1
|
Jiménez-Jiménez FJ, Alonso-Navarro H, Salgado-Cámara P, García-Martín E, Agúndez JAG. Antioxidant Therapies in the Treatment of Multiple Sclerosis. Biomolecules 2024; 14:1266. [PMID: 39456199 PMCID: PMC11506420 DOI: 10.3390/biom14101266] [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: 08/06/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Several studies have proposed a potential role for oxidative stress in the development of multiple sclerosis (MS). For this reason, it seems tentative to think that treatment with antioxidant substances could be useful in the treatment of this disease. In this narrative review, we provide a summary of the current findings on antioxidant treatments, both in experimental models of MS, especially in experimental autoimmune encephalomyelitis (EAE) and in the cuprizone-induced demyelination model, and clinical trials in patients diagnosed with MS. Practically all the antioxidants tested in experimental models of MS have shown improvement in clinical parameters, in delaying the evolution of the disease, and in improving histological and biochemical parameters, including decreased levels of markers of inflammation and oxidative stress in the central nervous system and other tissues. Only a few clinical trials have been carried out to investigate the potential efficacy of antioxidant substances in patients with MS, most of them in the short term and involving a short series of patients, so the results of these should be considered inconclusive. In this regard, it would be desirable to design long-term, randomized, multicenter clinical trials with a long series of patients, assessing several antioxidants that have demonstrated efficacy in experimental models of MS.
Collapse
Grants
- PI18/00540 Fondo de Investigación Sanitaria, Instituto de Salud Carlos, Madrid, Spain
- PI21/01683 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Madrid, Spain
- IB20134 Junta de Extremadura, Mérida, Spain
- GR21073 Junta de Extremadura, Mérida, Spain
Collapse
Affiliation(s)
| | - Hortensia Alonso-Navarro
- Section of Neurology, Hospital Universitario del Sureste, E28500 Arganda del Rey, Spain; (H.A.-N.); (P.S.-C.)
| | - Paula Salgado-Cámara
- Section of Neurology, Hospital Universitario del Sureste, E28500 Arganda del Rey, Spain; (H.A.-N.); (P.S.-C.)
| | - Elena García-Martín
- University Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, E10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| | - José A. G. Agúndez
- University Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, E10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| |
Collapse
|
2
|
Zhang C, Qiu M, Fu H. Oligodendrocytes in central nervous system diseases: the effect of cytokine regulation. Neural Regen Res 2024; 19:2132-2143. [PMID: 38488548 PMCID: PMC11034588 DOI: 10.4103/1673-5374.392854] [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: 07/31/2023] [Revised: 11/08/2023] [Accepted: 12/06/2023] [Indexed: 04/24/2024] Open
Abstract
Cytokines including tumor necrosis factor, interleukins, interferons, and chemokines are abundantly produced in various diseases. As pleiotropic factors, cytokines are involved in nearly every aspect of cellular functions such as migration, survival, proliferation, and differentiation. Oligodendrocytes are the myelin-forming cells in the central nervous system and play critical roles in the conduction of action potentials, supply of metabolic components for axons, and other functions. Emerging evidence suggests that both oligodendrocytes and oligodendrocyte precursor cells are vulnerable to cytokines released under pathological conditions. This review mainly summarizes the effects of cytokines on oligodendrocyte lineage cells in central nervous system diseases. A comprehensive understanding of the effects of cytokines on oligodendrocyte lineage cells contributes to our understanding of central nervous system diseases and offers insights into treatment strategies.
Collapse
Affiliation(s)
- Chengfu Zhang
- Center for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Mengsheng Qiu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Hui Fu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Lee JE, Kim M, Ochiai S, Kim SH, Yeo H, Bok J, Kim J, Park M, Kim D, Lamiable O, Lee M, Kim MJ, Kim HY, Ronchese F, Kwon SW, Lee H, Kim TG, Chung Y. Tonic type 2 immunity is a critical tissue checkpoint controlling autoimmunity in the skin. Cell Rep 2024; 43:114364. [PMID: 38900635 DOI: 10.1016/j.celrep.2024.114364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/26/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
Immunoregulatory mechanisms established in the lymphoid organs are vital for preventing autoimmunity. However, the presence of similar mechanisms in non-lymphoid tissues remains unclear. Through transcriptomic and lipidomic analyses, we find a negative association between psoriasis and fatty acid metabolism, as well as Th2 signature. Homeostatic expression of liver X receptor (LXR) and peroxisome proliferator-activated receptor gamma (PPARγ) is essential for maintaining fatty acid metabolism and for conferring resistance to psoriasis in mice. Perturbation of signal transducer and activator of transcription 6 (STAT6) diminishes the homeostatic levels of LXR and PPARγ. Furthermore, mice lacking STAT6, interleukin 4 receptor alpha (IL-4Rα), or IL-13, but not IL-4, exhibit increased susceptibility to psoriasis. Under steady state, innate lymphoid cells (ILCs) are the primary producers of IL-13. In human skin, inhibiting tonic type 2 immunity exacerbates psoriasis-like inflammation and IL-17A, while activating LXR or PPARγ inhibits them. Hence, we propose that tonic type 2 immunity, driven by IL-13-producing ILCs, represents a crucial tissue checkpoint that represses autoimmunity and maintains lipid homeostasis in the skin.
Collapse
Affiliation(s)
- Jeong-Eun Lee
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Mina Kim
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sotaro Ochiai
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sung-Hee Kim
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeonuk Yeo
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jahyun Bok
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jiyeon Kim
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Miso Park
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea; College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Daehong Kim
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | | | - Myunggyo Lee
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Min-Ju Kim
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Hye Young Kim
- College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand.
| | - Sung Won Kwon
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| | - Haeseung Lee
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea.
| | - Tae-Gyun Kim
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Yeonseok Chung
- Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
da Silva MI, Oli N, Gambonini F, Ott T. Effects of parity and early pregnancy on peripheral blood leukocytes in dairy cattle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592827. [PMID: 38766084 PMCID: PMC11100682 DOI: 10.1101/2024.05.06.592827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Subfertility remains a major problem in the dairy industry. Only 35-40% of high-yielding dairy cows and 55-65% of nonlactating heifers become pregnant after their first service. The immune system plays a critical role in the establishment of pregnancy. However, it can also create challenges for embryo survival and contribute to reduced fertility. We conducted 2 separate experiments to characterize changes in subsets of peripheral blood leukocytes (PBL) and their phenotype over the estrous cycle and early pregnancy in heifers and cows. We used flow cytometry and RT-qPCR to assess protein and mRNA expression of molecules important for immune function. We observed that monocytes and T cells were most affected by pregnancy status in heifers, whereas, CD8+ lymphocytes and natural killer (NK) cells were most affected during early pregnancy in cows. Changes in immune parameters measured appeared to be greater in heifers than cows including changes in expression of numerous immune function molecules. To test the hypothesis, we conducted a third experiment to simultaneously analyze the immunological responses to pregnancy between cows and heifers. We observed that cows had greater expression of proinflammatory cytokines and molecules associated with leukocyte migration and phagocytosis compared to heifers. Moreover, animals that failed to become pregnant showed altered expression of anti-inflammatory molecules. Overall, these findings support the hypothesis that early pregnancy signaling alters the proportions and functions of peripheral blood immune cells and differences between cows and heifers may yield insight into the reduced fertility of mature lactating dairy cows.
