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023]
Abstract
Currently there is critical need for the identification of reliable biomarkers to help guide clinical management of multiple sclerosis (MS) patients. We investigated the combined roles of Response Gene to Complement 32 (RGC-32), FasL, CDC2, AKT, and IL-21 as possible biomarkers of relapse and response to glatiramer acetate (GA) treatment in relapsing-remitting MS (RRMS) patients. Over the course of 2 years, a cohort of 15 GA-treated RRMS patients was clinically monitored and peripheral blood mononuclear cells (PBMCs) were collected at 0, 3, 6, and 12 months. Target gene mRNA expression was measured in patients' isolated PBMCs by real-time qRT-PCR. Compared to stable MS patients, those with acute relapses exhibited decreased expression of RGC-32 (p<0.0001) and FasL (p<0.0001), increased expression of IL-21 (p=0.04), but no change in CDC2 or AKT. Compared to non-responders, responders to GA treatment showed increased expression of RGC-32 (p<0.0001) and FasL (p<0.0001), and decreased expression of IL-21 (p=0.02). Receiver operating characteristic (ROC) analysis was used to assess the predictive accuracy of each putative biomarker. The probability of accurately detecting relapse was 90% for RGC-32, 88% for FasL, and 75% for IL-21. The probability of accurately detecting response to GA was 85% for RGC-32, 90% for FasL, and 85% for IL-21. Our data suggest that RGC-32, FasL, and IL-21 could serve as potential biomarkers for the detection of MS relapse and response to GA therapy.
Collapse
Affiliation(s)
- Adam M Kruszewski
- Department of Neurology, University of Maryland, School of Medicine, United States
| | - Gautam Rao
- Department of Neurology, University of Maryland, School of Medicine, United States
| | - Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, United States
| | - Daniel Hewes
- Department of Neurology, University of Maryland, School of Medicine, United States
| | - Cosmin A Tegla
- Department of Neurology, University of Maryland, School of Medicine, United States; Research Service, Veterans Administration Maryland Health Care System, United States
| | - Cornelia D Cudrici
- Department of Neurology, University of Maryland, School of Medicine, United States
| | - Vingh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, United States
| | - Walter Royal
- Department of Neurology, University of Maryland, School of Medicine, United States; Veterans Administration Multiple Sclerosis Center of Excellence East, Baltimore, MD, USA
| | - Christopher T Bever
- Department of Neurology, University of Maryland, School of Medicine, United States; Research Service, Veterans Administration Maryland Health Care System, United States; Veterans Administration Multiple Sclerosis Center of Excellence East, Baltimore, MD, USA
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, United States
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, United States; Research Service, Veterans Administration Maryland Health Care System, United States; Veterans Administration Multiple Sclerosis Center of Excellence East, Baltimore, MD, USA.
| |
Collapse
|
6
|
Wu W, Shao J, Lu H, Xu J, Zhu A, Fang W, Hui G. Guard of delinquency? A role of microglia in inflammatory neurodegenerative diseases of the CNS. Cell Biochem Biophys 2015; 70:1-8. [PMID: 24633457 DOI: 10.1007/s12013-014-9872-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Activation of microglia and inflammation-mediated neurotoxicity are believed to play an important role in the pathogenesis of several neurodegenerative disorders, including multiple sclerosis. Studies demonstrate complex functions of activated microglia that can lead to either beneficial or detrimental outcomes, depending on the form and the timing of activation. Combined with genetic and environmental factors, overactivation and dysregulation of microglia cause progressive neurotoxic consequences which involve a vicious cycle of neuron injury and unregulated neuroinflammation. Thus, modulation of microglial activation appears to be a promising new therapeutic target. While current therapies do attempt to block activation of microglia, they indiscriminately inhibit inflammation thus also curbing beneficial effects of inflammation and delaying recovery. Multiple signaling cascades, often cross-talking, are involved in every step of microglial activation. One of the key challenges is to understand the molecular mechanisms controlling cytokine expression and phagocytic activity, as well as cell-specific consequences of dysregulated cytokine expression. Further, a better understanding of how the integration of multiple cytokine signals influences the function or activity of individual microglia remains an important research objective to identify potential therapeutic targets for clinical intervention to promote repair.
