1
|
Bose A, Pahan K. Build muscles and protect myelin. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:175-182. [PMID: 39741558 PMCID: PMC11683878 DOI: 10.1515/nipt-2024-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/02/2024] [Indexed: 01/03/2025]
Abstract
Multiple sclerosis (MS) is a chronic and debilitating autoimmune disease of the central nervous system (CNS) in which a CNS-driven immune response destroys myelin, leading to wide range of symptoms including numbness and tingling, vision problems, mobility impairment, etc. Oligodendrocytes are the myelinating cells in the CNS, which are generated from oligodendroglial progenitor cells (OPCs) via differentiation. However, for multiple reasons, OPCs fail to differentiate to oligodendrocytes in MS and as a result, stimulating the differentiation of OPCs to oligodendrocytes is considered beneficial for MS. The β-hydroxy β-methylbutyrate (HMB) is a widely-used muscle-building supplement in human and recently it has been shown that low-dose HMB is capable of stimulating the differentiation of cultured OPCs to oligodendrocytes for remyelination. Moreover, other causes of autoimmune demyelination are the decrease and/or suppression of Foxp3-expressing anti-autoimmune regulatory T cells (Tregs) and upregulation of autoimmune T-helper 1(Th1) and Th17 cells. Experimental autoimmune encephalomyelitis (EAE) is an animal model of MS in which the autoimmune demyelination is nicely visible. It has been reported that in EAE mice, oral HMB upregulates Tregs and decreases Th1 and Th17 responses, leading to remyelination in the CNS. Here, we analyze these newly-described features of HMB, highlighting the putative promyelinating nature of this supplement.
Collapse
Affiliation(s)
- Ahana Bose
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
2
|
Jana M, Prieto S, Gorai S, Dasarathy S, Kundu M, Pahan K. Muscle-building supplement β-hydroxy β-methylbutyrate stimulates the maturation of oligodendroglial progenitor cells to oligodendrocytes. J Neurochem 2024; 168:1340-1358. [PMID: 38419348 PMCID: PMC11260247 DOI: 10.1111/jnc.16084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/10/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Oligodendrocytes are the myelinating cells in the CNS and multiple sclerosis (MS) is a demyelinating disorder that is characterized by progressive loss of myelin. Although oligodendroglial progenitor cells (OPCs) should be differentiated into oligodendrocytes, for multiple reasons, OPCs fail to differentiate into oligodendrocytes in MS. Therefore, increasing the maturation of OPCs to oligodendrocytes may be of therapeutic benefit for MS. The β-hydroxy β-methylbutyrate (HMB) is a muscle-building supplement in humans and this study underlines the importance of HMB in stimulating the maturation of OPCs to oligodendrocytes. HMB treatment upregulated the expression of different maturation markers including PLP, MBP, and MOG in cultured OPCs. Double-label immunofluorescence followed by immunoblot analyses confirmed the upregulation of OPC maturation by HMB. While investigating mechanisms, we found that HMB increased the maturation of OPCs isolated from peroxisome proliferator-activated receptor β-/- (PPARβ-/-) mice, but not PPARα-/- mice. Similarly, GW6471 (an antagonist of PPARα), but not GSK0660 (an antagonist of PPARβ), inhibited HMB-induced maturation of OPCs. GW9662, a specific inhibitor of PPARγ, also could not inhibit HMB-mediated stimulation of OPC maturation. Furthermore, PPARα agonist GW7647, but neither PPARβ agonist GW0742 nor PPARγ agonist GW1929, alone increased the maturation of OPCs. Finally, HMB treatment of OPCs led to the recruitment of PPARα, but neither PPARβ nor PPARγ, to the PLP gene promoter. These results suggest that HMB stimulates the maturation of OPCs via PPARα and that HMB may have therapeutic prospects in remyelination.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Shelby Prieto
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sridevi Dasarathy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
3
|
Poddar J, Rangasamy SB, Pahan K. Therapeutic efficacy of cinnamein, a component of balsam of Tolu/Peru, in controlled cortical impact mouse model of TBI. Neurochem Int 2024; 176:105742. [PMID: 38641028 DOI: 10.1016/j.neuint.2024.105742] [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: 01/26/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 04/21/2024]
Abstract
Traumatic brain injury (TBI) remains a major health concern which causes long-term neurological disability particularly in war veterans, athletes and young adults. In spite of intense clinical and research investigations, there is no effective therapy to cease the pathogenesis of the disease. It is believed that axonal injury during TBI is potentiated by neuroinflammation and demyelination and/or failure to remyelination. This study highlights the use of naturally available cinnamein, also chemically known as benzyl cinnamate, in inhibiting neuroinflammation, promoting remyelination and combating the disease process of controlled cortical impact (CCI)-induced TBI in mice. Oral delivery of cinnamein through gavage brought down the activation of microglia and astrocytes to decrease the expression of inducible nitric oxide synthase (iNOS), glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) in hippocampus and cortex of TBI mice. Cinnamein treatment also stimulated remyelination in TBI mice as revealed by PLP and A2B5 double-labeling, luxol fast blue (LFB) staining and axonal double-labeling for neurofilament and MBP. Furthermore, oral cinnamein reduced the size of lesion cavity in the brain, improved locomotor functions and restored memory and learning in TBI mice. These results suggest a new neuroprotective property of cinnamein that may be valuable in the treatment of TBI.
Collapse
Affiliation(s)
- Jit Poddar
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Suresh B Rangasamy
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
4
|
Mondal S, Sheinin M, Rangasamy SB, Pahan K. Amelioration of experimental autoimmune encephalomyelitis by gemfibrozil in mice via PPARβ/δ: implications for multiple sclerosis. Front Cell Neurosci 2024; 18:1375531. [PMID: 38835441 PMCID: PMC11148333 DOI: 10.3389/fncel.2024.1375531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
It is important to describe effective and non-toxic therapies for multiple sclerosis (MS), an autoimmune demyelinating disease. Experimental autoimmune encephalomyelitis (EAE) is an immune-mediated inflammatory disease that serves as a model for MS. Earlier we and others have shown that, gemfibrozil, a lipid-lowering drug, exhibits therapeutic efficacy in EAE. However, the underlying mechanism was poorly understood. Although gemfibrozil is a known ligand of peroxisome proliferator-activated receptor α (PPARα), here, we established that oral administration of gemfibrozil preserved the integrity of blood-brain barrier (BBB) and blood-spinal cord barrier (BSB), decreased the infiltration of mononuclear cells into the CNS and inhibited the disease process of EAE in both wild type and PPARα-/- mice. On the other hand, oral gemfibrozil was found ineffective in maintaining the integrity of BBB/BSB, suppressing inflammatory infiltration and reducing the disease process of EAE in mice lacking PPARβ (formerly PPARδ), indicating an important role of PPARβ/δ, but not PPARα, in gemfibrozil-mediated preservation of BBB/BSB and protection of EAE. Regulatory T cells (Tregs) play a critical role in the disease process of EAE/MS and we also demonstrated that oral gemfibrozil protected Tregs in WT and PPARα-/- EAE mice, but not PPARβ-/- EAE mice. Taken together, our findings suggest that gemfibrozil, a known ligand of PPARα, preserves the integrity of BBB/BSB, enriches Tregs, and inhibits the disease process of EAE via PPARβ, but not PPARα.
Collapse
Affiliation(s)
- Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| | - Monica Sheinin
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Suresh B Rangasamy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| |
Collapse
|
5
|
Hinsinger G, Du Trieu De Terdonck L, Urbach S, Salvetat N, Rival M, Galoppin M, Ripoll C, Cezar R, Laurent-Chabalier S, Demattei C, Agherbi H, Castelnovo G, Lehmann S, Rigau V, Marin P, Thouvenot E. CD138 as a Specific CSF Biomarker of Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200230. [PMID: 38669615 PMCID: PMC11057439 DOI: 10.1212/nxi.0000000000200230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/30/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND AND OBJECTIVES The aim of this study was to identify novel biomarkers for multiple sclerosis (MS) diagnosis and prognosis, addressing the critical need for specific and prognostically valuable markers in the field. METHODS We conducted an extensive proteomic investigation, combining analysis of (1) CSF proteome from symptomatic controls, fast and slow converters after clinically isolated syndromes, and patients with relapsing-remitting MS (n = 10 per group) using label-free quantitative proteomics and (2) oligodendrocyte secretome changes under proinflammatory or proapoptotic conditions using stable isotope labeling by amino acids in cell culture. Proteins exhibiting differential abundance in both proteomic analyses were combined with other putative MS biomarkers, yielding a comprehensive list of 87 proteins that underwent quantification through parallel reaction monitoring (PRM) in a novel cohort, comprising symptomatic controls, inflammatory neurologic disease controls, and patients with MS at various disease stages (n = 10 per group). The 11 proteins that passed this qualification step were subjected to a new PRM assay within an expanded cohort comprising 158 patients with either MS at different disease stages or other inflammatory or noninflammatory neurologic disease controls. RESULTS This study unveiled a promising biomarker signature for MS, including previously established candidates, such as chitinase 3-like protein 1, chitinase 3-like protein 2, chitotriosidase, immunoglobulin kappa chain region C, neutrophil gelatinase-associated lipocalin, and CD27. In addition, we identified novel markers, namely cat eye syndrome critical region protein 1 (adenosine deaminase 2, a therapeutic target in multiple sclerosis) and syndecan-1, a proteoglycan, also known as plasma cell surface marker CD138 and acting as chitinase 3-like protein 1 receptor implicated in inflammation and cancer signaling. CD138 exhibited good diagnostic accuracy in distinguishing MS from inflammatory neurologic disorders (area under the curve [AUC] = 0.85, CI 0.75-0.95). CD138 immunostaining was also observed in the brains of patients with MS and cultured oligodendrocyte precursor cells but was absent in astrocytes. DISCUSSION These findings identify CD138 as a specific CSF biomarker for MS and suggest the selective activation of the chitinase 3-like protein 1/CD138 pathway within the oligodendrocyte lineage in MS. They offer promising prospects for improving MS diagnosis and prognosis by providing much-needed specificity and clinical utility. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that CD138 distinguishes multiple sclerosis from other inflammatory neurologic disorders with an AUC of 0.85 (95% CI 0.75-0.95).
Collapse
Affiliation(s)
- Geoffrey Hinsinger
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Lucile Du Trieu De Terdonck
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Serge Urbach
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Nicolas Salvetat
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Manon Rival
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Manon Galoppin
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Chantal Ripoll
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Renaud Cezar
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Sabine Laurent-Chabalier
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Christophe Demattei
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Hanane Agherbi
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Giovanni Castelnovo
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Sylvain Lehmann
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Valérie Rigau
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Philippe Marin
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Eric Thouvenot
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| |
Collapse
|
6
|
Jana M, Mondal S, Jana A, Pahan K. Induction of IL-2 by interleukin-12 p40 homodimer and IL-12, but not IL-23, in microglia and macrophages: Implications for multiple sclerosis. Cytokine 2024; 174:156457. [PMID: 38056248 PMCID: PMC10872483 DOI: 10.1016/j.cyto.2023.156457] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/11/2023] [Accepted: 11/26/2023] [Indexed: 12/08/2023]
Abstract
The level of IL-2 increases markedly in serum and central nervous system (CNS) of patients with multiple sclerosis (MS) and animals with experimental allergic encephalomyelitis (EAE). However, mechanisms by which IL-2 is induced under autoimmune demyelinating conditions are poorly understood. The present study underlines the importance of IL-12p40 homodimer (p402), the so-called biologically inactive molecule, in inducing the expression of IL-2 in mouse BV-2 microglial cells, primary mouse and human microglia, mouse peritoneal macrophages, RAW264.7 macrophages, and T cells. Interestingly, we found that p402 and IL-12p70 (IL-12), but not IL-23, dose-dependently induced the production of IL-2 and the expression of IL-2 mRNA in microglial cells. Similarly, p402 also induced the activation of IL-2 promoter in microglial cells and RAW264.7 cells. Among various stimuli tested, p402 was the most potent stimulus followed by IFN-γ, bacterial lipopolysaccharide, HIV-1 gp120, and IL-12 in inducing the activation of IL-2 promoter in microglial cells. Moreover, p402, but not IL-23, increased NFATc2 mRNA expression and the transcriptional activity of NFAT. Furthermore, induction of IL-2 mRNA expression by over-expression of p40, but not by p19, cDNA indicated that p40, but not p19, is responsible for the induction of IL-2 mRNA in microglia. Finally, by using primary microglia from IL to 12 receptor β1 deficient (IL-12Rβ1-/-) and IL-12 receptor β2 deficient (IL-12Rβ2-/-) mice, we demonstrate that p402 induces the expression of IL-2 via IL-12Rβ1, but not IL-12Rβ2. In experimental autoimmune encephalomyelitis, an animal model of MS, neutralization of p402 by mAb a3-1d led to decrease in clinical symptoms and reduction in IL-2 in T cells and microglia. These results delineate a new biological function of p402, which is missing in the so-called autoimmune cytokine IL-23, and raise the possibility of controlling increased IL-2 and the disease process of MS via neutralization of p402.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Arundhati Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue, Chicago, IL, USA.