Collapse
Affiliation(s)
- M I da Silva
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - N Oli
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - F Gambonini
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - T Ott
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
5
|
He K, Zhou X, Du H, Zhao J, Deng R, Wang J. A review on the relationship between Arachidonic acid 15-Lipoxygenase (ALOX15) and diabetes mellitus. PeerJ 2023; 11:e16239. [PMID: 37849828 PMCID: PMC10578307 DOI: 10.7717/peerj.16239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Arachidonic acid 15-lipoxygenase (ALOX15), as one of the lipoxygenase family, is mainly responsible for catalyzing the oxidation of various fatty acids to produce a variety of lipid components, contributing to the pathophysiological processes of various immune and inflammatory diseases. Studies have shown that ALOX15 and its related products are widely distributed in human tissues and related to multiple diseases such as liver, cardiovascular, cerebrovascular diseases, diabetes mellitus and other diseases. Diabetes mellitus (DM), the disease studied in this article, is a metabolic disease characterized by a chronic increase in blood glucose levels, which is significantly related to inflammation, oxidative stress, ferroptosis and other mechanisms, and it has a high incidence in the population, accompanied by a variety of complications. Figuring out how ALOX15 is involved in DM is critical to understanding its role in diseases. Therefore, ALOX15 inhibitors or combination therapy containing inhibitors may deliver a novel research direction for the treatment of DM and its complications. This article aims to review the biological effect and the possible function of ALOX15 in the pathogenesis of DM.
Collapse
Affiliation(s)
- Kaiying He
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Xiaochun Zhou
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Hongxuan Du
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jing Zhao
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Rongrong Deng
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jianqin Wang
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| |
Collapse
|
6
|
Inflammation- and Metastasis-Related Proteins Expression Changes in Early Stages in Tumor and Non-Tumor Adjacent Tissues of Colorectal Cancer Samples. Cancers (Basel) 2022; 14:cancers14184487. [PMID: 36139645 PMCID: PMC9497293 DOI: 10.3390/cancers14184487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Non-tumor adjacent tissue plays a key role in colorectal cancer development, as well as chronic inflammation, but their role has not yet been dilucidated. In addition, inflammation is a process which is related to epithelial-mesenchymal transition and metastasis, but their changes across the different colorectal cancer stages are not fully studied. Understanding how these processes participate in all colorectal cancer phases can be key to a better understanding of the disease. Abstract Chronic inflammation can induce malignant cell transformation, having an important role in all colorectal cancer (CRC) phases. Non-tumor adjacent tissue plays an important role in tumor progression, but its implication in CRC has not yet been fully elucidated. The aim was to analyze the expression of inflammatory, epithelial-mesenchymal transition (EMT), and metastasis-related proteins in both tumor and non-tumor adjacent tissues from CRC patients by western blot. Tumor tissue presented an increase in metastasis and EMT-related proteins compared to non-tumor adjacent tissue, especially in stage II. Tumor tissue stage II also presented an increase in inflammatory-related proteins compared to other stages, which was also seen in non-tumor adjacent tissue stage II. Additionally, the relapse-free survival study of Vimentin and VEGF-B expression levels in stage II patients showed that the higher the expression levels of each protein, the lower 10-year relapse-free survival. These could suggest that some metastasis-related signalling pathways may be activated in stage II in tumor tissue, accompanied by an increase in inflammation. Furthermore, non-tumor adjacent tissue presented an increase of the inflammatory status that could be the basis for future tumor progression. In conclusion, these proteins could be useful as biomarkers of diagnosis for CRC at early stages.
Collapse
|
7
|
Stifel U, Caratti G, Tuckermann J. Novel insights into the regulation of cellular catabolic metabolism in macrophages through nuclear receptors. FEBS Lett 2022; 596:2617-2629. [PMID: 35997656 DOI: 10.1002/1873-3468.14474] [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: 02/15/2022] [Revised: 06/14/2022] [Accepted: 07/04/2022] [Indexed: 11/07/2022]
Abstract
Regulation of cellular catabolic metabolism in immune cells has recently become a major concept for resolution of inflammation. Nuclear receptors (NRs), including peroxisome proliferator activator receptors (PPARs), 1,25-dihydroxyvitamin D(3) receptor (VDR), liver X receptors (LXRs), glucocorticoid receptors (GRs), estrogen-related receptor α (ERRα) and Nur77, have been identified as major modulators of inflammation, affecting innate immune cells, such as macrophages. Evidence emerges on how NRs regulate cellular metabolism in macrophages during inflammatory processes and contribute to the resolution of inflammation. This could have new implications for our understanding of how NRs shape immune responses and inform anti-inflammatory drug design. This review will highlight the recent developments about NRs and their role in cellular metabolism in macrophages.
Collapse
Affiliation(s)
- Ulrich Stifel
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany
| | - Giorgio Caratti
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany
| |
Collapse
|
8
|
Zorrilla Veloz RI, McKenzie T, Palacios BE, Hu J. Nuclear hormone receptors in demyelinating diseases. J Neuroendocrinol 2022; 34:e13171. [PMID: 35734821 PMCID: PMC9339486 DOI: 10.1111/jne.13171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Demyelination results from the pathological loss of myelin and is a hallmark of many neurodegenerative diseases. Despite the prevalence of demyelinating diseases, there are no disease modifying therapies that prevent the loss of myelin or promote remyelination. This review aims to summarize studies in the field that highlight the importance of nuclear hormone receptors in the promotion and maintenance of myelination and the relevance of nuclear hormone receptors as potential therapeutic targets for demyelinating diseases. These nuclear hormone receptors include the estrogen receptor, progesterone receptor, androgen receptor, vitamin D receptor, thyroid hormone receptor, peroxisome proliferator-activated receptor, liver X receptor, and retinoid X receptor. Pre-clinical studies in well-established animal models of demyelination have shown a prominent role of these nuclear hormone receptors in myelination through their promotion of oligodendrocyte maturation and development. The activation of the nuclear hormone receptors by their ligands also promotes the synthesis of myelin proteins and lipids in mouse models of demyelination. There are limited clinical studies that focus on how the activation of these nuclear hormone receptors could alleviate demyelination in patients with diseases such as multiple sclerosis (MS). However, the completed clinical trials have reported improved clinical outcome in MS patients treated with the ligands of some of these nuclear hormone receptors. Together, the positive results from both clinical and pre-clinical studies point to nuclear hormone receptors as promising therapeutic targets to counter demyelination.