Collapse
Affiliation(s)
- Weijiang Wu
- Department of Neurosurgery, Wuxi Third People's Hospital, Wuxi, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
7
|
Aharoni R. New findings and old controversies in the research of multiple sclerosis and its model experimental autoimmune encephalomyelitis. Expert Rev Clin Immunol 2014; 9:423-40. [DOI: 10.1586/eci.13.21] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
8
|
Hartung HP, Aktas O, Menge T, Kieseier BC. Immune regulation of multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:3-14. [PMID: 24507511 DOI: 10.1016/b978-0-444-52001-2.00001-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Multiple sclerosis (MS) is considered a prototype inflammatory autoimmune disorder of the central nervous system (CNS). The etiology of this disease remains unknown, but an interplay between as yet unidentified environmental factors and susceptibility genes appears most likely. In consequence, these factors trigger a cascade, involving an inflammatory response within the CNS that results in demyelination, oligodendrocyte death, axonal damage, gliosis, and neurodegeneration. How these complex traits translate into the clinical presentation of the disease is a focus of ongoing research. The central hypothesis is that T lymphocytes with receptors for CNS myelin components are driving the disease. The initial activation of autoreactive lymphocytes is thought to take place in the systemic lymphoid organs, most likely through molecular mimickry or nonspecifically through bystander activation. These autoreactive lymphocytes can migrate to the CNS where they become reactivated upon encountering their target antigen, initiating an autoimmune inflammatory attack. This ultimately leads to demyelination and axonal damage. This chapter focuses on the role of T and B lymphocytes in the immunopathogenesis of MS.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Til Menge
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Bernd C Kieseier
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
9
|
Gui T, Wang Y, Zhang L, Wang W, Zhu H, Ding W. Krüppel-like factor 6 rendered rat Schwann cell more sensitive to apoptosis via upregulating FAS expression. PLoS One 2013; 8:e82449. [PMID: 24324791 PMCID: PMC3853331 DOI: 10.1371/journal.pone.0082449] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/02/2013] [Indexed: 12/12/2022] Open
Abstract
Krüppel-like factor 6 (KLF6) is a tumor suppressor gene and play a role in the regulation of cell proliferation and apoptosis. After the peripheral nerve injury (PNI), the microenvironment created by surrounding Schwann cells (SCs) is a critical determinant of its regenerative potential. In this study, we examined the effects of KLF6 on SCs responses during PNI. Both KLF6 mRNA and protein expression levels were upregulated in the injured sciatic nerve, and immunofluorescence results showed that many KLF6-positive cells simultaneously expressed the SC markers S-100 and p75NTR. The apoptosis inducers TNFα and cisplatin upregulated KLF6 expression in primary cultured SCs and the SC line RSC96. Although KLF6 overexpression exacerbated cisplatin- and TNFα-induced apoptosis, expression levels of the apoptosis regulators Bcl2 and Bax were not significantly affected in either KLF6-overexpressing or KLF6-depleted RSC96 cells. Realtime PCR arrays and qRT-PCR demonstrated that KLF6 overexpression upregulated four pro-apoptotic genes, FAS, TNF, TNFSF12, and PYCARD, and inhibited expression of the anti-apoptotic IL10 gene expression. Further analysis revealed that FAS protein expression was positively correlated with KLF6 expression in SCs. These data suggest that KLF6 upregulation may render SCs more vulnerable to apoptosis after injury via upregulating FAS expression.
Collapse
Affiliation(s)
- Ting Gui
- Department of Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueming Wang
- Department of Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lixing Zhang
- State Key Laboratrory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjing Wang
- Department of Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Zhu
- Department of Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenlong Ding
- Department of Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
10
|
Moreno M, Sáenz-Cuesta M, Castilló J, Cantó E, Negrotto L, Vidal-Jordana A, Montalban X, Comabella M. Circulating levels of soluble apoptosis-related molecules in patients with multiple sclerosis. J Neuroimmunol 2013; 263:152-4. [DOI: 10.1016/j.jneuroim.2013.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/16/2013] [Accepted: 07/17/2013] [Indexed: 11/26/2022]
|
11
|
Wangdong X. CD70: probably being a therapeutic target in human multiple sclerosis. Rheumatol Int 2013; 33:815. [DOI: 10.1007/s00296-011-2112-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 08/22/2011] [Indexed: 11/24/2022]
|
12
|
Tegla CA, Cudrici CD, Azimzadeh P, Singh AK, Trippe R, Khan A, Chen H, Andrian-Albescu M, Royal W, Bever C, Rus V, Rus H. Dual role of Response gene to complement-32 in multiple sclerosis. Exp Mol Pathol 2012; 94:17-28. [PMID: 23000427 DOI: 10.1016/j.yexmp.2012.09.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/10/2012] [Indexed: 01/17/2023]
Abstract
Response gene to complement (RGC)-32 is a novel molecule that plays an important role in cell proliferation. We investigated the expression of RGC-32 in multiple sclerosis (MS) brain and in peripheral blood mononuclear cells (PBMCs) obtained from patients with relapsing-remitting multiple sclerosis. We found that CD3(+), CD68(+), and glial fibrillar acidic protein (GFAP)(+) cells in MS plaques co-localized with RGC-32. Our results show a statistically significant decrease in RGC-32 mRNA expression in PBMCs during relapses when compared to the levels in stable MS patients. This decrease might be useful in predicting disease activity in patients with relapsing-remitting MS. RGC-32 expression was also correlated with that of FasL mRNA during relapses. FasL mRNA expression was significantly reduced after RGC-32 silencing, indicating a role for RGC-32 in the regulation of FasL expression. In addition, the expression of Akt1, cyclin D1, and IL-21 mRNA was significantly increased during MS relapses when compared to levels in healthy controls. Furthermore, we investigated the role of RGC-32 in TGF-β-induced extracellular matrix expression in astrocytes. Blockage of RGC-32 using small interfering RNA significantly inhibits TGF-β induction of procollagen I, fibronectin and of the reactive astrocyte marker α-smooth muscle actin (α-SMA). Our data suggest that RGC-32 plays a dual role in MS, both as a regulator of T-cells mediated apoptosis and as a promoter of TGF-β-mediated profibrotic effects in astrocytes.