| |
Collapse
|
7
|
Suhail H, Nematullah M, Rashid F, Sajad M, Fatma M, Singh J, Zahoor I, Cheung WL, Tiwari N, Ayasolla K, Kumar A, Hoda N, Rattan R, Giri S. An early glycolysis burst in microglia regulates mitochondrial dysfunction in oligodendrocytes under neuroinflammation. iScience 2023; 26:107921. [PMID: 37841597 PMCID: PMC10568429 DOI: 10.1016/j.isci.2023.107921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/10/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Metabolism and energy processes governing oligodendrocyte function during neuroinflammatory disease are of great interest. However, how varied cellular environments affect oligodendrocyte activity during neuroinflammation is unknown. We demonstrate that activated microglial energy metabolism controls oligodendrocyte mitochondrial respiration and activity. Lipopolysaccharide/interferon gamma promote glycolysis and decrease mitochondrial respiration and myelin protein synthesis in rat brain glial cells. Enriched microglia showed an early burst in glycolysis. In microglia-conditioned medium, oligodendrocytes did not respire and expressed less myelin. SCENITH revealed metabolic derangement in microglia and O4-positive oligodendrocytes in endotoxemia and experimental autoimmune encephalitogenic models. The early burst of glycolysis in microglia was mediated by PDPK1 and protein kinase B/AKT signaling. We found that microglia-produced NO and itaconate, a tricarboxylic acid bifurcated metabolite, reduced mitochondrial respiration in oligodendrocytes. During inflammation, we discovered a signaling pathway in microglia that could be used as a therapeutic target to restore mitochondrial function in oligodendrocytes and induce remyelination.
Collapse
Affiliation(s)
- Hamid Suhail
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | | | - Faraz Rashid
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Mir Sajad
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Mena Fatma
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Jaspreet Singh
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Insha Zahoor
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Wing Lee Cheung
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Nivedita Tiwari
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Kameshwar Ayasolla
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Ashok Kumar
- Department of Ophthalmology/Kresge Eye Institute, Department of Anatomy and Cell Biology, Department of Immunology and Microbiology, Wayne State University, Detroit, MI, USA
| | - Nasrul Hoda
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women’s Health Services, Henry Ford Health System, Detroit, MI 48202, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| |
Collapse
|
8
|
Schang AL, Van Steenwinckel J, Ioannidou ZS, Lipecki J, Rich-Griffin C, Woolley-Allen K, Dyer N, Le Charpentier T, Schäfer P, Fleiss B, Ott S, Sabéran-Djoneidi D, Mezger V, Gressens P. Epigenetic priming of immune/inflammatory pathways activation and abnormal activity of cell cycle pathway in a perinatal model of white matter injury. Cell Death Dis 2022; 13:1038. [PMID: 36513635 PMCID: PMC9748018 DOI: 10.1038/s41419-022-05483-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/10/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
Prenatal inflammatory insults accompany prematurity and provoke diffuse white matter injury (DWMI), which is associated with increased risk of neurodevelopmental pathologies, including autism spectrum disorders. DWMI results from maturation arrest of oligodendrocyte precursor cells (OPCs), a process that is poorly understood. Here, by using a validated mouse model of OPC maturation blockade, we provide the genome-wide ID card of the effects of neuroinflammation on OPCs that reveals the architecture of global cell fate issues underlining their maturation blockade. First, we find that, in OPCs, neuroinflammation takes advantage of a primed epigenomic landscape and induces abnormal overexpression of genes of the immune/inflammatory pathways: these genes strikingly exhibit accessible chromatin conformation in uninflamed OPCs, which correlates with their developmental, stage-dependent expression, along their normal maturation trajectory, as well as their abnormal upregulation upon neuroinflammation. Consistently, we observe the positioning on DNA of key transcription factors of the immune/inflammatory pathways (IRFs, NFkB), in both unstressed and inflamed OPCs. Second, we show that, in addition to the general perturbation of the myelination program, neuroinflammation counteracts the physiological downregulation of the cell cycle pathway in maturing OPCs. Neuroinflammation therefore perturbs cell identity in maturing OPCs, in a global manner. Moreover, based on our unraveling of the activity of genes of the immune/inflammatory pathways in prenatal uninflamed OPCs, the mere suppression of these proinflammatory mediators, as currently proposed in the field, may not be considered as a valid neurotherapeutic strategy.
Collapse
Affiliation(s)
- Anne-Laure Schang
- grid.464155.7Université Paris Cité, Epigenetics and Cell Fate, CNRS, F-75013 Paris, France ,grid.513208.dUniversité Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France ,grid.7429.80000000121866389Present Address: Inserm, UMR1153, Epidemiology and Biostatistics Sorbonne Paris Cité Center (CRESS) HERA team. Université Paris Cité, Faculté de Santé, Faculté de Pharmacie de Paris, 4 avenue de l’Observatoire, 75006 Paris, France
| | | | - Zoi S. Ioannidou
- grid.7372.10000 0000 8809 1613School of Life Sciences, University of Warwick, Coventry, CV4 7AL UK
| | - Julia Lipecki
- grid.7372.10000 0000 8809 1613School of Life Sciences, University of Warwick, Coventry, CV4 7AL UK
| | - Charlotte Rich-Griffin
- grid.7372.10000 0000 8809 1613School of Life Sciences, University of Warwick, Coventry, CV4 7AL UK
| | - Kate Woolley-Allen
- grid.7372.10000 0000 8809 1613School of Life Sciences, University of Warwick, Coventry, CV4 7AL UK
| | - Nigel Dyer
- grid.7372.10000 0000 8809 1613Bioinformatics Research Technology Platform, Warwick University, Coventry, CV4 7AL UK
| | | | - Patrick Schäfer
- grid.7372.10000 0000 8809 1613School of Life Sciences, University of Warwick, Coventry, CV4 7AL UK
| | - Bobbi Fleiss
- grid.513208.dUniversité Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France ,grid.1017.70000 0001 2163 3550Present Address: School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC Australia
| | - Sascha Ott
- grid.7372.10000 0000 8809 1613Warwick Medical School, University of Warwick, Coventry, CV4 7AL UK
| | | | - Valérie Mezger
- grid.464155.7Université Paris Cité, Epigenetics and Cell Fate, CNRS, F-75013 Paris, France
| | - Pierre Gressens
- grid.513208.dUniversité Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France ,grid.7372.10000 0000 8809 1613School of Life Sciences, University of Warwick, Coventry, CV4 7AL UK
| |
Collapse
|
9
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by irreversible deterioration of upper and lower motor neurons (MNs). Previously, studies on the involvement of glial cells in the pathogenic process of ALS have mainly revolved around astrocytes and microglia. And oligodendrocytes (OLs) have only recently been highlighted. Grey matter demyelination within the motor cortex and proliferation of the oligodendrocyte precursor cells (OPCs) was observed in ALS patients. The selective ablation of mutant SOD1 (the dysfunctional superoxide dismutase) from the oligodendrocyte progenitors after birth significantly delayed disease onset and prolonged the overall survival in ALS mice model (SOD1G37R). In this study, we review the several mechanisms of oligodendrocyte dysfunction involved in the pathological process of myelin damage and MNs death during ALS. Particularly, we examined the insufficient local energy supply from OLs to axons, impaired differentiation from OPCs into OLs mediated by oxidative stress damage, and inflammatory injury to the OLs. Since increasing evidence depicted that ALS is not a disease limited to MNs damage, exploring the mechanisms by which oligodendrocyte dysfunction is involved in MNs death would contribute to a more comprehensive understanding of ALS and identifying potential drug targets.
Collapse
Affiliation(s)
- Zhenxiang Gong
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Li Ba
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Zhang
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Min Zhang, Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Qiaokou District, Wuhan, Hubei 430030, China. Tel: +86-27-83663895, E-mail:
| |
Collapse
|
10
|
Spaas J, van Veggel L, Schepers M, Tiane A, van Horssen J, Wilson DM, Moya PR, Piccart E, Hellings N, Eijnde BO, Derave W, Schreiber R, Vanmierlo T. Oxidative stress and impaired oligodendrocyte precursor cell differentiation in neurological disorders. Cell Mol Life Sci 2021; 78:4615-4637. [PMID: 33751149 PMCID: PMC8195802 DOI: 10.1007/s00018-021-03802-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/12/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) account for 5% of the resident parenchymal central nervous system glial cells. OPCs are not only a back-up for the loss of oligodendrocytes that occurs due to brain injury or inflammation-induced demyelination (remyelination) but are also pivotal in plastic processes such as learning and memory (adaptive myelination). OPC differentiation into mature myelinating oligodendrocytes is controlled by a complex transcriptional network and depends on high metabolic and mitochondrial demand. Mounting evidence shows that OPC dysfunction, culminating in the lack of OPC differentiation, mediates the progression of neurodegenerative disorders such as multiple sclerosis, Alzheimer's disease and Parkinson's disease. Importantly, neurodegeneration is characterised by oxidative and carbonyl stress, which may primarily affect OPC plasticity due to the high metabolic demand and a limited antioxidant capacity associated with this cell type. The underlying mechanisms of how oxidative/carbonyl stress disrupt OPC differentiation remain enigmatic and a focus of current research efforts. This review proposes a role for oxidative/carbonyl stress in interfering with the transcriptional and metabolic changes required for OPC differentiation. In particular, oligodendrocyte (epi)genetics, cellular defence and repair responses, mitochondrial signalling and respiration, and lipid metabolism represent key mechanisms how oxidative/carbonyl stress may hamper OPC differentiation in neurodegenerative disorders. Understanding how oxidative/carbonyl stress impacts OPC function may pave the way for future OPC-targeted treatment strategies in neurodegenerative disorders.
Collapse
Affiliation(s)
- Jan Spaas
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Lieve van Veggel
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Melissa Schepers
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Assia Tiane
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Jack van Horssen
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam University Medical Center, Location VUmc, Amsterdam, The Netherlands
| | - David M Wilson
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Pablo R Moya
- Facultad de Ciencias, Instituto de Fisiología, Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| | - Elisabeth Piccart
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Niels Hellings
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Bert O Eijnde
- University MS Center (UMSC), Hasselt-Pelt, Belgium
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Faculty of Medicine and Life Sciences, SMRC-Sportsmedical Research Center, BIOMED Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Wim Derave
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Rudy Schreiber
- Department Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Tim Vanmierlo
- University MS Center (UMSC), Hasselt-Pelt, Belgium.
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.
- Department Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
11
|
Li F, Song X, Xu J, Shi Y, Hu R, Ren Z, Qi Q, Lü H, Cheng X, Hu J. Morroniside protects OLN-93 cells against H 2O 2-induced injury through the PI3K/Akt pathway-mediated antioxidative stress and antiapoptotic activities. Cell Cycle 2021; 20:661-675. [PMID: 33734020 DOI: 10.1080/15384101.2021.1889186] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative disorders, including spinal cord injury (SCI), result in oxidative stress-induced cell damage. Morroniside (MR), a major active ingredient of the Chinese herb Shan Zhu Yu, has been shown to ameliorate oxidative stress and inflammatory response. Our previous study also confirmed that morroniside protects SK-N-SH cell line (human neuroblastoma cells) against oxidative impairment. However, it remains unclear whether MR also plays a protective role for oligodendrocytes that are damaged following SCI. The present study investigated the protective effects of MR against hydrogen peroxide (H2O2)-induced cell death in OLN-93 cells. MR protected OLN-93 cells from H2O2-induced injury, attenuated H2O2-induced increase in reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and blocked the reduction of mitochondrial membrane potential (MMP) induced by H2O2. MR enhanced the activity of the antioxidant enzyme superoxide dismutase (SOD) and suppressed H2O2-induced downregulation of the antiapoptotic protein Bcl-2 and activation of the proapoptotic protein caspase-3. Finally, we found that LY294002, a specific inhibitor of the PI3K/Akt pathway, inhibited the protective effect of MR against H2O2-induced OLN-93 cell injury in the MTT and TUNEL assays. LY294002 also inhibited the expression of SOD and Bcl-2, and increased the expression of iNOS and c-caspase-3 induced by MR treatment. MR exerts protective effects against H2O2-induced OLN-93 cell injury through the PI3K/Akt signaling pathway-mediated antioxidative stress and antiapoptotic activities. MR may provide a potential strategy for SCI treatment or other related neurodegeneration.