Collapse
Affiliation(s)
- Rocío I Zorrilla Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Takese McKenzie
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Bridgitte E Palacios
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
9
|
Mannan A, Garg N, Singh TG, Kang HK. Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ɣ): Molecular Effects and Its Importance as a Novel Therapeutic Target for Cerebral Ischemic Injury. Neurochem Res 2021; 46:2800-2831. [PMID: 34282491 DOI: 10.1007/s11064-021-03402-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Cerebral ischemic injury is a leading cause of death and long-term disability throughout the world. Peroxisome proliferator-activated receptor gamma (PPAR-ɣ) is a ligand-activated nuclear transcription factor that is a member of the PPAR family. PPAR-ɣ has been shown in several in vitro and in vivo models to prevent post-ischemic inflammation and neuronal damage by negatively controlling the expression of genes modulated by cerebral ischemic injury, indicating a neuroprotective effect during cerebral ischemic injury. A extensive literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on the mechanistic role of Peroxisome proliferator activated receptor gamma and its modulation in Cerebral ischemic injury. PPAR-ɣ can interact with specific DNA response elements to control gene transcription and expression when triggered by its ligand. It regulates lipid metabolism, improves insulin sensitivity, modulates antitumor mechanisms, reduces oxidative stress, and inhibits inflammation. This review article provides insights on the current state of research into the neuroprotective effects of PPAR-ɣ in cerebral ischemic injury, as well as the cellular and molecular mechanisms by which these effects are modulated, such as inhibition of inflammation, reduction of oxidative stress, suppression of pro-apoptotic production, modulation of transcription factors, and restoration of injured tissue through neurogenesis and angiogenesis.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Harmeet Kaur Kang
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| |
Collapse
|
10
|
Sandi D, Fricska-Nagy Z, Bencsik K, Vécsei L. Neurodegeneration in Multiple Sclerosis: Symptoms of Silent Progression, Biomarkers and Neuroprotective Therapy-Kynurenines Are Important Players. Molecules 2021; 26:molecules26113423. [PMID: 34198750 PMCID: PMC8201043 DOI: 10.3390/molecules26113423] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/17/2022] Open
Abstract
Neurodegeneration is one of the driving forces behind the pathogenesis of multiple sclerosis (MS). Progression without activity, pathopsychological disturbances (cognitive impairment, depression, fatigue) and even optic neuropathy seems to be mainly routed in this mechanism. In this article, we aim to give a comprehensive review of the clinical aspects and symptomology, radiological and molecular markers and potential therapeutic targets of neurodegeneration in connection with MS. As the kynurenine pathway (KP) was evidenced to play an important role in the pathogenesis of other neurodegenerative conditions (even implied to have a causative role in some of these diseases) and more and more recent evidence suggest the same central role in the neurodegenerative processes of MS as well, we pay special attention to the KP. Metabolites of the pathway are researched as biomarkers of the disease and new, promising data arising from clinical evaluations show the possible therapeutic capability of KP metabolites as neuroprotective drugs in MS. Our conclusion is that the kynurenine pathway is a highly important route of research both for diagnostic and for therapeutic values and is expected to yield concrete results for everyday medicine in the future.
Collapse
Affiliation(s)
- Dániel Sandi
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
| | - Zsanett Fricska-Nagy
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
| | - Krisztina Bencsik
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
| | - László Vécsei
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
- MTA-SZTE Neuroscience Research Group, University of Szeged, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Centre, University of Szeged, H-6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-384; Fax: +36-62-545-597
| |
Collapse
|
11
|
Pu H, Zheng X, Jiang X, Mu H, Xu F, Zhu W, Ye Q, Jizhang Y, Hitchens TK, Shi Y, Hu X, Leak RK, Dixon CE, Bennett MV, Chen J. Interleukin-4 improves white matter integrity and functional recovery after murine traumatic brain injury via oligodendroglial PPARγ. J Cereb Blood Flow Metab 2021; 41:511-529. [PMID: 32757740 PMCID: PMC7922743 DOI: 10.1177/0271678x20941393] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Long-term neurological recovery after severe traumatic brain injury (TBI) is strongly linked to the repair and functional restoration of injured white matter. Emerging evidence suggests that the anti-inflammatory cytokine interleukin-4 (IL-4) plays an important role in promoting white matter integrity after cerebral ischemic injury. Here, we report that delayed intranasal delivery of nanoparticle-packed IL-4 boosted sensorimotor neurological recovery in a murine model of controlled cortical impact, as assessed by a battery of neurobehavioral tests for up to five weeks. Post-injury IL-4 treatment failed to reduce macroscopic brain lesions after TBI, but preserved the structural and functional integrity of white matter, at least in part through oligodendrogenesis. IL-4 directly facilitated the differentiation of oligodendrocyte progenitor cells (OPCs) into mature myelin-producing oligodendrocytes in primary cultures, an effect that was attenuated by selective PPARγ inhibition. IL-4 treatment after TBI in vivo also failed to stimulate oligodendrogenesis or improve white matter integrity in OPC-specific PPARγ conditional knockout (cKO) mice. Accordingly, IL-4-afforded improvements in sensorimotor neurological recovery after TBI were markedly impaired in the PPARγ cKO mice compared to wildtype controls. These results support IL-4 as a potential novel neurorestorative therapy to improve white matter functionality and mitigate the long-term neurological consequences of TBI.