Collapse
Affiliation(s)
- Cosmin A Tegla
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Brambilla R, Ashbaugh JJ, Magliozzi R, Dellarole A, Karmally S, Szymkowski DE, Bethea JR. Inhibition of soluble tumour necrosis factor is therapeutic in experimental autoimmune encephalomyelitis and promotes axon preservation and remyelination. ACTA ACUST UNITED AC 2011; 134:2736-54. [PMID: 21908877 DOI: 10.1093/brain/awr199] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tumour necrosis factor is linked to the pathophysiology of various neurodegenerative disorders including multiple sclerosis. Tumour necrosis factor exists in two biologically active forms, soluble and transmembrane. Here we show that selective inhibition of soluble tumour necrosis factor is therapeutic in experimental autoimmune encephalomyelitis. Treatment with XPro1595, a selective soluble tumour necrosis factor blocker, improves the clinical outcome, whereas non-selective inhibition of both forms of tumour necrosis factor with etanercept does not result in protection. The therapeutic effect of XPro1595 is associated with axon preservation and improved myelin compaction, paralleled by increased expression of axon-specific molecules (e.g. neurofilament-H) and reduced expression of non-phosphorylated neurofilament-H which is associated with axon damage. XPro1595-treated mice show significant remyelination accompanied by elevated expression of myelin-specific genes and increased numbers of oligodendrocyte precursors. Immunohistochemical characterization of tumour necrosis factor receptors in the spinal cord following experimental autoimmune encephalomyelitis shows tumour necrosis factor receptor 1 expression in neurons, oligodendrocytes and astrocytes, while tumour necrosis factor receptor 2 is localized in oligodendrocytes, oligodendrocyte precursors, astrocytes and macrophages/microglia. Importantly, a similar pattern of expression is found in post-mortem spinal cord of patients affected by progressive multiple sclerosis, suggesting that pharmacological modulation of tumour necrosis factor receptor signalling may represent an important target in affecting not only the course of mouse experimental autoimmune encephalomyelitis but human multiple sclerosis as well. Collectively, our data demonstrate that selective inhibition of soluble tumour necrosis factor improves recovery following experimental autoimmune encephalomyelitis, and that signalling mediated by transmembrane tumour necrosis factor is essential for axon and myelin preservation as well as remyelination, opening the possibility of a new avenue of treatment for multiple sclerosis.
Collapse
Affiliation(s)
- Roberta Brambilla
- The Miami Project To Cure Paralysis, Miller School of Medicine, University of Miami 1095 NW 14th Terrace, Miami, FL 33136, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Neuroprotective effect of combination therapy of glatiramer acetate and epigallocatechin-3-gallate in neuroinflammation. PLoS One 2011; 6:e25456. [PMID: 22022398 PMCID: PMC3192751 DOI: 10.1371/journal.pone.0025456] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 09/05/2011] [Indexed: 01/14/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory autoimmune disease of the central nervous system. However, studies of MS and the animal model, experimental autoimmune encephalomyelitis (EAE), indicate that neuronal pathology is the principle cause of clinical disability. Thus, there is need to develop new therapeutic strategies that not only address immunomodulation but also neuroprotection. Here we show that the combination therapy of Glatiramer acetate (GA), an immunomodulatory MS therapeutic, and the neuroprotectant epigallocatechin-3-gallate (EGCG), the main phenol in green tea, have synergistic protective effects in vitro and in the EAE model. EGCG and GA together led to increased protection from glutamate- and TRAIL-induced neuronal cell death in vitro. EGCG combined with GA induced regeneration of hippocampal axons in an outgrowth assay. The combined application of EGCG and GA did not result in unexpected adverse events in vivo. Neuroprotective and neuroregenerative effects could be translated in the in vivo model, where combination treatment with EGCG and GA significantly delayed disease onset, strongly reduced clinical severity, even after onset of symptoms and reduced inflammatory infiltrates. These results illustrate the promise of combining neuroprotective and anti-inflammatory treatments and strengthen the prospects of EGCG as an adjunct therapy for neuroinflammatory and neurodegenerative diseases.