Collapse
Affiliation(s)
- Fengzhi Li
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Xue Song
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Jiaxin Xu
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Yujiao Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Ruina Hu
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Zhen Ren
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Qi Qi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Hezuo Lü
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Xiaoxin Cheng
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Jianguo Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| |
Collapse
|
12
|
Talebi M, Majdi A, Nasiri E, Naseri A, Sadigh-Eteghad S. The correlation between circulating inflammatory, oxidative stress, and neurotrophic factors level with the cognitive outcomes in multiple sclerosis patients. Neurol Sci 2020; 42:2291-2300. [DOI: 10.1007/s10072-020-04807-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/06/2020] [Indexed: 10/23/2022]
|
13
|
The Influence of Reactive Oxygen Species in the Immune System and Pathogenesis of Multiple Sclerosis. Autoimmune Dis 2020; 2020:5793817. [PMID: 32789026 PMCID: PMC7334772 DOI: 10.1155/2020/5793817] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/14/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023] Open
Abstract
Multiple roles have been indicated for reactive oxygen species (ROS) in the immune system in recent years. ROS have been extensively studied due to their ability to damage DNA and other subcellular structures. Noticeably, they have been identified as a pivotal second messenger for T-cell receptor signaling and T-cell activation and participate in antigen cross-presentation and chemotaxis. As an agent with direct toxic effects on cells, ROS lead to the initiation of the autoimmune response. Moreover, ROS levels are regulated by antioxidant systems, which include enzymatic and nonenzymatic antioxidants. Enzymatic antioxidants include superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase. Nonenzymatic antioxidants contain vitamins C, A, and E, glutathione, and thioredoxin. Particularly, cellular antioxidant systems have important functions in maintaining the redox system homeostasis. This review will discuss the significant roles of ROS generation and antioxidant systems under normal conditions, in the immune system, and pathogenesis of multiple sclerosis.
Collapse
|
14
|
Musella A, Fresegna D, Rizzo FR, Gentile A, De Vito F, Caioli S, Guadalupi L, Bruno A, Dolcetti E, Buttari F, Bullitta S, Vanni V, Centonze D, Mandolesi G. 'Prototypical' proinflammatory cytokine (IL-1) in multiple sclerosis: role in pathogenesis and therapeutic targeting. Expert Opin Ther Targets 2020; 24:37-46. [PMID: 31899994 DOI: 10.1080/14728222.2020.1709823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: It has been recognized for about 20 years that interleukin (IL)-1 signaling is implicated in Multiple Sclerosis (MS), a disabling, chronic, inflammatory and neurodegenerative disease of the central nervous system (CNS). Only recently, multifaceted roles of IL-1 emerged in MS pathophysiology as a result of both clinical and preclinical studies. Notably, drugs that directly target the IL-1 system have not been tested so far in MS.Areas covered: Recent studies in animal models, together with the development of ex vivo chimeric MS models, have disclosed a critical role for IL-1 not only at the peripheral level but also within the CNS. In the present review, we highlight the IL-1-dependent neuropathological aspects of MS, by providing an overview of the cells of the immune and CNS systems that respond to IL-1 signaling, and by emphasizing the subsequent effects on the CNS, from demyelinating processes, to synaptopathy, and excitotoxicity.Expert opinion: Drugs that act on the IL-1 system show a therapeutic potential in several autoinflammatory diseases and preclinical studies have highlighted the effects of these compounds in MS. We will discuss why anti-IL-1 therapies in MS have been neglected to date.
Collapse
Affiliation(s)
- Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele University, Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Francesca Romana Rizzo
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | - Silvia Caioli
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Antonio Bruno
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Ettore Dolcetti
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Silvia Bullitta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Diego Centonze
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy.,Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele University, Rome, Italy
| |
Collapse
|
15
|
Chakrabarti S, Jana M, Roy A, Pahan K. Upregulation of Suppressor of Cytokine Signaling 3 in Microglia by Cinnamic Acid. Curr Alzheimer Res 2019; 15:894-904. [PMID: 29732971 DOI: 10.2174/1567205015666180507104755] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/15/2018] [Accepted: 04/23/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neuroinflammation plays an important role in the pathogenesis of various neurodegenerative diseases including Alzheimer's disease (AD). Suppressor of cytokine signaling 3 (SOCS3) is an anti-inflammatory molecule that suppresses cytokine signaling and inflammatory gene expression in different cells including microglia. OBJECTIVE The pathways through which SOCS3 could be upregulated are poorly described. Cinnamic acid is a metabolite of cinnamon, a natural compound that is being widely used all over the world as a spice or flavoring agent. Here, we examined if cinnamic acid could upregulate SOCS3 in microglia. METHOD Microglia and astroglia isolated from mouse brain as well as BV-2 microglial cells were treated with cinnamic acid followed by monitoring the level of SOCS3 and different proinflammatory molecules by RT-PCR and real-time PCR. To nail down the mechanism, we also performed ChIP analysis to monitore the recruitment of cAMP response element binding (CREB) to the socs3 gene promoter and carried out siRNA knockdown of CREB. RESULTS Cinnamic acid upregulated the expression of SOCS3 mRNA and protein in mouse BV-2 microglial cells in dose- and time-dependent manner. Accordingly, cinnamic acid also increased the level of SOCS3 and suppressed the expression of inducible nitric oxide synthase and proinflammatory cytokines (TNFα, IL-1β and IL-6) in LPSstimulated BV-2 microglial cells. Similar to BV-2 microglial cells, cinnamic acid also increased the expression of SOCS3 in primary mouse microglia and astrocytes. We have seen that cAMP response element is present in the promoter of socs3 gene, that cinnamic acid induces the activation of CREB, that siRNA knockdown of CREB abrogates cinnamic acid-mediated upregulation of SOCS3, and that cinnamic acid treatment leads to the recruitment of CREB to the socs3 gene. CONCLUSIONS These studies suggest that cinnamic acid upregulates the expression of SOCS3 in glial cells via CREB pathway, which may be of importance in neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Sudipta Chakrabarti
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| | - Malabendu Jana
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| | - Avik Roy
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| |
Collapse
|
16
|
Janowska J, Sypecka J. Therapeutic Strategies for Leukodystrophic Disorders Resulting from Perinatal Asphyxia: Focus on Myelinating Oligodendrocytes. Mol Neurobiol 2018; 55:4388-4402. [PMID: 28660484 PMCID: PMC5884907 DOI: 10.1007/s12035-017-0647-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/07/2017] [Indexed: 12/12/2022]
Abstract
Perinatal asphyxia results from the action of different risk factors like complications during pregnancy, preterm delivery, or long and difficult labor. Nowadays, it is still the leading cause of neonatal brain injury known as hypoxic-ischemic encephalopathy (HIE) and resulting neurological disorders. A temporal limitation of oxygen, glucose, and trophic factors supply results in alteration of neural cell differentiation and functioning and/or leads to their death. Among the affected cells are oligodendrocytes, responsible for myelinating the central nervous system (CNS) and formation of white matter. Therefore, one of the major consequences of the experienced HIE is leukodystrophic diseases resulting from oligodendrocyte deficiency or malfunctioning. The therapeutic strategies applied after perinatal asphyxia are aimed at reducing brain damage and promoting the endogenous neuroreparative mechanisms. In this review, we focus on the biology of oligodendrocytes and discuss present clinical treatments in the context of their efficiency in preserving white matter structure and preventing cognitive and behavioral deficits after perinatal asphyxia.
Collapse
Affiliation(s)
- Justyna Janowska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego str., 02-106, Warsaw, Poland
| | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego str., 02-106, Warsaw, Poland.
| |
Collapse
|
17
|
Jana M, Ghosh S, Pahan K. Upregulation of Myelin Gene Expression by a Physically-Modified Saline via Phosphatidylinositol 3-Kinase-Mediated Activation of CREB: Implications for Multiple Sclerosis. Neurochem Res 2017; 43:407-419. [PMID: 29143164 PMCID: PMC5799355 DOI: 10.1007/s11064-017-2435-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/03/2017] [Accepted: 11/09/2017] [Indexed: 12/29/2022]
Abstract
An increase in central nervous system (CNS) remyelination and a decrease in CNS inflammation are important steps to halt the progression of multiple sclerosis (MS). RNS60 is a bioactive aqueous solution generated by subjecting normal saline to Taylor–Couette–Poiseuille flow under elevated oxygen pressure. Recently we have demonstrated that RNS60 exhibits anti-inflammatory properties. Here, we describe promyelinating property of RNS60. RNS60, but not normal saline (NS), RNS10.3 (TCP-modified saline without excess oxygen) or PNS60 (saline containing excess oxygen without TCP modification), stimulated the expression of myelin-specific genes and proteins (myelin basic protein, MBP; myelin oligodendrocyte glycoprotein, MOG and proteolipid protein, PLP) in primary mouse oligodendroglia and mixed glial cells. While investigating the mechanisms, we found that RNS60 treatment induced the activation of cAMP response element binding protein (CREB) in oligodendrocytes, ultimately leading to the recruitment of CREB to the promoters of myelin-specific genes. Furthermore, activation of type 1A p110β/α, but not type 1B p110γ, phosphatidylinositol-3 (PI-3) kinase by RNS60 together with abrogation of RNS60-mediated activation of CREB and upregulation of myelin genes by LY294002 (a specific inhibitor of PI-3 kinase) suggest that RNS60 upregulates the activation of CREB and the expression of myelin-specific molecules in oligodendrocytes via activation of PI3 kinase. These results highlight a novel promyelinating property of RNS60, which may be of benefit for MS and other demyelinating disorders.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Supurna Ghosh
- Revalesio Corporation, 1200 East D Street, Tacoma, WA, 98421, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA.
| |
Collapse
|
18
|
Murphy NP, Lampe KJ. Fabricating PLGA microparticles with high loads of the small molecule antioxidant N-acetylcysteine that rescue oligodendrocyte progenitor cells from oxidative stress. Biotechnol Bioeng 2017; 115:246-256. [PMID: 28872660 DOI: 10.1002/bit.26443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/12/2017] [Accepted: 08/28/2017] [Indexed: 01/02/2023]
Abstract
Reactive oxygen species (ROS), encompassing all oxygen radical or non-radical oxidizing agents, play key roles in disease progression. Controlled delivery of antioxidants is therapeutically relevant in such oxidant-stressed environments. Encapsulating small hydrophilic molecules into hydrophobic polymer microparticles via traditional emulsion methods has long been a challenge due to rapid mass transport of small molecules out of particle pores. We have developed a simple alteration to the existing water-in-oil-in-water (W/O/W) drug encapsulation method that dramatically improves loading efficiency: doping external water phases with drug to mitigate drug diffusion out of the particle during fabrication. PLGA microparticles with diameters ranging from 0.6 to 0.9 micrometers were fabricated, encapsulating high loads of 0.6-0.9 µm diameter PLGA microparticles were fabricated, encapsulating high loads of the antioxidant N-acetylcysteine (NAC), and released active, ROS-scavenging NAC for up to 5 weeks. Encapsulation efficiencies, normalized to the theoretical load of traditional encapsulation without doping, ranged from 96% to 400%, indicating that NAC-loaded external water phases not only prevented drug loss due to diffusion, but also doped the particles with additional drug. Antioxidant-doped particles positively affected the metabolism of oligodendrocyte progenitor cells (OPCs) under H2 O2 -mediated oxidative stress when administered both before (protection) or after (rescue) injury. Antioxidant doped particles improved outcomes of OPCs experiencing multiple doses of H2 O2 by increasing the intracellular glutathione content and preserving cellular viability relative to the injury control. Furthermore, antioxidant-doped particles preserve cell number, number of process extensions, cytoskeletal morphology, and nuclear size of H2 O2 -stressed OPCs relative to the injury control. These NAC-doped particles have the potential to provide temporally-controlled antioxidant therapy in neurodegenerative disorders such as multiple sclerosis (MS) that are characterized by continuous oxidative stress.