Collapse
Affiliation(s)
- Hongjian Pu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xuan Zheng
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoyan Jiang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hongfeng Mu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fei Xu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wen Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Ye
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yunneng Jizhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - T Kevin Hitchens
- Animal Imaging Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yejie Shi
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - C Edward Dixon
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Vl Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jun Chen
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Zhang J, Rong P, Zhang L, He H, Zhou T, Fan Y, Mo L, Zhao Q, Han Y, Li S, Wang Y, Yan W, Chen H, You Z. IL4-driven microglia modulate stress resilience through BDNF-dependent neurogenesis. SCIENCE ADVANCES 2021; 7:eabb9888. [PMID: 33731342 PMCID: PMC7968840 DOI: 10.1126/sciadv.abb9888] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 01/28/2021] [Indexed: 05/15/2023]
Abstract
Adult neurogenesis in the dentate gyrus of the hippocampus is regulated by specific microglia groups and functionally implicated in behavioral responses to stress. However, the role of microglia in hippocampal neurogenesis and stress resilience remains unclear. We identified interleukin 4 (IL4)-driven microglia characterized by high expression of Arg1, which is critical in maintaining hippocampal neurogenesis and stress resistance. Decreasing Arg1+ microglia in the hippocampus by knocking down the microglial IL4R suppressed hippocampal neurogenesis and enhanced stress vulnerability. Increasing Arg1+ microglia in the hippocampus by enhancing IL4 signaling restored hippocampal neurogenesis and the resilience to stress-induced depression. Brain-derived neurotrophic factor (BDNF) was found necessary for the proneurogenesis effects of IL4-driven microglia. Together, our findings suggest that IL4-driven microglia in the hippocampus trigger BDNF-dependent neurogenesis responding to chronic stress, helping protect against depressive-like symptoms. These findings identify the modulation of a specific microglial phenotype as a treatment strategy for mood disorders.
Collapse
Affiliation(s)
- Jinqiang Zhang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Peijing Rong
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lijuan Zhang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hui He
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Tao Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Yonghua Fan
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Li Mo
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Qiuying Zhao
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yue Han
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Shaoyuan Li
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yifei Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wan Yan
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Huafu Chen
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Zili You
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| |
Collapse
|
13
|
Vaes JEG, Brandt MJV, Wanders N, Benders MJNL, de Theije CGM, Gressens P, Nijboer CH. The impact of trophic and immunomodulatory factors on oligodendrocyte maturation: Potential treatments for encephalopathy of prematurity. Glia 2020; 69:1311-1340. [PMID: 33595855 PMCID: PMC8246971 DOI: 10.1002/glia.23939] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Encephalopathy of prematurity (EoP) is a major cause of morbidity in preterm neonates, causing neurodevelopmental adversities that can lead to lifelong impairments. Preterm birth-related insults, such as cerebral oxygen fluctuations and perinatal inflammation, are believed to negatively impact brain development, leading to a range of brain abnormalities. Diffuse white matter injury is a major hallmark of EoP and characterized by widespread hypomyelination, the result of disturbances in oligodendrocyte lineage development. At present, there are no treatment options available, despite the enormous burden of EoP on patients, their families, and society. Over the years, research in the field of neonatal brain injury and other white matter pathologies has led to the identification of several promising trophic factors and cytokines that contribute to the survival and maturation of oligodendrocytes, and/or dampening neuroinflammation. In this review, we discuss the current literature on selected factors and their therapeutic potential to combat EoP, covering a wide range of in vitro, preclinical and clinical studies. Furthermore, we offer a future perspective on the translatability of these factors into clinical practice.
Collapse
Affiliation(s)
- Josine E G Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Myrna J V Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Nikki Wanders
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | | | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
14
|
Bozic I, Savic D, Lavrnja I. Astrocyte phenotypes: Emphasis on potential markers in neuroinflammation. Histol Histopathol 2020; 36:267-290. [PMID: 33226087 DOI: 10.14670/hh-18-284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes, the most abundant glial cells in the central nervous system (CNS), have numerous integral roles in all CNS functions. They are essential for synaptic transmission and support neurons by providing metabolic substrates, secreting growth factors and regulating extracellular concentrations of ions and neurotransmitters. Astrocytes respond to CNS insults through reactive astrogliosis, in which they go through many functional and molecular changes. In neuroinflammatory conditions reactive astrocytes exert both beneficial and detrimental functions, depending on the context and heterogeneity of astrocytic populations. In this review we profile astrocytic diversity in the context of neuroinflammation; with a specific focus on multiple sclerosis (MS) and its best-described animal model experimental autoimmune encephalomyelitis (EAE). We characterize two main subtypes, protoplasmic and fibrous astrocytes and describe the role of intermediate filaments in the physiology and pathology of these cells. Additionally, we outline a variety of markers that are emerging as important in investigating astrocytic biology in both physiological conditions and neuroinflammation.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
15
|
Xu L, Ma X, Verma N, Perie L, Pendse J, Shamloo S, Marie Josephson A, Wang D, Qiu J, Guo M, Ping X, Allen M, Noguchi A, Springer D, Shen F, Liu C, Zhang S, Li L, Li J, Xiao J, Lu J, Du Z, Luo J, Aleman JO, Leucht P, Mueller E. PPARγ agonists delay age-associated metabolic disease and extend longevity. Aging Cell 2020; 19:e13267. [PMID: 33219735 PMCID: PMC7681041 DOI: 10.1111/acel.13267] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022] Open
Abstract
Aging leads to a number of disorders caused by cellular senescence, tissue damage, and organ dysfunction. It has been reported that anti‐inflammatory and insulin‐sensitizing compounds delay, or reverse, the aging process and prevent metabolic disorders, neurodegenerative disease, and muscle atrophy, improving healthspan and extending lifespan. Here we investigated the effects of PPARγ agonists in preventing aging and increasing longevity, given their known properties in lowering inflammation and decreasing glycemia. Our molecular and physiological studies show that long‐term treatment of mice at 14 months of age with low doses of the PPARγ ligand rosiglitazone (Rosi) improved glucose metabolism and mitochondrial functionality. These effects were associated with decreased inflammation and reduced tissue atrophy, improved cognitive function, and diminished anxiety‐ and depression‐like conditions, without any adverse effects on cardiac and skeletal functionality. Furthermore, Rosi treatment of mice started when they were 14 months old was associated with lifespan extension. A retrospective analysis of the effects of the PPARγ agonist pioglitazone (Pio) on longevity showed decreased mortality in patients receiving Pio compared to those receiving a PPARγ‐independent insulin secretagogue glimepiride. Taken together, these data suggest the possibility of using PPARγ agonists to promote healthy aging and extend lifespan.