Collapse
|
15
|
Mc Guire C, Beyaert R, van Loo G. Death receptor signalling in central nervous system inflammation and demyelination. Trends Neurosci 2011; 34:619-28. [PMID: 21999927 DOI: 10.1016/j.tins.2011.09.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 09/13/2011] [Accepted: 09/18/2011] [Indexed: 12/31/2022]
Abstract
Death receptors (DRs) are members of the tumor necrosis factor receptor (TNF-R) superfamily that are characterised by the presence of a conserved intracellular death domain and are able to trigger a signalling pathway leading to apoptosis. Strong evidence suggests that DRs contribute to the pathology of tissue destructive diseases, including multiple sclerosis (MS), the most common inflammatory demyelinating disease of the central nervous system (CNS). Here, we review the evidence supporting a role for DRs in MS pathology and its implications for the development of therapeutic strategies for MS and other demyelinating pathologies of the CNS.
Collapse
Affiliation(s)
- Conor Mc Guire
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium
| | | | | |
Collapse
|
16
|
Hagman S, Raunio M, Rossi M, Dastidar P, Elovaara I. Disease-associated inflammatory biomarker profiles in blood in different subtypes of multiple sclerosis: Prospective clinical and MRI follow-up study. J Neuroimmunol 2011; 234:141-7. [DOI: 10.1016/j.jneuroim.2011.02.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 01/03/2011] [Accepted: 02/15/2011] [Indexed: 12/29/2022]
|
17
|
Kurne A, Guc D, Canpinar H, Aydin OF, Sayat G, Yörübulut M, Esendagli G, Karabudak R. Analysis of BAFF and TRAIL expression levels in multiple sclerosis patients: evaluation of expression under immunomodulatory therapy. Acta Neurol Scand 2011; 123:8-12. [PMID: 20456241 DOI: 10.1111/j.1600-0404.2010.01346.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and B cell-activating factor (BAFF), the members of tumor necrosis factor superfamily, play essential roles in immune homeostasis and may have potential contributions to the autoimmune process in multiple sclerosis (MS). MATERIAL AND METHODS Thirty-five relapsing remitting MS (RRMS) patients and 19 healthy individuals were enrolled in the study. The expression of TRAIL on peripheral blood lymphocytes was analyzed by flow cytometry. The serum levels of soluble TRAIL (sTRAIL) and soluble BAFF (sBAFF) were determined by ELISA. Further, we evaluated the effect of IFN-β on sTRAIL, sBAFF levels and on TRAIL surface expression in these patients on the third and sixth months following the treatment. RESULTS AND CONCLUSION These preliminary results signify that MS patients are heterogenous in TRAIL expression. Additionally, during the IFN-β treatment, the soluble form of TRAIL increases concomitantly as its surface expression decreases on lymphocytes. The basal sBAFF levels of patients were significantly higher than the control group and no significant change was observed. Thus, the changes in TRAIL expression may be a potential parameter indicating the response to IFN-β1 therapy at individual level.
Collapse
Affiliation(s)
- A Kurne
- Department of Neurology, Hacettepe University Hospitals, Neuroimmunology Unit, Ankara, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Hebb ALO, Moore CS, Bhan V, Robertson GS. Effects of IFN-B on TRAIL and Decoy Receptor Expression in Different Immune Cell Populations from MS Patients with Distinct Disease Subtypes. Autoimmune Dis 2010; 2011:485752. [PMID: 21253524 PMCID: PMC3022173 DOI: 10.4061/2011/485752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 11/16/2010] [Indexed: 12/11/2022] Open
Abstract
Using quantitative RT-PCR, we compared mRNA levels for TRAIL [tumor necrosis factor (TNF)-related apoptosis-inducing ligand] and its receptors in various immune cell subsets derived from the peripheral blood of untreated normal subjects (NS) and patients with distinct subtypes of multiple sclerosis (MS): active relapsing-remitting MS (RRA), quiescent relapsing-remitting MS (RRQ), secondary-progressive MS (SPMS) or primary-progressive MS (PPMS). Consistent with a role for TRAIL in the mechanism of action of interferon-β (IFN-β), TRAIL mRNA levels were increased in monocytes from patients clinically responsive to IFN-β (RRQ) but not those unresponsive to this therapeutic (RRA). TRAIL-R3 (decoy receptor) expression was elevated in T cells from untreated RRMS patients while IFN-β therapy reversed this increase suggesting that IFN-β may promote the apoptotic elimination of autoreactive T cells by increasing the amount of TRAIL available to activate TRAIL death receptors. Serum concentrations of soluble TRAIL were increased to a similar extent by IFN-β therapy in RRQ, RRA and SPMS patients that had not generated neutralizing antibodies against this cytokine. Although our findings suggest altered TRAIL signaling may play a role in MS pathogenesis and IFN-β therapy, they do not support use of TRAIL as a surrogate marker for clinical responsiveness to this therapeutic.