Collapse
Affiliation(s)
- Nicholas P Murphy
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
19
|
Mahata D, Jana M, Jana A, Mukherjee A, Mondal N, Saha T, Sen S, Nando GB, Mukhopadhyay CK, Chakraborty R, Mandal SM. Lignin-graft-Polyoxazoline Conjugated Triazole a Novel Anti-Infective Ointment to Control Persistent Inflammation. Sci Rep 2017; 7:46412. [PMID: 28401944 PMCID: PMC5401907 DOI: 10.1038/srep46412] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 03/01/2017] [Indexed: 11/26/2022] Open
Abstract
Lignin, one of the most abundant renewable feedstock, is used to develop a biocompatible hydrogel as anti-infective ointment. A hydrophilic polyoxazoline chain is grafted through ring opening polymerization, possess homogeneous spherical nanoparticles of 10-15 nm. The copolymer was covalently modified with triazole moiety to fortify the antimicrobial and antibiofilm activities. The hydrogel was capable of down regulating the expression level of IL-1β in LPS induced macrophage cells, and to cause significant reduction of iNOS production. It supported cellular anti-inflammatory activity which was confirmed with luciferase assay, western blot, and NF-κB analysis. This novel lignin-based hydrogel tested in-vivo has shown the abilities to prevent infection of burn wound, aid healing, and an anti-inflammatory dressing material. The hydrogel reported here provides a new material platform to introduce a cost-effective and efficient ointment option after undertaking further work to look at its use in the area of clinical practice.
Collapse
Affiliation(s)
- Denial Mahata
- Central Research Facility, Rubber Technology Centre, Indian Institute of Technology Kharagpur, Kharagpur 721302, WB, India
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Centre, Chicago, IL, USA
| | - Arundhuti Jana
- Department of Neurological Sciences, Rush University Medical Centre, Chicago, IL, USA
| | - Abhishek Mukherjee
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110 067, India
| | - Nibendu Mondal
- OMICS Laboratory, Department of Biotechnology, University of North Bengal, Siliguri-734 013, WB, India
| | - Tilak Saha
- OMICS Laboratory, Department of Biotechnology, University of North Bengal, Siliguri-734 013, WB, India
| | - Subhajit Sen
- OMICS Laboratory, Department of Biotechnology, University of North Bengal, Siliguri-734 013, WB, India
| | - Golok B. Nando
- Central Research Facility, Rubber Technology Centre, Indian Institute of Technology Kharagpur, Kharagpur 721302, WB, India
| | - Chinmay K. Mukhopadhyay
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110 067, India
| | - Ranadhir Chakraborty
- OMICS Laboratory, Department of Biotechnology, University of North Bengal, Siliguri-734 013, WB, India
| | - Santi M. Mandal
- Central Research Facility, Rubber Technology Centre, Indian Institute of Technology Kharagpur, Kharagpur 721302, WB, India
| |
Collapse
|
20
|
Lim JL, van der Pol SMA, Baron W, McCord JM, de Vries HE, van Horssen J. Protandim Protects Oligodendrocytes against an Oxidative Insult. Antioxidants (Basel) 2016; 5:antiox5030030. [PMID: 27618111 PMCID: PMC5039579 DOI: 10.3390/antiox5030030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/02/2016] [Indexed: 01/23/2023] Open
Abstract
Oligodendrocyte damage and loss are key features of multiple sclerosis (MS) pathology. Oligodendrocytes appear to be particularly vulnerable to reactive oxygen species (ROS) and cytokines, such as tumor necrosis factor-α (TNF), which induce cell death and prevent the differentiation of oligodendrocyte progenitor cells (OPCs). Here, we investigated the efficacy of sulforaphane (SFN), monomethyl fumarate (MMF) and Protandim to induce Nrf2-regulated antioxidant enzyme expression, and protect oligodendrocytes against ROS-induced cell death and ROS-and TNF-mediated inhibition of OPC differentiation. OLN-93 cells and primary rat oligodendrocytes were treated with SFN, MMF or Protandim resulting in significant induction of Nrf2-driven (antioxidant) proteins heme oygenase-1, nicotinamide adenine dinucleotide phosphate (NADPH): quinone oxidoreductase-1 and p62/SQSTM1, as analysed by Western blotting. After incubation with the compounds, oligodendrocytes were exposed to hydrogen peroxide. Protandim most potently promoted oligodendrocyte cell survival as measured by live/death viability assay. Moreover, OPCs were treated with Protandim or vehicle control prior to exposing them to TNF or hydrogen peroxide for five days, which inhibited OPC differentiation. Protandim significantly promoted OPC differentiation under influence of ROS, but not TNF. Protandim, a combination of five herbal ingredients, potently induces antioxidants in oligodendrocytes and is able to protect oligodendrocytes against oxidative stress by preventing ROS-induced cell death and promoting OPC differentiation.
Collapse
Affiliation(s)
- Jamie L Lim
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Neuroscience Campus Amsterdam, 1081 HZ Amsterdam, the Netherlands.
| | - Susanne M A van der Pol
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Neuroscience Campus Amsterdam, 1081 HZ Amsterdam, the Netherlands.
| | - Wia Baron
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands.
| | - Joe M McCord
- Department of Medicine, Division of Pulmonary Science and Critical Care Medicine, University of Colorado at Denver, Aurora, CO 80045, USA.
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Neuroscience Campus Amsterdam, 1081 HZ Amsterdam, the Netherlands.
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Neuroscience Campus Amsterdam, 1081 HZ Amsterdam, the Netherlands.
| |
Collapse
|
21
|
Steullet P, Cabungcal JH, Monin A, Dwir D, O'Donnell P, Cuenod M, Do KQ. Redox dysregulation, neuroinflammation, and NMDA receptor hypofunction: A "central hub" in schizophrenia pathophysiology? Schizophr Res 2016; 176:41-51. [PMID: 25000913 PMCID: PMC4282982 DOI: 10.1016/j.schres.2014.06.021] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/06/2014] [Accepted: 06/08/2014] [Indexed: 12/18/2022]
Abstract
Accumulating evidence points to altered GABAergic parvalbumin-expressing interneurons and impaired myelin/axonal integrity in schizophrenia. Both findings could be due to abnormal neurodevelopmental trajectories, affecting local neuronal networks and long-range synchrony and leading to cognitive deficits. In this review, we present data from animal models demonstrating that redox dysregulation, neuroinflammation and/or NMDAR hypofunction (as observed in patients) impairs the normal development of both parvalbumin interneurons and oligodendrocytes. These observations suggest that a dysregulation of the redox, neuroimmune, and glutamatergic systems due to genetic and early-life environmental risk factors could contribute to the anomalies of parvalbumin interneurons and white matter in schizophrenia, ultimately impacting cognition, social competence, and affective behavior via abnormal function of micro- and macrocircuits. Moreover, we propose that the redox, neuroimmune, and glutamatergic systems form a "central hub" where an imbalance within any of these "hub" systems leads to similar anomalies of parvalbumin interneurons and oligodendrocytes due to the tight and reciprocal interactions that exist among these systems. A combination of vulnerabilities for a dysregulation within more than one of these systems may be particularly deleterious. For these reasons, molecules, such as N-acetylcysteine, that possess antioxidant and anti-inflammatory properties and can also regulate glutamatergic transmission are promising tools for prevention in ultra-high risk patients or for early intervention therapy during the first stages of the disease.
Collapse
Affiliation(s)
- P Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - J H Cabungcal
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - A Monin
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - D Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - P O'Donnell
- Neuroscience Research Unit, Pfizer, Inc., 700 Main Street, Cambridge, MA 02139, USA
| | - M Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - K Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland.
| |
Collapse
|
22
|
Burm SM, Peferoen LAN, Zuiderwijk-Sick EA, Haanstra KG, 't Hart BA, van der Valk P, Amor S, Bauer J, Bajramovic JJ. Expression of IL-1β in rhesus EAE and MS lesions is mainly induced in the CNS itself. J Neuroinflammation 2016; 13:138. [PMID: 27266875 PMCID: PMC4895983 DOI: 10.1186/s12974-016-0605-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/30/2016] [Indexed: 12/21/2022] Open
Abstract
Background Interleukin (IL)-1β is a pro-inflammatory cytokine that plays a role in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), the animal model for MS. Yet, detailed studies on IL-1β expression in different stages of MS lesion development and a comparison of IL-1β expression in MS and EAE are lacking. Methods Here, we performed an extensive characterization of IL-1β expression in brain tissue of MS patients, which included different MS lesion types, and in brain tissue of rhesus macaques with EAE. Results In rhesus EAE brain tissue, we observed prominent IL-1β staining in MHC class II+ cells within perivascular infiltrates and at the edges of large demyelinating lesions. Surprisingly, staining was localized to resident microglia or differentiated macrophages rather than to infiltrating monocytes, suggesting that IL-1β expression is induced within the central nervous system (CNS). By contrast, IL-1β staining in MS brain tissue was much less pronounced. Staining was found in the parenchyma of active and chronic active MS lesions and in nodules of MHC class II+ microglia in otherwise normal appearing white matter. IL-1β expression was detected in a minority of the nodules only, which could not be distinguished by the expression of pro- and anti-inflammatory markers. These nodules were exclusively found in MS, and it remains to be determined whether IL-1β+ nodules are destined to progress into active lesions or whether they merely reflect a transient response to cellular stress. Conclusions Although the exact localization and relative intensity of IL-1β expression in EAE and MS is different, the staining pattern in both neuroinflammatory disorders is most consistent with the idea that the expression of IL-1β during lesion development is induced in the tissue rather than in the periphery. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0605-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Saskia Maria Burm
- Alternatives Unit, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | | | - Ella Alwine Zuiderwijk-Sick
- Alternatives Unit, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Krista Geraldine Haanstra
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Bert Adriaan 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Paul van der Valk
- Department of Pathology, VU Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Sandra Amor
- Department of Pathology, VU Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Jan Bauer
- Department of Neuroimmunology, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria
| | - Jeffrey John Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands.
| |
Collapse
|
23
|
Guzmán-Soto I, Salinas E, Quintanar JL. Leuprolide Acetate Inhibits Spinal Cord Inflammatory Response in Experimental Autoimmune Encephalomyelitis by Suppressing NF-κB Activation. Neuroimmunomodulation 2016; 23:33-40. [PMID: 26445405 DOI: 10.1159/000438927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/17/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Recent findings have shown that gonadotropin-releasing hormone (GnRH) administration in an animal model of multiple sclerosis (experimental autoimmune encephalomyelitis, EAE) improves clinical signs of locomotion. The present study was designed to determine whether the administration of the synthetic analog of GnRH, leuprolide acetate (LA) - besides its effects on clinical signs of locomotion - also has an effect on the activation/expression levels of molecular markers of EAE, namely transcription nuclear factor (NF)-κB and the proinflammatory cytokines IL-1β, IL-17A, IL-23 and TNF-α. METHODS EAE spinal cords were collected from control and LA-administered rats. Lumbar sections were processed at four different time points during the course of the disease to analyze NF-κB activation by chemiluminescent Western blot, and during the EAE recovery phase to evaluate proinflammatory cytokine levels by quantitative real-time PCR. RESULTS It was found that LA administration to EAE rats promoted a significant reduction of NF-κB activation during the course of the disease and also decreased the mRNA expression levels of the proinflammatory cytokines IL-1β, IL-17A and TNF-α in the EAE recovery phase; both effects are consistent with the decrease in the severity of clinical signs of locomotion induced by the treatment. CONCLUSION LA causes a reduction in the severity of locomotor activity, as well as in the activation of NF-κB and the number of proinflammatory markers in rats with EAE. These results suggest the use of this agonist as a potential therapeutic approach for multiple sclerosis.