Collapse
Affiliation(s)
- Lingyan Xu
- Division of Endocrinology Diabetes and MetabolismNYU Grossman School of Medicine New York NY USA
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Xinran Ma
- Division of Endocrinology Diabetes and MetabolismNYU Grossman School of Medicine New York NY USA
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Narendra Verma
- Division of Endocrinology, Diabetes and Metabolism NYU Grossman School of Medicine New York NY USA
| | - Luce Perie
- Division of Endocrinology, Diabetes and Metabolism NYU Grossman School of Medicine New York NY USA
| | - Jay Pendse
- Division of Endocrinology, Diabetes and Metabolism NYU Grossman School of Medicine New York NY USA
- Medical Service Veterans Affairs New York Harbor Healthcare System New York NY USA
| | - Sama Shamloo
- Division of Endocrinology, Diabetes and Metabolism NYU Grossman School of Medicine New York NY USA
| | - Anne Marie Josephson
- Department of Orthopedic Surgery NYU Grossman School of Medicine New York NY USA
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Jin Qiu
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Xiaodan Ping
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Michele Allen
- Murine Phenotyping Core facility NHLBI National Institutes of Health Bethesda MD USA
| | - Audrey Noguchi
- Murine Phenotyping Core facility NHLBI National Institutes of Health Bethesda MD USA
| | - Danielle Springer
- Murine Phenotyping Core facility NHLBI National Institutes of Health Bethesda MD USA
| | - Fei Shen
- School of Physical Education & Health Care East China Normal University Shanghai China
| | - Caizhi Liu
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Shiwei Zhang
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Lingyu Li
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Jin Li
- Cardiac Regeneration and Ageing Lab Institute of Cardiovascular Sciences School of Life Science Shanghai University Shanghai China
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab Institute of Cardiovascular Sciences School of Life Science Shanghai University Shanghai China
| | - Jian Lu
- School of Physical Education & Health Care East China Normal University Shanghai China
| | - Zhenyu Du
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Jian Luo
- Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences East China Normal University Shanghai China
| | - Jose O. Aleman
- Division of Endocrinology, Diabetes and Metabolism NYU Grossman School of Medicine New York NY USA
- Medical Service Veterans Affairs New York Harbor Healthcare System New York NY USA
| | - Philipp Leucht
- Department of Orthopedic Surgery NYU Grossman School of Medicine New York NY USA
| | - Elisabetta Mueller
- Division of Endocrinology Diabetes and MetabolismNYU Grossman School of Medicine New York NY USA
- Division of Endocrinology, Diabetes and Metabolism NYU Grossman School of Medicine New York NY USA
- Cardiac Regeneration and Ageing Lab Institute of Cardiovascular Sciences School of Life Science Shanghai University Shanghai China
| |
Collapse
|
16
|
Gaojian T, Dingfei Q, Linwei L, Xiaowei W, Zheng Z, Wei L, Tong Z, Benxiang N, Yanning Q, Wei Z, Jian C. Parthenolide promotes the repair of spinal cord injury by modulating M1/M2 polarization via the NF-κB and STAT 1/3 signaling pathway. Cell Death Discov 2020; 6:97. [PMID: 33083018 PMCID: PMC7538575 DOI: 10.1038/s41420-020-00333-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disease; however, there is no effective treatment for spinal cord injury. Neuroinflammation involves the activation of resident microglia and the infiltration of macrophages is the major pathogenesis of SCI secondary injury and considered to be the therapeutic target of SCI. Parthenolide (PN) has been reported to exert anti-inflammatory effects in fever, migraines, arthritis, and superficial inflammation; however, the role of PN in SCI therapeutics has not been clarified. In this study, we showed that PN could improve the functional recovery of spinal cord in mice as revealed by increased BMS scores and decreased cavity of spinal cord injury in vivo. Immunofluorescence staining experiments confirmed that PN could promote axonal regeneration, increase myelin reconstitution, reduce chondroitin sulfate formation, inhibit scar hyperplasia, suppress the activation of A1 neurotoxic reactive astrocytes and facilitate shift from M1 to M2 polarization of microglia/macrophages. To verify how PN exerts its effects on microglia/macrophages polarization, we performed the mechanism study in vitro in microglia cell line BV-2. PN could significantly reduce M1 polarization in BV2 cells and partially rescue the decrease in the expression of M2 phenotype markers of microglia/macrophage induced by LPS, but no significant effect on M2 polarization stimulated with IL-4 was observed. Further study demonstrated PN inhibited NF-κB signal pathway directly or indirectly, and suppressed activation of signal transducer and activator of transcription 1 or 3 (STAT1/3) via reducing the expression of HDAC1 and subsequently increasing the levels of STAT1/3 acetylation. Overall, our study illustrated that PN may be a promising strategy for traumatic SCI.
Collapse
Affiliation(s)
- Tao Gaojian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
- Department of Pain Management, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008 China
| | - Qian Dingfei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Li Linwei
- Department of Orthopedic, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| | - Wang Xiaowei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhou Zheng
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Liu Wei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhu Tong
- Department of Pain Management, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008 China
| | - Ning Benxiang
- Department of Pain Management, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008 China
| | - Qian Yanning
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhou Wei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Chen Jian
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
17
|
Zanno AE, Romer MA, Fox L, Golden T, Jaeckle-Santos L, Simmons RA, Grinspan JB. Reducing Th2 inflammation through neutralizing IL-4 antibody rescues myelination in IUGR rat brain. J Neurodev Disord 2019; 11:34. [PMID: 31839002 PMCID: PMC6913005 DOI: 10.1186/s11689-019-9297-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) is a common complication of pregnancy and is associated with significant neurological deficits in infants, including white matter damage. Previous work using an animal model of IUGR has demonstrated that IUGR rats exhibit neurobehavioral deficits and developmental delays in oligodendrocyte maturation and myelination, but the mechanisms which cause this delay are unknown. Inflammation may be an important etiological factor in IUGR and has been recognized as playing a fundamental role in the pathogenesis of myelin disorders, including cerebral palsy. METHODS To create the model, the uterine arteries of pregnant rats were ligated at embryonic day 15. Rats delivered spontaneously. Cytokine and chemokine expression was evaluated at one prenatal and three postnatal time points, and myelin protein expression and oligodendrocyte cell numbers were evaluated by several methods at postnatal day 14. IL-4 was identified as a potential inhibitor of myelination, and rat pups were injected with IL-4 function blocking antibody from postnatal days 1-5 and myelination was assessed. RESULTS Here, we show a novel mechanism of white matter injury. IUGR induces an exaggerated Th2 response in the developing rat brain, including upregulation of several Th2 cytokines. Of these, IL-4 is significantly increased during the period corresponding to robust developmental myelination. We show that neutralizing IL-4 antibody therapy given in the newborn period ameliorates inflammation and restores myelin protein expression and oligodendrocyte cell number in the IUGR brain to control levels, demonstrating a novel role for Th2 responses and IL-4 in IUGR and white matter injury. In addition, IL-4 directly affects oligodendrocytes in vitro decreasing differentiation. CONCLUSIONS In this study, we have identified inflammation as a factor in the decrease in myelin seen in an animal model of IUGR. IL-4, an inflammatory protein often thought to be protective in the adult, is specifically increased, and treatment of these animals to prevent this increase ameliorates white matter damage. Our results suggest that the immune system plays a role in IUGR that is different in the perinatal period than in the adult and preventing this exaggerated Th2 response may be a potential therapeutic target.