Collapse
Affiliation(s)
- Andrea L O Hebb
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada B3H 1X5
| | | | | | | |
Collapse
|
19
|
The role of FasL and Fas in health and disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 647:64-93. [PMID: 19760067 DOI: 10.1007/978-0-387-89520-8_5] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The FS7-associated cell surface antigen (Fas, also named CD95, APO-1 or TNFRSF6) attracted considerable interest in the field of apoptosis research since its discovery in 1989. The groups of Shin Yonehara and Peter Krammer were the first reporting extensive apoptotic cell death induction upon treating cells with Fas-specific monoclonal antibodies.1,2 Cloning of Fas3 and its ligand,4,5 FasL (also known as CD178, CD95L or TNFSF6), laid the cornerstone in establishing this receptor-ligand system as a central regulator of apoptosis in mammals. Therapeutic exploitation of FasL-Fas-mediated cytotoxicity was soon an ambitous goal and during the last decade numerous strategies have been developed for its realization. In this chapter, we will briefly introduce essential general aspects of the FasL-Fas system before reviewing its physiological and pathophysiological relevance. Finally, FasL-Fas-related therapeutic tools and concepts will be addressed.
Collapse
|
20
|
Merson TD, Binder MD, Kilpatrick TJ. Role of cytokines as mediators and regulators of microglial activity in inflammatory demyelination of the CNS. Neuromolecular Med 2010; 12:99-132. [PMID: 20411441 DOI: 10.1007/s12017-010-8112-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Accepted: 02/26/2010] [Indexed: 12/11/2022]
Abstract
As the resident innate immune cells of the central nervous system (CNS), microglia fulfil a critical role in maintaining tissue homeostasis and in directing and eliciting molecular responses to CNS damage. The human disease Multiple Sclerosis and animal models of inflammatory demyelination are characterized by a complex interplay between degenerative and regenerative processes, many of which are regulated and mediated by microglia. Cellular communication between microglia and other neural and immune cells is controlled to a large extent by the activity of cytokines. Here we review the role of cytokines as mediators and regulators of microglial activity in inflammatory demyelination, highlighting their importance in potentiating cell damage, promoting neuroprotection and enhancing cellular repair in a context-dependent manner.
Collapse
Affiliation(s)
- Tobias D Merson
- Florey Neuroscience Institutes, Centre for Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.
| | | | | |
Collapse
|
21
|
Kumar M, Makonchuk DY, Li H, Mittal A, Kumar A. TNF-like weak inducer of apoptosis (TWEAK) activates proinflammatory signaling pathways and gene expression through the activation of TGF-beta-activated kinase 1. THE JOURNAL OF IMMUNOLOGY 2009; 182:2439-48. [PMID: 19201899 DOI: 10.4049/jimmunol.0803357] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TWEAK, TNF-like weak inducer of apoptosis, is a relatively recently identified proinflammatory cytokine that functions through binding to Fn14 receptor in target cells. Although TWEAK has been shown to modulate several biological responses, the TWEAK-induced signaling pathways remain poorly understood. In this study, we tested the hypothesis that TAK1 (TGF-beta-activated kinase 1) is involved in TWEAK-induced activation of NF-kappaB and MAPK and expression of proinflammatory protein. TWEAK increased the phosphorylation and kinase activity of TAK1 in cultured myoblast and fibroblast cells. The activation of NF-kappaB was significantly inhibited in TAK1-deficient (TAK1(-/-)) mouse embryonic fibroblasts (MEF) compared with wild-type MEF. Deficiency of TAK1 also inhibited the TWEAK-induced activation of IkappaB kinase and the phosphorylation and degradation of IkappaBalpha protein. However, there was no difference in the levels of p100 protein in TWEAK-treated wild-type and TAK1(-/-) MEF. Furthermore, TWEAK-induced transcriptional activation of NF-kappaB was significantly reduced in TAK1(-/-) MEF and in C2C12 myoblasts transfected with a dominant-negative TAK1 or TAK1 short interfering RNA. TAK1 was also required for the activation of AP-1 in response to TWEAK. Activation of JNK1 and p38 MAPK, but not ERK1/2 or Akt kinase, was significantly inhibited in TAK1(-/-) MEF compared with wild-type MEF upon treatment with TWEAK. TWEAK-induced expression of proinflammatory genes such as MMP-9, CCL-2, and VCAM-1 was also reduced in TAK1(-/-) MEF compared with wild-type MEF. Furthermore, the activation of NF-kappaB and the expression of MMP-9 in response to TWEAK involved the upstream activation of Akt kinase. Collectively, our study demonstrates that TAK1 and Akt are the important components of TWEAK-induced proinflammatory signaling and gene expression.