Collapse
Affiliation(s)
- Irene Guzmán-Soto
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Centro de Ciencias Bx00E1;sicas, Universidad Autx00F3;noma de Aguascalientes, Aguascalientes, Mexico
| | | | | |
Collapse
|
24
|
Gruber RC, LaRocca D, Minchenberg SB, Christophi GP, Hudson CA, Ray AK, Shafit-Zagardo B, Massa PT. The control of reactive oxygen species production by SHP-1 in oligodendrocytes. Glia 2015; 63:1753-71. [PMID: 25919645 DOI: 10.1002/glia.22842] [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: 10/01/2014] [Accepted: 04/02/2015] [Indexed: 11/09/2022]
Abstract
We have previously described reduced myelination and corresponding myelin basic protein (MBP) expression in the central nervous system of Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) deficient motheaten (me/me) mice compared with normal littermate controls. Deficiency in myelin and MBP expression in both brains and spinal cords of motheaten mice correlated with reduced MBP mRNA expression levels in vivo and in purified oligodendrocytes in vitro. Therefore, SHP-1 activity seems to be a critical regulator of oligodendrocyte gene expression and function. Consistent with this role, this study demonstrates that oligodendrocytes of motheaten mice and SHP-1-depleted N20.1 cells produce higher levels of reactive oxygen species (ROS) and exhibit corresponding markers of increased oxidative stress. In agreement with these findings, we demonstrate that increased production of ROS coincides with ROS-induced signaling pathways known to affect myelin gene expression in oligodendrocytes. Antioxidant treatment of SHP-1-deficient oligodendrocytes reversed the pathological changes in these cells, with increased myelin protein gene expression and decreased expression of nuclear factor (erythroid-2)-related factor 2 (Nrf2) responsive gene, heme oxygenase-1 (HO-1). Furthermore, we demonstrate that SHP-1 is expressed in human white matter oligodendrocytes, and there is a subset of multiple sclerosis subjects that demonstrate a deficiency of SHP-1 in normal-appearing white matter. These studies reveal critical pathways controlled by SHP-1 in oligodendrocytes that relate to susceptibility of SHP-1-deficient mice to both developmental defects in myelination and to inflammatory demyelinating diseases.
Collapse
Affiliation(s)
- Ross C Gruber
- Department of Neurology, SUNY Upstate Medical University, Syracuse, New York.,Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Daria LaRocca
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York
| | - Scott B Minchenberg
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York
| | - George P Christophi
- Department of Neurology, SUNY Upstate Medical University, Syracuse, New York.,Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Chad A Hudson
- Department of Neurology, SUNY Upstate Medical University, Syracuse, New York.,Department of Pathology, University of Rochester, Rochester, New York
| | - Alex K Ray
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
| | | | - Paul T Massa
- Department of Neurology, SUNY Upstate Medical University, Syracuse, New York.,Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York
| |
Collapse
|
25
|
Ghosh A, Jana M, Modi K, Gonzalez FJ, Sims KB, Berry-Kravis E, Pahan K. Activation of peroxisome proliferator-activated receptor α induces lysosomal biogenesis in brain cells: implications for lysosomal storage disorders. J Biol Chem 2015; 290:10309-24. [PMID: 25750174 PMCID: PMC4400343 DOI: 10.1074/jbc.m114.610659] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 03/04/2015] [Indexed: 11/06/2022] Open
Abstract
Lysosomes are ubiquitous membrane-enclosed organelles filled with an acidic interior and are central to the autophagic, endocytic, or phagocytic pathway. In contrast to its classical function as the waste management machinery, lysosomes are now considered to be an integral part of various cellular signaling processes. The diverse functionality of this single organelle requires a very complex and coordinated regulation of its activity with transcription factor EB (TFEB), a master regulator of lysosomal biogenesis, at its core. However, mechanisms by which TFEB is regulated are poorly understood. This study demonstrates that gemfibrozil, an agonist of peroxisome proliferator-activated receptor (PPAR) α, alone and in conjunction with all-trans-retinoic acid is capable of enhancing TFEB in brain cells. We also observed that PPARα, but not PPARβ and PPARγ, is involved in gemfibrozil-mediated up-regulation of TFEB. Reporter assay and chromatin immunoprecipitation studies confirmed the recruitment of retinoid X receptor α, PPARα, and PGC1α on the PPAR-binding site on the Tfeb promoter as well. Subsequently, the drug-mediated induction of TFEB caused an increase in lysosomal protein and the lysosomal abundance in cell. Collectively, this study reinforces the link between lysosomal biogenesis and lipid metabolism with TFEB at the crossroads. Furthermore, gemfibrozil may be of therapeutic value in the treatment of lysosomal storage disorders in which autophagy-lysosome pathway plays an important role.
Collapse
Affiliation(s)
| | | | - Khushbu Modi
- From the Departments of Neurological Sciences and
| | - Frank J Gonzalez
- the Laboratory of Metabolism, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Katherine B Sims
- the Department of Neurology, Harvard Medical School, Boston, Massachusetts 02114, and
| | - Elizabeth Berry-Kravis
- Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, Illinois 60612
| | - Kalipada Pahan
- From the Departments of Neurological Sciences and the Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| |
Collapse
|
26
|
Protective effects of N-acetyl-L-cysteine in human oligodendrocyte progenitor cells and restoration of motor function in neonatal rats with hypoxic-ischemic encephalopathy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:764251. [PMID: 25918547 PMCID: PMC4396975 DOI: 10.1155/2015/764251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/15/2015] [Accepted: 03/16/2015] [Indexed: 12/29/2022]
Abstract
Objective. Since oligodendrocyte progenitor cells (OPCs) are the target cells of neonatal hypoxic-ischemic encephalopathy (HIE), the present study was aimed at investigating the protective effects of N-acetyl-l-cysteine (NAC), a well-known antioxidant and precursor of glutathione, in OPCs as well as in neonatal rats. Methods. In in vitro study, protective effects of NAC on KCN cytotoxicity in F3.Olig2 OPCs were investigated via MTT assay and apoptotic signal analysis. In in vivo study, NAC was administered to rats with HIE induced by hypoxia-ischemia surgery at postnatal day 7, and their motor functions and white matter demyelination were analyzed. Results. NAC decreased KCN cytotoxicity in F3.Olig2 cells and especially suppressed apoptosis by regulating Bcl2 and p-ERK. Administration of NAC recovered motor functions such as the using ratio of forelimb contralateral to the injured brain, locomotor activity, and rotarod performance of neonatal HIE animals. It was also confirmed that NAC attenuated demyelination in the corpus callosum, a white matter region vulnerable to HIE. Conclusion. The results indicate that NAC exerts neuroprotective effects in vitro and in vivo by preserving OPCs, via regulation of antiapoptotic signaling, and that F3.Olig2 human OPCs could be a good tool for screening of candidates for demyelinating diseases.
Collapse
|
27
|
Jana M, Mondal S, Jana A, Pahan K. Interleukin-12 (IL-12), but not IL-23, induces the expression of IL-7 in microglia and macrophages: implications for multiple sclerosis. Immunology 2014; 141:549-63. [PMID: 24224652 DOI: 10.1111/imm.12214] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 11/05/2013] [Accepted: 11/08/2013] [Indexed: 12/28/2022] Open
Abstract
Interleukin-12 (IL-12) p70 and IL-23 are bioactive cytokines and their biological functions are becoming clear. Increased expression of IL-7 in the central nervous system as well as in peripheral immune cells is associated with multiple sclerosis and experimental allergic encephalomyelitis. Here, we describe the induction of IL-7 in primary mouse and human microglia, BV-2 microglial cells, mouse peritoneal macrophages and astrocytes by IL-12p70. Interestingly, IL-12 strongly induced the expression of IL-7 whereas IL-23 and other p40 family members remained weak inducers of IL-7 in these cell types. Consistently, IL-12, but not IL-23 and other p40 family members, induced IL-7 promoter-driven luciferase activity in microglial cells. Among various stimuli tested, IL-12 emerged as the most potent stimulus followed by bacterial lipopolysaccharide and HIV-1 gp120 in inducing the activation of IL-7 promoter in microglial cells. Furthermore, increase in IL-7 mRNA expression by over-expression of IL-12p35 subunit, but not p40 and IL-23 p19 subunit, confirm that p35, but not p40 and p19, is responsible for the induction of IL-7. Finally, by using primary microglia from IL-12 receptor β1-deficient (IL-12Rβ1(-/-)) and IL-12Rβ2(-/-) mice, we demonstrate that IL-12 induces the expression of IL-7 in microglia and macrophages via both IL-12Rβ2 and IL-12Rβ1. These studies delineate a novel biological function of IL-12 that is absent in IL-23 and other p40 family members.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | | | | |
Collapse
|
28
|
Prins M, Eriksson C, Wierinckx A, Bol JGJM, Binnekade R, Tilders FJH, Van Dam AM. Interleukin-1β and interleukin-1 receptor antagonist appear in grey matter additionally to white matter lesions during experimental multiple sclerosis. PLoS One 2013; 8:e83835. [PMID: 24376764 PMCID: PMC3871572 DOI: 10.1371/journal.pone.0083835] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 11/17/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) has been mainly attributed to white matter (WM) pathology. However, recent evidence indicated the presence of grey matter (GM) lesions. One of the principal mediators of inflammatory processes is interleukin-1β (IL-1β), which is known to play a role in MS pathogenesis. It is unknown whether IL-1β is solely present in WM or also in GM lesions. Using an experimental MS model, we questioned whether IL-1β and the IL-1 receptor antagonist (IL-1ra) are present in GM in addition to affected WM regions. METHODS The expression of IL-1β and IL-1ra in chronic-relapsing EAE (cr-EAE) rats was examined using in situ hybridization, immunohistochemistry and real-time PCR. Rats were sacrificed at the peak of the first disease phase, the trough of the remission phase, and at the peak of the relapse. Histopathological characteristics of CNS lesions were studied using immunohistochemistry for PLP, CD68 and CD3 and Oil-Red O histochemistry. RESULTS IL-1β and IL-ra expression appears to a similar extent in affected GM and WM regions in the brain and spinal cord of cr-EAE rats, particularly in perivascular and periventricular locations. IL-1β and IL-1ra expression was dedicated to macrophages and/or activated microglial cells, at sites of starting demyelination. The time-dependent expression of IL-1β and IL-1ra revealed that within the spinal cord IL-1β and IL-1ra mRNA remained present throughout the disease, whereas in the brain their expression disappeared during the relapse. CONCLUSIONS The appearance of IL-1β expressing cells in GM within the CNS during cr-EAE may explain the occurrence of several clinical deficits present in EAE and MS which cannot be attributed solely to the presence of IL-1β in WM. Endogenously produced IL-1ra seems not capable to counteract IL-1β-induced effects. We put forward that IL-1β may behold promise as a target to address GM, in addition to WM, related pathology in MS.
Collapse
Affiliation(s)
- Marloes Prins
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Charlotta Eriksson
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Anne Wierinckx
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
- UNIV UMR1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - John G. J. M. Bol
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Rob Binnekade
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Fred J. H. Tilders
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Anne-Marie Van Dam
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Jana M, Pahan K. Down-regulation of Myelin Gene Expression in Human Oligodendrocytes by Nitric Oxide: Implications for Demyelination in Multiple Sclerosis. ACTA ACUST UNITED AC 2013; 4. [PMID: 24273691 DOI: 10.4172/2155-9899.1000157] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disorder of the central nervous system (CNS) with unknown etiology. Several studies have shown that demyelination in MS is caused by proinflammatory mediators and nitric oxide (NO), which is released by perivascular infiltrates and/or activated glial cells. Both endogenous NO released by microglia and astrocytes; and NO generated from exogenous NO donors are known to induce oligodendrocytes death. However, the molecular mechanism of oligodendroglial death is poorly understood. Here we explore the role of NO in modulating the expression of myelin-specific genes that leads to oligodendroglial death. We investigated the effect of NO on the expression of myelin basic protein (MBP), 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase), myelin oligodendrocyte glycoprotein (MOG), and proteolipid protein (PLP) in human primary oligodendrocytes. Combination of IFN-γ and bacterial lipopolysaccharide (LPS) or double stranded RNA in the form of polyIC induced the production of NO and decreased the expression of myelin gene in human fetal mixed glial cultures. Either a scavenger of NO (PTIO) or an inhibitor of inducible nitric oxide synthase (L-NIL) abrogated (LPS+IFN-γ)- and polyIC-mediated suppression of myelin genes in human mixed glial cells. The role of NO was further corroborated by the inhibition of myelin gene expression in purified human oligodendroglia by several NO donors including SNP, NOC-7, SIN-1, and SNAP. This study illustrates a novel biological role of NO in down-regulating the expression of myelin genes preceding the death of oligodendrocytes.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | |
Collapse
|
30
|
Ghosh A, Corbett GT, Gonzalez FJ, Pahan K. Gemfibrozil and fenofibrate, Food and Drug Administration-approved lipid-lowering drugs, up-regulate tripeptidyl-peptidase 1 in brain cells via peroxisome proliferator-activated receptor α: implications for late infantile Batten disease therapy. J Biol Chem 2012; 287:38922-35. [PMID: 22989886 DOI: 10.1074/jbc.m112.365148] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The classical late infantile neuronal ceroid lipofuscinosis (LINCLs) is an autosomal recessive disease, where the defective gene is Cln2, encoding tripeptidyl-peptidase I (TPP1). At the molecular level, LINCL is caused by accumulation of autofluorescent storage materials in neurons and other cell types. Currently, there is no established treatment for this fatal disease. This study reveals a novel use of gemfibrozil and fenofibrate, Food and Drug Administration-approved lipid-lowering drugs, in up-regulating TPP1 in brain cells. Both gemfibrozil and fenofibrate up-regulated mRNA, protein, and enzymatic activity of TPP1 in primary mouse neurons and astrocytes as well as human astrocytes and neuronal cells. Because gemfibrozil and fenofibrate are known to activate peroxisome proliferator-activated receptor-α (PPARα), the role of PPARα in gemfibrozil- and fenofibrate-mediated up-regulation of TPP1 was investigated revealing that both drugs up-regulated TPP1 mRNA, protein, and enzymatic activity both in vitro and in vivo in wild type (WT) and PPARβ(-/-), but not PPARα(-/-), mice. In an attempt to delineate the mechanism of TPP1 up-regulation, it was found that the effects of the fibrate drugs were abrogated in the absence of retinoid X receptor-α (RXRα), a molecule known to form a heterodimer with PPARα. Accordingly, all-trans-retinoic acid, alone or together with gemfibrozil, up-regulated TPP1. Co-immunoprecipitation and ChIP studies revealed the formation of a PPARα/RXRα heterodimer and binding of the heterodimer to an RXR-binding site on the Cln2 promoter. Together, this study demonstrates a unique mechanism for the up-regulation of TPP1 by fibrate drugs via PPARα/RXRα pathway.