Collapse
Affiliation(s)
- Allison E. Zanno
- Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
| | - Micah A. Romer
- Department of Neurology, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 516D Abramson Center, 3615 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Lauren Fox
- Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
| | - Thea Golden
- Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
| | - Lane Jaeckle-Santos
- Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
| | - Rebecca A. Simmons
- Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
| | - Judith B. Grinspan
- Department of Neurology, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 516D Abramson Center, 3615 Civic Center Blvd, Philadelphia, PA 19104 USA
| |
Collapse
|
18
|
Multiplex profiling of inflammation-related bioactive lipid mediators in Toxocara canis- and Toxocara cati-induced neurotoxocarosis. PLoS Negl Trop Dis 2019; 13:e0007706. [PMID: 31557153 PMCID: PMC6762062 DOI: 10.1371/journal.pntd.0007706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/14/2019] [Indexed: 12/18/2022] Open
Abstract
Background Somatic migration of Toxocara canis- and T. cati-larvae in humans may cause neurotoxocarosis (NT) when larvae accumulate and persist in the central nervous system (CNS). Host- or parasite-induced immunoregulatory processes contribute to the pathogenesis; however, detailed data on involvement of bioactive lipid mediators, e.g. oxylipins or eico-/docosanoids, which are involved in the complex molecular signalling network during infection and inflammation, are lacking. Methodology/Principal findings To elucidate if T. canis- and T. cati-induced NT affects the homeostasis of oxylipins during the course of infection, a comprehensive lipidomic profiling in brains (cerebra and cerebella) of experimentally infected C57BL/6J mice was conducted at six different time points post infection (pi) by liquid-chromatography coupled to electrospray tandem mass spectrometry (LC-ESI-MS/MS). Only minor changes were detected regarding pro-inflammatory prostaglandins (cyclooxygenase pathway). In contrast, a significant increase of metabolites resulting from lipoxygenase pathways was observed for both infection groups and brain regions, implicating a predominantly anti-inflammatory driven immune response. This observation was supported by a significantly increased 13-hydroxyoctadecadienoic acid (HODE)/9-HODE ratio during the subacute phase of infection, indicating an anti-inflammatory response to neuroinfection. Except for the specialised pro-resolving mediator (SPM) neuroprotectin D1 (NPD1), which was detected in mice infected with both pathogens during the subacute phase of infection, no other SPMs were detected. Conclusions/Significance The obtained results demonstrate the influence of Toxocara spp. on oxylipins as part of the immune response of the paratenic hosts. Furthermore, this study shows differences in the alteration of the oxylipin composition between T. canis- and T. cati-brain infection. Results contribute to a further understanding of the largely unknown pathogenesis and mechanisms of host-parasite interactions during NT. Neurotoxocarosis (NT) is induced by larvae of the zoonotic roundworms Toxocara canis and T. cati migrating and persisting in the central nervous system of paratenic hosts, and may be accompanied by severe neurological symptoms. Toxocara spp. are known to modulate the hosts’ immune response, but data concerning involvement of signalling molecules are lacking. An important class of mediators participating in the complex molecular signalling network during infection and inflammation are bioactive regulatory lipids, derived from arachidonic acid and other polyunsaturated fatty acids. For a better understanding of inflammatory processes in the brain during an infection with Toxocara spp., a comprehensive analysis of regulatory lipids was conducted. The infection was predominantly characterised by only minor changes in the pattern of pro-inflammatory oxylipins, while anti-inflammatory metabolites, derived from lipoxygenase pathways, were significantly elevated in the subacute phase as well as in the beginning of the chronic phase of infection. This trend was also reflected in the 13-HODE/9-HODE ratio, a biomarker for the immunological status of an active infection. Obtained data provide a valuable insight in the host’s immune reaction as response against neuroinvasive Toxocara spp.-larvae, contributing to the characterisation of the mostly unknown pathogenesis of NT.
Collapse
|
19
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
20
|
The interleukin-4/PPARγ signaling axis promotes oligodendrocyte differentiation and remyelination after brain injury. PLoS Biol 2019; 17:e3000330. [PMID: 31226122 PMCID: PMC6608986 DOI: 10.1371/journal.pbio.3000330] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/03/2019] [Accepted: 05/31/2019] [Indexed: 11/29/2022] Open
Abstract
The repair of white matter damage is of paramount importance for functional recovery after brain injuries. Here, we report that interleukin-4 (IL-4) promotes oligodendrocyte regeneration and remyelination. IL-4 receptor expression was detected in a variety of glial cells after ischemic brain injury, including oligodendrocyte lineage cells. IL-4 deficiency in knockout mice resulted in greater deterioration of white matter over 14 d after stroke. Consistent with these findings, intranasal delivery of IL-4 nanoparticles after stroke improved white matter integrity and attenuated long-term sensorimotor and cognitive deficits in wild-type mice, as revealed by histological immunostaining, electron microscopy, diffusion tensor imaging, and electrophysiology. The selective effect of IL-4 on remyelination was verified in an ex vivo organotypic model of demyelination. By leveraging primary oligodendrocyte progenitor cells (OPCs), microglia-depleted mice, and conditional OPC-specific peroxisome proliferator-activated receptor gamma (PPARγ) knockout mice, we discovered a direct salutary effect of IL-4 on oligodendrocyte differentiation that was mediated by the PPARγ axis. Our findings reveal a new regenerative role of IL-4 in the central nervous system (CNS), which lies beyond its known immunoregulatory functions on microglia/macrophages or peripheral lymphocytes. Therefore, intranasal IL-4 delivery may represent a novel therapeutic strategy to improve white matter integrity in stroke and other brain injuries. The repair and remyelination of white matter are of paramount importance for functional recovery after brain injuries. This study shows that interleukin-4 plays an essential role in oligodendrocyte differentiation and long-term white matter recovery, beyond its well-known immunoregulatory functions, and is mediated by the PPARγ axis.