Collapse
Affiliation(s)
- Mukesh Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | | | |
Collapse
|
22
|
Hestvik ALK, Skorstad G, Vartdal F, Holmøy T. Idiotope-specific CD4(+) T cells induce apoptosis of human oligodendrocytes. J Autoimmun 2009; 32:125-32. [PMID: 19250800 DOI: 10.1016/j.jaut.2009.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 01/09/2009] [Accepted: 01/26/2009] [Indexed: 12/19/2022]
Abstract
CD4(+) T cells specific for immunologic non-self determinants on self-IgG, idiotopes (Id), can be raised from cerebrospinal fluid (CSF) and blood of patients with multiple sclerosis (MS). To test if Id-specific CD4(+) T cells have the potential to destroy oligodendrocytes (ODCs), we analyzed their ability to induce apoptosis of human ODC cell lines. Id-specific CD4(+) T cells stimulated with either Id-bearing B cells, Id-peptide presented by other antigen presenting cells, or by anti-CD3/anti-CD28 in the absence of accessory cells induced DNA fragmentation and killed ODCs. Killing required contact between the ODCs and the T cells, it did not depend on the cytokine profile of the T cells, it was independent of other cell types, and was inhibited by a general caspase inhibitor and an anti-Fas antibody. Activated CD4(+) T cells specific for glutamic acid decarboxylase 65 also induced apoptosis, showing that killing does not depend on cognate interaction between T cells and target cells but rather on the activation status of the T cells.
Collapse
Affiliation(s)
- Anne Lise Karlsgot Hestvik
- Institute of Immunology, Faculty of Medicine, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| | | | | | | |
Collapse
|
23
|
Razmara M, Hilliard B, Ziarani AK, Murali R, Yellayi S, Ghazanfar M, Chen YH, Tykocinski ML. Fn14-TRAIL, a chimeric intercellular signal exchanger, attenuates experimental autoimmune encephalomyelitis. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:460-74. [PMID: 19147815 DOI: 10.2353/ajpath.2009.080462] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Hallmarks of the pathogenesis of autoimmune encephalomyelitis include perivascular infiltration of inflammatory cells into the central nervous system, multifocal demyelination in the brain and spinal cord, and focal neuronal degeneration. Optimal treatment of this complex disease will ultimately call for agents that target the spectrum of underlying pathogenic processes. In the present study, Fn14-TRAIL is introduced as a unique immunotherapeutic fusion protein that is designed to exchange and redirect intercellular signals within inflammatory cell networks, and, in so doing, to impact multiple pathogenic events and yield a net anti-inflammatory effect. In this soluble protein product, a Fn14 receptor component (capable of blocking the pro-inflammatory TWEAK ligand) is fused to a TRAIL ligand (capable of inhibiting activated, pathogenic T cells). Sustained Fn14-TRAIL expression was obtained in vivo using a transposon-based eukaryotic expression vector. Fn14-TRAIL expression effectively prevented chronic, nonremitting, paralytic disease in myelin oligodendrocyte glycoprotein-challenged C57BL/6 mice. Disease suppression in this model was reflected by decreases in the clinical score, disease incidence, nervous tissue inflammation, and Th1, Th2, and Th17 cytokine responses. Significantly, the therapeutic efficacy of Fn14-TRAIL could not be recapitulated simply by administering its component parts (Fn14 and TRAIL) as soluble agents, either alone or in combination. Its functional pleiotropism was manifest in its additional ability to attenuate the enhanced permeability of the blood-brain barrier that typically accompanies autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Marjaneh Razmara
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Expression of TWEAK and Its Receptor Fn14 in the Multiple Sclerosis Brain: Implications for Inflammatory Tissue Injury. J Neuropathol Exp Neurol 2008; 67:1137-48. [DOI: 10.1097/nen.0b013e31818dab90] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
26
|
McTigue DM, Tripathi RB. The life, death, and replacement of oligodendrocytes in the adult CNS. J Neurochem 2008; 107:1-19. [PMID: 18643793 DOI: 10.1111/j.1471-4159.2008.05570.x] [Citation(s) in RCA: 329] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Oligodendrocytes (OLs) are mature glial cells that myelinate axons in the brain and spinal cord. As such, they are integral to functional and efficient neuronal signaling. The embryonic lineage and postnatal development of OLs have been well-studied and many features of the process have been described, including the origin, migration, proliferation, and differentiation of precursor cells. Less clear is the extent to which OLs and damaged/dysfunctional myelin are replaced following injury to the adult CNS. OLs and their precursors are very vulnerable to conditions common to CNS injury and disease sites, such as inflammation, oxidative stress, and elevated glutamate levels leading to excitotoxicity. Thus, these cells become dysfunctional or die in multiple pathologies, including Alzheimer's disease, spinal cord injury, Parkinson's disease, ischemia, and hypoxia. However, studies of certain conditions to date have detected spontaneous OL replacement. This review will summarize current information on adult OL progenitors, mechanisms that contribute to OL death, the consequences of their loss and the pathological conditions in which spontaneous oligodendrogenesis from endogenous precursors has been observed in the adult CNS.