Collapse
Affiliation(s)
- Arunava Ghosh
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
31
|
Jana M, Mondal S, Gonzalez FJ, Pahan K. Gemfibrozil, a lipid-lowering drug, increases myelin genes in human oligodendrocytes via peroxisome proliferator-activated receptor-β. J Biol Chem 2012; 287:34134-48. [PMID: 22879602 DOI: 10.1074/jbc.m112.398552] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
An increase in CNS remyelination and a decrease in CNS inflammation are important steps to halt the progression of multiple sclerosis. Earlier studies have shown that gemfibrozil, a lipid-lowering drug, has anti-inflammatory properties. The current study identified another novel property of gemfibrozil in stimulating the expression of myelin-specific genes (myelin basic protein, myelin oligodendrocyte glycoprotein, 2',3'-cyclic-nucleotide 3'-phosphodiesterase, and proteolipid protein (PLP)) in primary human oligodendrocytes, mixed glial cells, and spinal cord organotypic cultures. Although gemfibrozil is a known activator of peroxisome proliferator-activated receptor-α (PPAR-α), we were unable to detect PPAR-α in either gemfibrozil-treated or untreated human oligodendrocytes, and gemfibrozil increased the expression of myelin genes in oligodendrocytes isolated from both wild type and PPAR-α(-/-) mice. On the other hand, gemfibrozil markedly increased the expression of PPAR-β but not PPAR-γ. Consistently, antisense knockdown of PPAR-β, but not PPAR-γ, abrogated the stimulatory effect of gemfibrozil on myelin genes in human oligodendrocytes. Gemfibrozil also did not up-regulate myelin genes in oligodendroglia isolated from PPAR-β(-/-) mice. Chromatin immunoprecipitation analysis showed that gemfibrozil induced the recruitment of PPAR-β to the promoter of PLP and myelin oligodendrocyte glycoprotein genes in human oligodendrocytes. Furthermore, gemfibrozil treatment also led to the recruitment of PPAR-β to the PLP promoter in vivo in the spinal cord of experimental autoimmune encephalomyelitis mice and suppression of experimental autoimmune encephalomyelitis symptoms in PLP-T cell receptor transgenic mice. These results suggest that gemfibrozil stimulates the expression of myelin genes via PPAR-β and that gemfibrozil, a prescribed drug for humans, may find further therapeutic use in demyelinating diseases.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
32
|
Ljubisavljevic S, Stojanovic I, Pavlovic R, Sokolovic D, Pavlovic D, Cvetkovic T, Stevanovic I. Modulation of nitric oxide synthase by arginase and methylated arginines during the acute phase of experimental multiple sclerosis. J Neurol Sci 2012; 318:106-11. [DOI: 10.1016/j.jns.2012.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 10/28/2022]
|
33
|
Corbett GT, Roy A, Pahan K. Gemfibrozil, a lipid-lowering drug, upregulates IL-1 receptor antagonist in mouse cortical neurons: implications for neuronal self-defense. THE JOURNAL OF IMMUNOLOGY 2012; 189:1002-13. [PMID: 22706077 DOI: 10.4049/jimmunol.1102624] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chronic inflammation is becoming a hallmark of several neurodegenerative disorders and accordingly, IL-1β, a proinflammatory cytokine, is implicated in the pathogenesis of neurodegenerative diseases. Although IL-1β binds to its high-affinity receptor, IL-1R, and upregulates proinflammatory signaling pathways, IL-1R antagonist (IL-1Ra) adheres to the same receptor and inhibits proinflammatory cell signaling. Therefore, upregulation of IL-1Ra is considered important in attenuating inflammation. The present study underlines a novel application of gemfibrozil (gem), a Food and Drug Administration-approved lipid-lowering drug, in increasing the expression of IL-1Ra in primary mouse and human neurons. Gem alone induced an early and pronounced increase in the expression of IL-1Ra in primary mouse cortical neurons. Activation of type IA p110α PI3K and Akt by gem and abrogation of gem-induced upregulation of IL-1Ra by inhibitors of PI3K and Akt indicate a role of the PI3K-Akt pathway in the upregulation of IL-1Ra. Gem also induced the activation of CREB via the PI3K-Akt pathway, and small interfering RNA attenuation of CREB abolished the gem-mediated increase in IL-1Ra. Furthermore, gem was able to protect neurons from IL-1β insult. However, small interfering RNA knockdown of neuronal IL-1Ra abrogated the protective effect of gem against IL-1β, suggesting that this drug increases the defense mechanism of cortical neurons via upregulation of IL-1Ra. Taken together, these results highlight the importance of the PI3K-Akt-CREB pathway in mediating gem-induced upregulation of IL-1Ra in neurons and suggest gem as a possible therapeutic treatment for propagating neuronal self-defense in neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Grant T Corbett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | |
Collapse
|
34
|
Ghosh A, Pahan K. Gemfibrozil, a lipid-lowering drug, induces suppressor of cytokine signaling 3 in glial cells: implications for neurodegenerative disorders. J Biol Chem 2012; 287:27189-203. [PMID: 22685291 DOI: 10.1074/jbc.m112.346932] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glial inflammation is an important feature of several neurodegenerative disorders. Suppressor of cytokine signaling (SOCS) proteins play a crucial role in inhibiting cytokine signaling and inflammatory gene expression in various cell types, including glial cells. However, mechanisms by which SOCS genes could be up-regulated are poorly understood. This study underlines the importance of gemfibrozil, a Food and Drug Administration-approved lipid-lowering drug, in up-regulating the expression of SOCS3 in glial cells. Gemfibrozil increased the expression of Socs3 mRNA and protein in mouse astroglia and microglia in both a time- and dose-dependent manner. Interestingly, gemfibrozil induced the activation of type IA phosphatidylinositol (PI) 3-kinase and AKT. Accordingly, inhibition of PI 3-kinase and AKT by chemical inhibitors abrogated gemfibrozil-mediated up-regulation of SOCS3. Furthermore, we demonstrated that gemfibrozil induced the activation of Krüppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil also induced the recruitment of KLF4 to the distal, but not proximal, KLF4-binding site of the Socs3 promoter. This study delineates a novel property of gemfibrozil in up-regulating SOCS3 in glial cells via PI 3-kinase-AKT-mediated activation of KLF4 and suggests that gemfibrozil may find therapeutic application in neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Arunava Ghosh
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | |
Collapse
|
35
|
Kim DY, Hao J, Liu R, Turner G, Shi FD, Rho JM. Inflammation-mediated memory dysfunction and effects of a ketogenic diet in a murine model of multiple sclerosis. PLoS One 2012; 7:e35476. [PMID: 22567104 PMCID: PMC3342287 DOI: 10.1371/journal.pone.0035476] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 03/18/2012] [Indexed: 11/18/2022] Open
Abstract
A prominent clinical symptom in multiple sclerosis (MS), a progressive disorder of the central nervous system (CNS) due to heightened neuro-inflammation, is learning and memory dysfunction. Here, we investigated the effects of a ketogenic diet (KD) on memory impairment and CNS-inflammation in a murine model of experimental autoimmune encephalomyelitis (EAE), using electrophysiological, behavioral, biochemical and in vivo imaging approaches. Behavioral spatial learning deficits were associated with motor disability in EAE mice, and were observed concurrently with brain inflammation. The KD improved motor disability in the EAE model, as well as CA1 hippocampal synaptic plasticity (long-term potentiation) and spatial learning and memory (assessed with the Morris Water Maze). Moreover, hippocampal atrophy and periventricular lesions in EAE mice were reversed in KD-treated EAE mice. Finally, we found that the increased expression of inflammatory cytokines and chemokines, as well as the production of reactive oxygen species (ROS), in our EAE model were both suppressed by the KD. Collectively, our findings indicate that brain inflammation in EAE mice is associated with impaired spatial learning and memory function, and that KD treatment can exert protective effects, likely via attenuation of the robust immune response and increased oxidative stress seen in these animals.
Collapse
Affiliation(s)
- Do Young Kim
- Barrow Neurological Institute, Medical Center, St. Joseph’s Hospital, Phoenix, Arizona, United States of America
| | - Junwei Hao
- Barrow Neurological Institute, Medical Center, St. Joseph’s Hospital, Phoenix, Arizona, United States of America
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruolan Liu
- Barrow Neurological Institute, Medical Center, St. Joseph’s Hospital, Phoenix, Arizona, United States of America
| | - Gregory Turner
- Keller Center for Imaging Innovation, Barrow Neurological Institute, Phoenix, Arizona, United States of America
| | - Fu-Dong Shi
- Barrow Neurological Institute, Medical Center, St. Joseph’s Hospital, Phoenix, Arizona, United States of America
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- * E-mail: (JMR); (FDS)
| | - Jong M. Rho
- Barrow Neurological Institute, Medical Center, St. Joseph’s Hospital, Phoenix, Arizona, United States of America
- Departments of Pediatrics and Clinical Neurosciences, University of Calgary, Alberta Children’s Hospital, Calgary, Alberta, Canada
- * E-mail: (JMR); (FDS)
| |
Collapse
|
36
|
Jana M, Pahan K. Gemfibrozil, a lipid lowering drug, inhibits the activation of primary human microglia via peroxisome proliferator-activated receptor β. Neurochem Res 2012; 37:1718-29. [PMID: 22528839 DOI: 10.1007/s11064-012-0781-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 04/04/2012] [Accepted: 04/07/2012] [Indexed: 12/27/2022]
Abstract
Microglial activation participates in the pathogenesis of various neuroinflammatory and neurodegenerative diseases. However, mechanisms by which microglial activation could be controlled are poorly understood. Peroxisome proliferator-activated receptors (PPAR) are transcription factors belonging to the nuclear receptor super family with diverse effect. This study underlines the importance of PPARβ/δ in mediating the anti-inflammatory effect of gemfibrozil, an FDA-approved lipid-lowering drug, in primary human microglia. Bacterial lipopolysachharides (LPS) induced the expression of various proinflammatory molecules and upregulated the expression of microglial surface marker CD11b in human microglia. However, gemfibrozil markedly suppressed proinflammatory molecules and CD11b in LPS-stimulated microglia. Human microglia expressed PPAR-β and -γ, but not PPAR-α. Interestingly, either antisense knockdown of PPAR-β or antagonism of PPAR-β by a specific chemical antagonist abrogated gemfibrozil-mediated inhibition of microglial activation. On the other hand, blocking of PPAR-α and -γ had no effect on gemfibrozil-mediated anti-inflammatory effect in microglia. These results highlight the fact that gemfibrozil regulates microglial activation by inhibiting inflammatory gene expression in a PPAR-β dependent pathway and further reinforce its therapeutic application in several neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St., Suite 320, Chicago, IL 60612, USA
| | | |
Collapse
|
37
|
Volpe JJ. Systemic inflammation, oligodendroglial maturation, and the encephalopathy of prematurity. Ann Neurol 2012; 70:525-9. [PMID: 22028217 DOI: 10.1002/ana.22533] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Ljubisavljevic S, Stojanovic I, Pavlovic D, Sokolovic D, Stevanovic I. Aminoguanidine and N-acetyl-cysteine supress oxidative and nitrosative stress in EAE rat brains. Redox Rep 2011; 16:166-72. [PMID: 21888767 DOI: 10.1179/1351000211y.0000000007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a well-established animal model of human multiple sclerosis (MS). We have evaluated the role of oxidative and nitrosative stress, as the causal factors in the development of EAE, responsible for the damage of cardinal cellular components, such as lipids, proteins and nucleic acids, resulting in demyelination, axonal damage, and neuronal death. EAE was induced in female Sprague-Dawley rats, 3 months old (300±20 g), by immunization with myelin basic protein in combination with Complete Freund's adjuvant (CFA). The animals were divided into seven groups: control, EAE, CFA, EAE+aminoguanidine (AG), AG, EAE+N-acetyl-L-cysteine (NAC) and NAC. The animals were sacrificed 15 days after EAE induction, and the levels of nitrosative and oxidative stress were determined in 10% homogenate of the whole encephalitic mass. In EAE rats, brain NO production and MDA level were significantly increased (P<0.001) compared to the control values, whereas AG and NAC treatment decreased both parameters in EAE rats compared to EAE group (P<0.001). Glutathione (GSH) was reduced (P<0.001) in EAE rats in comparison with the control and CFA groups, but increased in EAE+AG and EAE+NAC group compared to the EAE group (P<0.01). Superoxide dismutase (SOD) activity was significantly decreased (P<0.001) in the EAE group compared to all other experimental groups. The clinical expression of EAE was significantly decreased (P<0.05) in the EAE groups treated with AG and NAC compared to EAE rats, during disease development. The obtained results prove an important role of oxidative and nitrosative stress in the pathogenesis of EAE, whereas AG and NAC protective effects offer new possibilities for a modified combined approach in MS therapy.