Collapse
|
21
|
Mahmoodi M, Amiri H, Ayoobi F, Rahmani M, Taghipour Z, Ghavamabadi RT, Jafarzadeh A, Sankian M. Carvacrol ameliorates experimental autoimmune encephalomyelitis through modulating pro- and anti-inflammatory cytokines. Life Sci 2019; 219:257-263. [DOI: 10.1016/j.lfs.2018.11.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 11/17/2022]
|
22
|
Adegoke EO, Wang X, Wang H, Wang C, Zhang H, Zhang G. Selenium (Na 2SeO 3) Upregulates Expression of Immune Genes and Blood-Testis Barrier Constituent Proteins of Bovine Sertoli Cell In Vitro. Biol Trace Elem Res 2018; 185:332-343. [PMID: 29383579 DOI: 10.1007/s12011-018-1248-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/11/2018] [Indexed: 01/03/2023]
Abstract
Sertoli cells were isolated from newborn calves and cultured in a medium supplemented with 0, 0.25, 0.50, 0.75, and 1.00 mg/L of sodium selenite to study their immune stimulatory effect, influence on cell's viability, and expression of blood-testis barrier proteins (occludin, connexin-43, zonula occluden, E-cadherin) using quantitative PCR and western blot analyses. Results showed that medium supplemented with 0.50 mg/L of selenium significantly (P < 0.05) promoted cell viability, upregulated toll-like receptor gene (TLR4), anti-inflammatory cytokines (IL-4, IL-10, TGFβ1), and expressions of blood-testis barrier proteins, and modulated expressions of pro-inflammatory cytokines (TNF-α, IL-1β, IFN-γ). Sertoli cells grown in culture medium supplemented with 0.25 mg/L of selenium significantly upregulated TLR4, IL-4, IL-10, TGFβ1, and blood-testis barrier proteins compared to the control group. Sodium selenite supplementation at 0.75 and 1.00 mg/L levels was cytotoxic and temporarily downregulated the expression of blood-testis barrier protein within 24 h after culture; however, commencing from 72 h post culture, increased cell viability and upregulation of expression of blood-testis barrier proteins were observed. In conclusion, the results of this study showed that selenium supplementation in the culture medium up to 0.50 mg/L concentration upregulates immune genes and blood-testis barrier constituent proteins of bovine Sertoli cells.
Collapse
Affiliation(s)
- E O Adegoke
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Xue Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Hao Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Chen Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Han Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Guixue Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China.
| |
Collapse
|
23
|
S100B promotes microglia M1 polarization and migration to aggravate cerebral ischemia. Inflamm Res 2018; 67:937-949. [DOI: 10.1007/s00011-018-1187-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/05/2018] [Accepted: 09/14/2018] [Indexed: 01/20/2023] Open
|
24
|
Vallée A, Vallée JN, Guillevin R, Lecarpentier Y. Interactions Between the Canonical WNT/Beta-Catenin Pathway and PPAR Gamma on Neuroinflammation, Demyelination, and Remyelination in Multiple Sclerosis. Cell Mol Neurobiol 2018; 38:783-795. [PMID: 28905149 PMCID: PMC11482031 DOI: 10.1007/s10571-017-0550-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/09/2017] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is marked by neuroinflammation and demyelination with loss of oligodendrocytes in the central nervous system. The immune response is regulated by WNT/beta-catenin pathway in MS. Activated NF-kappaB, a major effector of neuroinflammation, and upregulated canonical WNT/beta-catenin pathway positively regulate each other. Demyelinating events present an upregulation of WNT/beta-catenin pathway, whereas proper myelinating phases show a downregulation of WNT/beta-catenin pathway essential for the promotion of oligodendrocytes precursors cells proliferation and differentiation. The activation of WNT/beta-catenin pathway results in differentiation failure and impairment in remyelination. However, PI3K/Akt pathway and TCF7L2, two downstream targets of WNT/beta-catenin pathway, are upregulated and promote proper remyelination. The interactions of these signaling pathways remain unclear. PPAR gamma activation can inhibit NF-kappaB, and can also downregulate the WNT/beta-catenin pathway. PPAR gamma and canonical WNT/beta-catenin pathway act in an opposite manner. PPAR gamma agonists appear as a promising treatment for the inhibition of demyelination and the promotion of proper remyelination through the control of both NF-kappaB activity and canonical WNT/beta-catenin pathway.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France.
- Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France.
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France
- CHU Amiens Picardie, University of Picardie Jules Verne (UPJV), Amiens, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, University of Poitiers et CHU de Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| |
Collapse
|
25
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Demyelination in Multiple Sclerosis: Reprogramming Energy Metabolism and Potential PPARγ Agonist Treatment Approaches. Int J Mol Sci 2018; 19:ijms19041212. [PMID: 29659554 PMCID: PMC5979570 DOI: 10.3390/ijms19041212] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/20/2022] Open
Abstract
Demyelination in multiple sclerosis (MS) cells is the site of several energy metabolic abnormalities driven by dysregulation between the opposed interplay of peroxisome proliferator-activated receptor γ (PPARγ) and WNT/β-catenin pathways. We focus our review on the opposing interactions observed in demyelinating processes in MS between the canonical WNT/β-catenin pathway and PPARγ and their reprogramming energy metabolism implications. Demyelination in MS is associated with chronic inflammation, which is itself associated with the release of cytokines by CD4+ Th17 cells, and downregulation of PPARγ expression leading to the upregulation of the WNT/β-catenin pathway. Upregulation of WNT/β-catenin signaling induces activation of glycolytic enzymes that modify their energy metabolic behavior. Then, in MS cells, a large portion of cytosolic pyruvate is converted into lactate. This phenomenon is called the Warburg effect, despite the availability of oxygen. The Warburg effect is the shift of an energy transfer production from mitochondrial oxidative phosphorylation to aerobic glycolysis. Lactate production is correlated with increased WNT/β-catenin signaling and demyelinating processes by inducing dysfunction of CD4+ T cells leading to axonal and neuronal damage. In MS, downregulation of PPARγ decreases insulin sensitivity and increases neuroinflammation. PPARγ agonists inhibit Th17 differentiation in CD4+ T cells and then diminish release of cytokines. In MS, abnormalities in the regulation of circadian rhythms stimulate the WNT pathway to initiate the demyelination process. Moreover, PPARγ contributes to the regulation of some key circadian genes. Thus, PPARγ agonists interfere with reprogramming energy metabolism by directly inhibiting the WNT/β-catenin pathway and circadian rhythms and could appear as promising treatments in MS due to these interactions.
Collapse
Affiliation(s)
- Alexandre Vallée
- Délégation à la Recherche Clinique et à l'Innovation (DRCI), Hôpital Foch, 92150 Suresnes, France.
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), 77100 Meaux, France.
| | - Rémy Guillevin
- Data Analysis and Computations Through Imaging Modeling-Mathématiques (DACTIM), Unité mixte de recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7348 (Laboratoire de Mathématiques et Application), University of Poitiers, Centre Hospitalier Universitaire (CHU) de Poitiers, 86000 Poitiers, France.
| | - Jean-Noël Vallée
- Centre Hospitalier Universitaire (CHU) Amiens Picardie, University of Picardie Jules Verne (UPJV), 80000 Amiens, France.