Collapse
Affiliation(s)
- Dana M McTigue
- Department of Neuroscience and Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA.
| | | |
Collapse
|
27
|
Butts BD, Houde C, Mehmet H. Maturation-dependent sensitivity of oligodendrocyte lineage cells to apoptosis: implications for normal development and disease. Cell Death Differ 2008; 15:1178-86. [PMID: 18483490 DOI: 10.1038/cdd.2008.70] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Apoptosis plays a crucial role in brain development by ensuring that only appropriately growing, migrating, and synapse-forming neurons and their associated glial cells survive. This process involves an intimate relationship between cell-cell interactions and developmental cues and is further impacted by environmental stress during neurogenesis and disease. Oligodendrocytes (OLs), the major myelin-forming cells in the central nervous system, largely form after this wave of neurogenesis but also show a selective vulnerability to cell death stimuli depending on their stage of development. This can affect not only embryonic and early postnatal brain formation but also the response to demyelinating pathologies. In the present review, we discuss the stage-specific sensitivity of OL lineage cells to damage-induced death and how this might impact myelin survival and regeneration during injury or disease.
Collapse
Affiliation(s)
- B D Butts
- Apoptosis Research Group, Merck Research Laboratories, RY80Y-215, 126 East Lincoln Avenue, Rahway, NJ 07065, USA
| | | | | |
Collapse
|
28
|
Sellner J, Greeve I, Findling O, Grandgirard D, Leib SL, Mattle HP. Atorvastatin does not alter serum levels of sCD95 and sCD95L in multiple sclerosis. Clin Exp Immunol 2008; 152:280-4. [PMID: 18341614 DOI: 10.1111/j.1365-2249.2008.03630.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Elimination of autoreactive T cells by apoptosis is critical for restricting immune responses to self-antigens. An errant lytic interaction between the CD95 death receptor and its ligand CD95L is presumed to be involved in the pathogenesis of multiple sclerosis (MS). Statins are promising agents for the treatment of MS and were shown to modulate levels of soluble death receptors. Here, we evaluated the in vivo effects by interferon (IFN)-beta and atorvastatin on soluble CD95 (sCD95) and sCD95L in serum of patients with MS. Concentrations of sCD95 and sCD95L did not show any differences between MS and healthy control subjects. In patients with MS, treatment with IFN-beta increased serum levels of sCD95 and sCD95L significantly (P < 0.01 and P < 0.05 respectively). Addition of atorvastatin to IFN-beta did not alter serum levels of sCD95 and sCD95L significantly. Our study suggests that atorvastatin does not affect IFN-beta-induced increases of the soluble death receptors in the serum of patients with MS.
Collapse
Affiliation(s)
- J Sellner
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | | | | | | | | |
Collapse
|
29
|
Pedersen LB, Nashold FE, Spach KM, Hayes CE. 1,25-dihydroxyvitamin D3 reverses experimental autoimmune encephalomyelitis by inhibiting chemokine synthesis and monocyte trafficking. J Neurosci Res 2007; 85:2480-90. [PMID: 17600374 DOI: 10.1002/jnr.21382] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is a complex neurodegenerative disease whose pathogenesis involves genetic and environmental risk factors leading to an aberrant, neuroantigen-specific, CD4+ T cell-mediated autoimmune response. In support of the hypothesis that vitamin D3 may reduce MS risk and severity, we found that vitamin D3 and 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) inhibited induction of experimental autoimmune encephalomyelitis (EAE), an MS model. To investigate how 1,25-(OH)2D3 could carry out anti-inflammatory functions, we administered 1,25-(OH)2D3 or a placebo to mice with EAE, and subsequently analyzed clinical disease, chemokines, inducible nitric oxide synthase (iNOS), and recruitment of dye-labeled monocytes. The 1,25-(OH)2D3 treatment significantly reduced clinical EAE severity within 3 days. Sharp declines in chemokines, inducible iNOS, and CD11b+ monocyte recruitment into the central nervous system (CNS) preceded this clinical disease abatement in the 1,25-(OH)2D3-treated animals. The 1,25-(OH)2D3 did not directly and rapidly inhibit chemokine synthesis in vivo or in vitro. Rather, the 1,25-(OH)2D3 rapidly stimulated activated CD4+ T cell apoptosis in the CNS and spleen. Collectively, these results support a model wherein inflammation stimulates a natural anti-inflammatory feedback loop. The activated inflammatory cells produce 1,25-(OH)2D3, and this hormone subsequently enhances the apoptotic death of inflammatory CD4+ T cells, removing the driving force for continued inflammation. In this way, the sunlight-derived hormone could reduce the risk of chronic CNS inflammation and autoimmune-mediated neurodegenerative disease.