Collapse
|
39
|
Gankam Kengne F, Nicaise C, Soupart A, Boom A, Schiettecatte J, Pochet R, Brion JP, Decaux G. Astrocytes are an early target in osmotic demyelination syndrome. J Am Soc Nephrol 2011; 22:1834-45. [PMID: 21885671 DOI: 10.1681/asn.2010111127] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abrupt osmotic changes during rapid correction of chronic hyponatremia result in demyelinative brain lesions, but the sequence of events linking rapid osmotic changes to myelin loss is not yet understood. Here, in a rat model of osmotic demyelination syndrome, we found that massive astrocyte death occurred after rapid correction of hyponatremia, delineating the regions of future myelin loss. Astrocyte death caused a disruption of the astrocyte-oligodendrocyte network, rapidly upregulated inflammatory cytokines genes, and increased serum S100B, which predicted clinical manifestations and outcome of osmotic demyelination. These results support a model for the pathophysiology of osmotic brain injury in which rapid correction of hyponatremia triggers apoptosis in astrocytes followed by a loss of trophic communication between astrocytes and oligodendrocytes, secondary inflammation, microglial activation, and finally demyelination.
Collapse
Affiliation(s)
- Fabrice Gankam Kengne
- Erasme University Hospital, Department of General Internal Medicine, Research Unit on Hydromineral Metabolism, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Paintlia MK, Paintlia AS, Singh AK, Singh I. Synergistic activity of interleukin-17 and tumor necrosis factor-α enhances oxidative stress-mediated oligodendrocyte apoptosis. J Neurochem 2011; 116:508-21. [PMID: 21143599 DOI: 10.1111/j.1471-4159.2010.07136.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Th1 cytokine-induced loss of oligodendrocytes (OLs) is associated with axonal loss in CNS demyelinating diseases such as multiple sclerosis (MS)that contributes to neurological disabilities in affected individuals. Recent studies indicated that, in addition to Th1-phenotype cytokines including tumor necrosis factor (TNF)-α, Th17 phenotype cytokine, interleukin (IL)-17 also involved in the development of MS. In this study, we investigated the direct effect of IL-17 on the survival of OLs in the presence of TNF-α and individually in vitro settings. Our findings suggest that IL-17 alone, however, was not able to affect the survival of OLs, but it exacerbates the TNF-α-induced OL apoptosis as compared with individual TNF-α treatment. This effect of cytokines was ascribed to an inhibition of cell-survival mechanisms, co-localization of Bid/Bax proteins in the mitochondrial membrane and caspase 8 activation mediated release of apoptosis inducing factor from mitochondria in treated OLs. In addition, cytokine treatment disturbed the mitochondrial membrane potential in OLs with corresponding increase in the generation of reactive oxygen species, which were attenuated by N-acetyl cysteine treatment. In addition, combining of these cytokines induced cell-cycle arrest at G1/S phases in OL-like cells and inhibited the maturation of OL progenitor cells that was attenuated by peroxisome proliferator-activated receptor-γ/-β agonists. Collectively, these data provide initial evidence that IL-17 exacerbates TNF-α-induced OL loss and inhibits the differentiation of OL progenitor cells suggesting that antioxidant- or peroxisome proliferator-activated receptor agonist-based therapies have potential to limit CNS demyelination in MS or other related demyelinating disorders.
Collapse
Affiliation(s)
- Manjeet K Paintlia
- Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
41
|
Fibrillar amyloid-beta-activated human astroglia kill primary human neurons via neutral sphingomyelinase: implications for Alzheimer's disease. J Neurosci 2010; 30:12676-89. [PMID: 20861373 DOI: 10.1523/jneurosci.1243-10.2010] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Glial activation plays an important role in the pathogenesis of various neurodegenerative disorders including Alzheimer's disease. However, molecular mechanisms by which activated glia could kill neurons are poorly understood. The present study underlines the importance of neutral sphingomyelinase (N-SMase) in mediating the damaging effect of fibrillar amyloid-β 1-42 (Aβ1-42) peptide-activated astroglia on neurons. In transwell experiments, soluble products released from activated primary human astroglia induced the activation of neutral sphingomyelinase (N-SMase), production of ceramide, and cell death in primary human neurons. Protection of neurons from cytotoxic effects of activated astroglia by antisense knockdown of N-SMase, but not acidic sphingomyelinase (A-SMase), suggests that soluble products released from activated astroglia kill neurons via N-SMase but not A-SMase. Next we examined the role of N-SMase in the activation of human astroglia. Interestingly, knockdown of N-SMase, but not A-SMase, by either antisense oligonucleotides or chemical inhibitor, prevented the induction of proinflammatory molecules [tumor necrosis factor-α, inducible nitric oxide synthase, interleukin-1β (IL-1β), and IL-6] and the activation of nuclear factor-κB in Aβ1-42-activated astroglia. Subsequently, fibrillar Aβ peptides also induced the activation of N-SMase and ceramide in vivo in mouse cortex. Most importantly, antisense knockdown of N-SMase, but not A-SMase, decreased the activation of astroglia and protected neurons from fibrillar Aβ toxicity in vivo in the cortex. Together, it is apparent that both the activation of astroglia by Aβ and that the cytotoxicity of activated astroglia on neurons depend on N-SMase.
Collapse
|
42
|
Jana A, Pahan K. Sphingolipids in multiple sclerosis. Neuromolecular Med 2010; 12:351-61. [PMID: 20607622 DOI: 10.1007/s12017-010-8128-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2010] [Accepted: 06/26/2010] [Indexed: 02/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the CNS. Oligodendrocytes, the myelin-forming cells of the central nervous system (CNS), are target cells in MS. Although the etiology of MS is poorly known, new insights suggest oligodendrocyte apoptosis as one of the critical events followed by glial activation and infiltration of lymphocytes and macrophages. A major breakthrough in delineation of the mechanism of cell death, perivascular cuffing, and glial activation came from elucidation of the sphingolipid signal transduction pathway. The sphingolipid signal transduction pathway induces apoptosis, differentiation, proliferation, and growth arrest depending upon cell and receptor types, and downstream targets. Sphingomyelin, a major component of myelin membrane formed by mature oligodendrocytes, is abundant in the CNS and ceramide, its primary catabolic product released by activation of either neutral or acidic sphingomyelinase, serves as a potential lipid second messenger or mediator molecule modulating diverse cellular signaling pathways. Similarly, under certain conditions, sphingosine produced from ceramide by ceramidase is phosphorylated by sphingosine kinases to sphingosine-1 phosphate, another potent second messenger molecule. Both ceramide and sphingosine-1 phosphate regulate life and death of many cell types including brain cells and participate in pathogenic processes of MS. In this review, we have made an honest attempt to compile recent findings made by others and us relating to the role of sphingolipids in the disease process of MS.
Collapse
Affiliation(s)
- Arundhati Jana
- Department of Neurological Sciences, Rush University Medical Center, Cohn Research Building, Suite 320, 1735 West Harrison St., Chicago, IL 60612, USA
| | | |
Collapse
|
43
|
Jana M, Pahan K. IL-12 p40 homodimer, but not IL-12 p70, induces the expression of IL-16 in microglia and macrophages. Mol Immunol 2008; 46:773-83. [PMID: 19100623 DOI: 10.1016/j.molimm.2008.10.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 10/21/2008] [Accepted: 10/22/2008] [Indexed: 12/19/2022]
Abstract
IL-16, a leukocyte chemoattractant factor (LCF), is involved in the disease process of multiple sclerosis and other autoimmune disorders. However, mechanisms by which this LCF is expressed are poorly understood. The present study underlines the importance of IL-12 p40 homodimer (p40(2)), the so-called biologically inactive molecule, in inducing the expression of IL-16 in primary mouse and human microglia, mouse BV-2 microglial cells, mouse peritoneal macrophages, and RAW264.7 cells. In contrast, IL-12 p70, the bioactive heterodimeric cytokine, was unable to induce the expression of IL-16 in any of these cell types. Similarly IL-12 p40(2) also induced the activation of IL-16 promoter in microglia. Among various stimuli tested, p40(2) was the most potent one followed by p40 monomer, IL-16 and IL-23 in inducing the activation of IL-16 promoter in microglial cells. Furthermore, induction of IL-16 mRNA expression by over-expression of p40, but not p35, cDNA and induction of IL-16 expression by p40(2) in microglia isolated from IL-12p35 (-/-) mice confirm that p40, but not p35, is responsible for the induction of IL-16. Finally, by using primary microglia isolated from IL-12Rbeta1 (-/-) and IL-12Rbeta2 (-/-) mice, we demonstrate that p40(2) induces the expression of this LCF via IL-12Rbeta1 but not IL-12Rbeta2. These results delineate a novel biological function of p40(2) and raise the possibility that biological function of IL-12 p40(2) may be different from IL-12 p70.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Cohn Research Building, Suite 320, 1735 West Harrison St., Chicago, IL 60612, United States
| | | |
Collapse
|
44
|
Jana M, Palencia CA, Pahan K. Fibrillar amyloid-beta peptides activate microglia via TLR2: implications for Alzheimer's disease. THE JOURNAL OF IMMUNOLOGY 2008; 181:7254-62. [PMID: 18981147 DOI: 10.4049/jimmunol.181.10.7254] [Citation(s) in RCA: 249] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Microglial activation is an important pathological component in brains of patients with Alzheimer's disease (AD), and fibrillar amyloid-beta (Abeta) peptides play an important role in microglial activation in AD. However, mechanisms by which Abeta peptides induce the activation of microglia are poorly understood. The present study underlines the importance of TLR2 in mediating Abeta peptide-induced activation of microglia. Fibrillar Abeta1-42 peptides induced the expression of inducible NO synthase, proinflammatory cytokines (TNF-alpha, IL-1beta, and IL-6), and integrin markers (CD11b, CD11c, and CD68) in mouse primary microglia and BV-2 microglial cells. However, either antisense knockdown of TLR2 or functional blocking Abs against TLR2 suppressed Abeta1-42-induced expression of proinflammatory molecules and integrin markers in microglia. Abeta1-42 peptides were also unable to induce the expression of proinflammatory molecules and increase the expression of CD11b in microglia isolated from TLR2(-/-) mice. Finally, the inability of Abeta1-42 peptides to induce the expression of inducible NO synthase and to stimulate the expression of CD11b in vivo in the cortex of TLR2(-/-) mice highlights the importance of TLR2 in Abeta-induced microglial activation. In addition, ligation of TLR2 alone was also sufficient to induce microglial activation. Consistent to the importance of MyD88 in mediating the function of various TLRs, antisense knockdown of MyD88 also inhibited Abeta1-42 peptide-induced expression of proinflammatory molecules. Taken together, these studies delineate a novel role of TLR2 signaling pathway in mediating fibrillar Abeta peptide-induced activation of microglia.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | |
Collapse
|
45
|
Jana M, Pahan K. Induction of lymphotoxin-alpha by interleukin-12 p40 homodimer, the so-called biologically inactive molecule, but not IL-12 p70. Immunology 2008; 127:312-25. [PMID: 19019087 DOI: 10.1111/j.1365-2567.2008.02985.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Interleukin-12 (IL-12) p70 (p40:p35) is a bioactive cytokine and its biological functions are becoming clear. On the other hand, the IL-12 p40 homodimer (p40(2)) was considered an inactive or inhibitory molecule and its functions are poorly understood. It has been reported that increased expression of lymphotoxin-alpha (Lt-alpha) in the central nervous system as well as in peripheral immune cells is associated with multiple sclerosis and experimental allergic encephalomyelitis. Here we describe that p40(2) induces the expression of Lt-alpha in primary mouse and human microglia, BV-2 microglial cells, splenic macrophages, RAW 264.7 cells and splenic T cells. Interestingly, IL-12 p70 was either unable to induce Lt-alpha or was a very weak inducer of Lt-alpha in these cell types. Consistently, p40(2), but not p70, induced Lt-alpha promoter-driven luciferase activity in microglial cells. Among various stimuli tested, p40(2) emerged as the most potent followed by IL-16, lipopolyaccharide and double-stranded RNA in inducing the activation of Lt-alpha promoter in microglial cells. Furthermore, an increase in Lt-alpha messenger RNA expression by overexpression of p40, but not p35, complementary DNA and induction of Lt-alpha expression by p40(2) in microglia isolated from IL-12p35(-/-) mice confirm that p40, but not p35, is responsible for the induction of Lt-alpha. Finally, by using primary microglia from IUL-12 receptor beta1 deficient (IL-12Rbeta1(-/-)) and IL-12Rbeta2(-/-) mice, we demonstrate that p40(2) induced the expression of Lt-alpha in microglia and macrophages via IL-12Rbeta1, but not IL-12Rbeta2. These studies delineate a novel biological function of p40(2) that is absent in IL-12.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA.
| | | |
Collapse
|
46
|
Jana M, Jana A, Liu X, Ghosh S, Pahan K. Involvement of phosphatidylinositol 3-kinase-mediated up-regulation of I kappa B alpha in anti-inflammatory effect of gemfibrozil in microglia. THE JOURNAL OF IMMUNOLOGY 2007; 179:4142-52. [PMID: 17785853 PMCID: PMC2604815 DOI: 10.4049/jimmunol.179.6.4142] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The present study underlines the importance of PI3K in mediating the anti-inflammatory effect of gemfibrozil, a prescribed lipid-lowering drug for humans, in mouse microglia. Gemfibrozil inhibited LPS-induced expression of inducible NO synthase (iNOS) and proinflammatory cytokines in mouse BV-2 microglial cells and primary microglia. By overexpressing wild-type and dominant-negative constructs of peroxisome proliferator-activated receptor-alpha (PPAR-alpha) in microglial cells and isolating primary microglia from PPAR-alpha-/- mice, we have demonstrated that gemfibrozil inhibits the activation of microglia independent of PPAR-alpha. Interestingly, gemfibrozil induced the activation of p85alpha-associated PI3K (p110beta but not p110alpha) and inhibition of that PI3K by either chemical inhibitors or dominant-negative mutants abrogated the inhibitory effect of gemfibrozil. Conversely, overexpression of the constitutively active mutant of p110 enhanced the inhibitory effect of gemfibrozil on LPS-induced expression of proinflammatory molecules. Similarly, gemfibrozil also inhibited fibrillar amyloid beta (Abeta)-, prion peptide (PrP)-, dsRNA (poly IC)-, HIV-1 Tat-, and 1-methyl-4-phenylpyridinium (MPP+)-, but not IFN-gamma-, induced microglial expression of iNOS. Inhibition of PI3K also abolished the inhibitory effect of gemfibrozil on Abeta-, PrP-, poly IC-, Tat-, and MPP+-induced microglial expression of iNOS. Involvement of NF-kappaB activation in LPS-, Abeta-, PrP-, poly IC-, Tat-, and MPP+-, but not IFN-gamma-, induced microglial expression of iNOS and stimulation of IkappaBalpha expression and inhibition of NF-kappaB activation by gemfibrozil via the PI3K pathway suggests that gemfibrozil inhibits the activation of NF-kappaB and the expression of proinflammatory molecules in microglia via PI3K-mediated up-regulation of IkappaBalpha.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Arundhati Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Xiaojuan Liu
- Section of Neuroscience, Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, NE 68583
| | - Sankar Ghosh
- Section of Immunobiology and Department of Molecular Biophysics and Biochemistry, School of Medicine, Yale University, New Haven, CT 06536
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
- Section of Neuroscience, Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, NE 68583
- Address correspondence and reprint requests to Dr. Kalipada Pahan, Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Suite 320, Chicago, IL 60612. E-mail address:
| |
Collapse
|
47
|
Antony JM, Ellestad KK, Hammond R, Imaizumi K, Mallet F, Warren KG, Power C. The human endogenous retrovirus envelope glycoprotein, syncytin-1, regulates neuroinflammation and its receptor expression in multiple sclerosis: a role for endoplasmic reticulum chaperones in astrocytes. THE JOURNAL OF IMMUNOLOGY 2007; 179:1210-24. [PMID: 17617614 DOI: 10.4049/jimmunol.179.2.1210] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Retroviral envelopes are pathogenic glycoproteins which cause neuroinflammation, neurodegeneration, and endoplasmic reticulum stress responses. The human endogenous retrovirus (HERV-W) envelope protein, Syncytin-1, is highly expressed in CNS glia of individuals with multiple sclerosis (MS). In this study, we investigated the mechanisms by which Syncytin-1 mediated neuroimmune activation and oligodendrocytes damage. In brain tissue from individuals with MS, ASCT1, a receptor for Syncytin-1 and a neutral amino acid transporter, was selectively suppressed in astrocytes (p < 0.05). Syncytin-1 induced the expression of the endoplasmic reticulum stress sensor, old astrocyte specifically induced substance (OASIS), in cultured astrocytes, similar to findings in MS brains. Overexpression of OASIS in astrocytes increased inducible NO synthase expression but concurrently down-regulated ASCT1 (p < 0.01). Treatment of astrocytes with a NO donor enhanced expression of early growth response 1, with an ensuing reduction in ASCT1 expression (p < 0.05). Small-interfering RNA molecules targeting Syncytin-1 selectively down-regulated its expression, preventing the suppression of ASCT1 and the release of oligodendrocyte cytotoxins by astrocytes. A Syncytin-1-transgenic mouse expressing Syncytin-1 under the glial fibrillary acidic protein promoter demonstrated neuroinflammation, ASCT1 suppression, and diminished levels of myelin proteins in the corpus callosum, consistent with observations in CNS tissues from MS patients together with neurobehavioral abnormalities compared with wild-type littermates (p < 0.05). Thus, Syncytin-1 initiated an OASIS-mediated suppression of ASCT1 in astrocytes through the induction of inducible NO synthase with ensuing oligodendrocyte injury. These studies provide new insights into the role of HERV-mediated neuroinflammation and its contribution to an autoimmune disease.
Collapse
Affiliation(s)
- Joseph M Antony
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
48
|
Roy A, Liu X, Pahan K. Myelin basic protein-primed T cells induce neurotrophins in glial cells via alphavbeta3 [corrected] integrin. J Biol Chem 2007; 282:32222-32. [PMID: 17823123 PMCID: PMC2637617 DOI: 10.1074/jbc.m702899200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Increasing the level of neurotrophins within the central nervous system may have therapeutic efficacy in patients with various neurological diseases. Earlier we have demonstrated that myelin basic protein (MBP)-primed T cells induce the expression of various proinflammatory molecules in glial cells via cell-to-cell contact. Here we describe that after Th2 polarization by gemfibrozil or other drugs, MBP-primed T cells induced the expression of neurotrophic molecules such as brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), but not proinflammatory molecules in microglia and astroglia via cell-to-cell contact. MBP-primed Th2 cells expressed alpha5 and beta3 integrins and functional blocking antibodies against both alpha5 and beta3 integrins inhibited the ability of MBP-primed Th2 cells to induce glial neurotrophins. On the other hand, glial cells expressed PDGF-Rbeta and neutralization of this glial receptor abrogated the ability of Th2 cells to induce neurotrophins in glia. Activation of glial cAMP response element-binding protein (CREB) by MBP-primed Th2 cell contact and inhibition of contact-mediated expression of neurotrophins by antisense knockdown of glial CREB suggest that MBP-primed Th2 cell-glia contact induces the expression of neurotrophins through glial activation of CREB. Accordingly, blocking of either alpha5beta3 integrins on T cells or PDGF-Rbeta on glial cells impaired the ability of MBP-primed Th2 cells to induce glial activation of CREB. Furthermore, we demonstrate that these MBP-primed Th2 cells entered into the central nervous system and increased the expression of neurotrophins in vivo in the brain. This study illuminates the importance of alpha5beta3 and PDGF-Rbeta in guiding the novel neurotrophic property of neuroantigen-primed T cells via activation of CREB that may be of therapeutic importance in various neurological disorders.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Xiaojuan Liu
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center, College of Dentistry, Lincoln, Nebraska 68583
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center, College of Dentistry, Lincoln, Nebraska 68583
| |
Collapse
|
49
|
Jana M, Jana A, Pal U, Pahan K. A simplified method for isolating highly purified neurons, oligodendrocytes, astrocytes, and microglia from the same human fetal brain tissue. Neurochem Res 2007; 32:2015-22. [PMID: 17447141 PMCID: PMC2085356 DOI: 10.1007/s11064-007-9340-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Accepted: 03/21/2007] [Indexed: 01/05/2023]
Abstract
Elucidation of the underlying pathogenic mechanisms leading to apoptosis of neurons and oligodendrocytes and activation of microglia and astrocytes in different neurodegenerative and neuroinflammatory disorders remains a challenge in neuroscience. In order to overcome the challenge and find out therapeutic remedies, it is important to study live and death processes in each and every cell type of the brain. Here we present a protocol of isolating highly purified microglia, astrocytes, oligodendrocytes, and neurons, all four major cell types of the CNS, from the same human fetal brain tissue. As found in vivo, these primary neurons and oligodendroglia underwent apoptosis and cell death in response to neurodegenerative challenges. On the other hand, astroglia, and microglia, cells that do not die in neurodegenerative brains, became activated after inflammatory challenge. The availability of highly purified human brain cells will increase the possibility of developing therapies for different neurodegenerative disorders.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
50
|
Jana A, Pahan K. Oxidative stress kills human primary oligodendrocytes via neutral sphingomyelinase: implications for multiple sclerosis. J Neuroimmune Pharmacol 2007; 2:184-93. [PMID: 18040843 DOI: 10.1007/s11481-007-9066-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Accepted: 02/22/2007] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis (MS) is the most common human demyelinating disease of the central nervous system where oxidative stress has been proposed to play an important role in oligodendroglial death. However, molecular mechanisms that couple oxidative stress to the loss of oligodendrocytes are poorly understood. This study underlines the importance of neutral sphingomyelinase-ceramide pathway in mediating oxidative stress-induced apoptosis and cell death of human primary oligodendrocytes. Various oxidative stress-inducing agents, such as, superoxide radical produced by hypoxanthine and xanthine oxidase, hydrogen peroxide, aminotriazole capable of inhibiting catalase and increasing intracellular level of H2O2, or reduced glutathione-depleting diamide induced the activation of neutral sphingomyelinase and the production of ceramide. It is interesting to note that antisense knockdown of neutral but not acidic sphingomyelinase ablated oxidative stress-induced apoptosis and cell death in human primary oligodendrocytes. This study identifies neutral but not acidic sphingomyelinase as a target for possible therapeutic intervention in MS.
Collapse
Affiliation(s)
- Arundhati Jana
- Division of Neuroscience, Department of Neurological Sciences, Rush University Medical Center, Cohn Research Building, Suite 320, 1735 West Harrison St, Chicago, IL 60612, USA
| | | |
Collapse
|