Collapse
Affiliation(s)
- Laura B Pedersen
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | |
Collapse
|
30
|
Abstract
Hypoxic acclimatization includes increased brain capillary density. Adaptive angiogenesis, which occurs over a 3-week period, is mediated by upregulation of vascular endothelial growth factor induced by hypoxia-inducible factor-1 in concert with the capillary remodeling molecule angiopoietin-2, which is upregulated through cyclooxygenase-2 production of prostaglandin E2. The process is apparently orchestrated by pericytes, which regulate the microvascular milieu and coordinate the interactions within the neurovascular unit. The return to normoxia is accompanied by microvascular regression and decreasing numbers of capillaries to prehypoxic densities. Regression is the result of endothelial cell apoptosis, suggesting the existence of physiologic mechanisms for adjusting capillary density to balance oxygen availability and oxygen consumption. The capacity for adaptation is diminished in older rats because of the attenuation of the hypoxia-inducible factor-1 response.
Collapse
Affiliation(s)
- Paula Dore-Duffy
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | |
Collapse
|
31
|
Schulze-Topphoff U, Prat A, Bader M, Zipp F, Aktas O. Roles of the kallikrein/kinin system in the adaptive immune system. Int Immunopharmacol 2007; 8:155-60. [PMID: 18182219 DOI: 10.1016/j.intimp.2007.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Revised: 08/01/2007] [Accepted: 08/01/2007] [Indexed: 01/22/2023]
Abstract
This review deals with the effects of kinins, a family of octa- to decapeptides structurally related to bradykinin (BK), in adaptive immune responses. Herein, we discuss the experimental evidence that kinins may exert influence on multiple players of the immune system (i.e. macrophages, dendritic cells, T and B lymphocytes), and modulate the activation, proliferation, migration and effector functions of these cells. We also give an overview of the possible impact of kinins in human autoimmune diseases and corresponding animal models, with special emphasis on autoimmune neuroinflammation and arthritis. These studies indicate a possible immunomodulatory capacity of kinins beyond our current knowledge of kinin actions regarding the vascular system, and thus the way towards future therapeutic approaches.
Collapse
Affiliation(s)
- U Schulze-Topphoff
- Cecilie-Vogt-Clinic for Molecular Neurology, Charité-Universitätsmedizin Berlin, Germany
| | | | | | | | | |
Collapse
|
32
|
Siffrin V, Brandt AU, Herz J, Zipp F. New insights into adaptive immunity in chronic neuroinflammation. Adv Immunol 2007; 96:1-40. [PMID: 17981203 DOI: 10.1016/s0065-2776(07)96001-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Understanding the immune response in the central nervous system (CNS) is crucial for the development of new therapeutic concepts in chronic neuroinflammation, which differs considerably from other autoimmune diseases. Special immunologic properties of inflammatory processes in the CNS, which is often referred to as an immune privileged site, imply distinct features of CNS autoimmune disease in terms of disease initiation, perpetuation, and therapeutic accessibility. Furthermore, the CNS is a stress-sensitive organ with a low capacity for self-renewal and is highly prone to bystander damage caused by CNS inflammation. This leads to neuronal degeneration that contributes considerably to the phenotype of the disease. In this chapter, we discuss recent findings emphasizing the predominant role of the adaptive immune system in the pathogenesis of chronic neuroinflammation, that is, multiple sclerosis (MS) in patients and experimental autoimmune encephalomyelitis (EAE) in rodents. In addition, we report on efforts to translate these findings into clinical practice with the aim of developing selective treatment regimens.
Collapse
Affiliation(s)
- Volker Siffrin
- Cecilie-Vogt-Clinic for Molecular Neurology, Charité-Universitaetsmedizin Berlin, 10117 Berlin, Germany
| | | | | | | |
Collapse
|
33
|
Abstract
Headache and migraine are common features in multiple sclerosis (MS) and can influence the diagnosis, radiological evaluation, treatment, and quality of life of these patients. Similarities in symptoms between patients with migraine and MS presenting with headache can lead to misdiagnosis. Likewise, MRI lesions which may be found in migraine patients without other neurological symptoms or signs may cause diagnostic confusion and patient anxiety. Studies addressing the pathophysiology of these comorbid conditions have not found a clear link, but brainstem lesions and inflammatory processes have been proposed. Management of de novo or treatment-induced headache in the MS patient is fairly straightforward if the physician is aware of the problem.
Collapse
Affiliation(s)
- Debra G Elliott
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103, USA
| |
Collapse
|