- LMA (Laboratoire de Mathématiques et Applications), Unité mixte de recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7348, Université de Poitiers, 86000 Poitiers, France.
| |
Collapse
|
26
|
Werion A, Joris V, Hepp M, Papasokrati L, Marique L, de Ville de Goyet C, Van Regemorter V, Mourad M, Lengelé B, Daumerie C, Marbaix E, Brichard S, Many MC, Craps J. Pioglitazone, a PPARγ Agonist, Upregulates the Expression of Caveolin-1 and Catalase, Essential for Thyroid Cell Homeostasis: A Clue to the Pathogenesis of Hashimoto's Thyroiditis. Thyroid 2016; 26:1320-31. [PMID: 27324467 DOI: 10.1089/thy.2015.0625] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor γ (PPARγ) is a transcription factor that regulates the expression of multiple target genes involved in several metabolic pathways as well as in inflammation. The expression and cell localization of caveolin-1 (Cav-1), thyroperoxidase (TPO), and dual oxidase (DUOX), involved in extracellular iodination, is modulated by Th1 cytokines in human normal thyroid cells and in Hashimoto's thyroiditis (HT). OBJECTIVES The objectives of this study were (i) to analyze the PPARγ protein and mRNA expression at the follicular level in HT versus controls in correlation with the one of Cav-1; (ii) to study the effects of Th1 cytokines on PPARγ and catalase expression in human thyrocyte primary cultures; and (iii) to study the effects of pioglitazone, a PPARγ agonist, on thyroxisome components (Cav-1, TPO, DUOX) and on catalase, involved in antioxidant defense. RESULTS Although the global expression of PPARγ in the whole gland of patients with HT was not modified compared with controls, there was great heterogeneity among glands and among follicles within the same thyroid. Besides normal (type 1) follicles, there were around inflammatory zones, hyperactive (type 2) follicles with high PPARγ and Cav-1 expression, and inactive (type 3) follicles which were unable to form thyroxine and did not express PPARγ or Cav-1. In human thyrocytes in primary culture, Th1 cytokines decreased PPARγ and catalase expression; pioglitazone increased Cav-1, TPO, and catalase expression. CONCLUSION PPARγ may play a central role in normal thyroid physiology by upregulating Cav-1, essential for the organization of the thyroxisome and extracellular iodination. By upregulating catalase, PPARγ may also contribute to cell homeostasis. The inhibitory effect of Th1 cytokines on PPARγ expression may be considered as a new pathogenetic mechanism for HT, and the use of PPARγ agonists could open a new therapeutic approach.
Collapse
Affiliation(s)
- Alexis Werion
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Virginie Joris
- 2 de Pharmacologie et Thérapeutique, et, Université Catholique de Louvain , Brussels, Belgium
| | - Michael Hepp
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Lida Papasokrati
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Lancelot Marique
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | | | | | - Michel Mourad
- 3 de Chirurgie, et, Université Catholique de Louvain , Brussels, Belgium
| | - Benoit Lengelé
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Chantal Daumerie
- 4 Départements d'Endocrinologie, Université Catholique de Louvain , Brussels, Belgium
| | - Etienne Marbaix
- 5 d'Anatomo-pathologie, Secteur des Sciences de la Santé, Faculté de Médecine, Université Catholique de Louvain , Brussels, Belgium
| | - Sonia Brichard
- 4 Départements d'Endocrinologie, Université Catholique de Louvain , Brussels, Belgium
- 6 d'Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain , Brussels, Belgium
| | | | - Julie Craps
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| |
Collapse
|
27
|
Lloyd AF, Miron VE. Cellular and Molecular Mechanisms Underpinning Macrophage Activation during Remyelination. Front Cell Dev Biol 2016; 4:60. [PMID: 27446913 PMCID: PMC4914869 DOI: 10.3389/fcell.2016.00060] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/06/2016] [Indexed: 12/13/2022] Open
Abstract
Remyelination is an example of central nervous system (CNS) regeneration, whereby myelin is restored around demyelinated axons, re-establishing saltatory conduction and trophic/metabolic support. In progressive multiple sclerosis, remyelination is limited or fails altogether which is considered to contribute to axonal damage/loss and consequent disability. Macrophages have critical roles in both CNS damage and regeneration, such as remyelination. This diverse range in functions reflects the ability of macrophages to acquire tissue microenvironment-specific activation states. This activation is dynamically regulated during efficient regeneration, with a switch from pro-inflammatory to inflammation-resolution/pro-regenerative phenotypes. Although, some molecules and pathways have been implicated in the dynamic activation of macrophages, such as NFκB, the cellular and molecular mechanisms underpinning plasticity of macrophage activation are unclear. Identifying mechanisms regulating macrophage activation to pro-regenerative phenotypes may lead to novel therapeutic strategies to promote remyelination in multiple sclerosis.
Collapse
Affiliation(s)
- Amy F Lloyd
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh Edinburgh, UK
| | - Veronique E Miron
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh Edinburgh, UK
| |
Collapse
|
28
|
Kopitar-Jerala N. Innate Immune Response in Brain, NF-Kappa B Signaling and Cystatins. Front Mol Neurosci 2015; 8:73. [PMID: 26696821 PMCID: PMC4673337 DOI: 10.3389/fnmol.2015.00073] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/16/2015] [Indexed: 12/29/2022] Open
Abstract
Recently several reports have demonstrated that innate immune response and inflammation have an important role in major neurodegenerative diseases. The activation of the NF-κB family of transcription factors is a key step in the regulation of pro inflammatory cytokine expression. Microglia and other cell types in the brain can be activated in response to endogenous danger molecules as well as aggregated proteins and brain injury. During the past couple of years several studies reported the role of cystatins in neuroinflammation and neurodegeneration. In the present review, I will summarize and analyze recent findings regarding the role of cystatins in inflammation and NF-κB activation. Type I cystatin stefin B (cystatin B) is an endogenous cysteine cathepsin inhibitor localized in the cytosol, mitochondria and nucleus. Mutations in the gene of stefin B are associated with the neurodegenerative disease known as Unverricht-Lundborg disease and microglial activation plays an important role in the pathogenesis of the disease. Stefin B deficient mice have increased caspase-11 expression and secreted higher amounts of pro-inflammatory cytokines. The increased caspase-11 gene expression, was a consequence of increased NF-κB activation.
Collapse
Affiliation(s)
- Nataša Kopitar-Jerala
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute Ljubljana, Slovenia
| |
Collapse
|
29
|
α-TLR2 antibody attenuates the Aβ-mediated inflammatory response in microglia through enhanced expression of SIGIRR. Brain Behav Immun 2015; 46:70-9. [PMID: 25620736 DOI: 10.1016/j.bbi.2015.01.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 01/11/2023] Open
Abstract
The immunoregulatory function of single-Ig-interleukin-1 related receptor (SIGIRR) is derived from its ability to constrain the inflammatory consequences of interleukin (IL)-1R and toll-like receptor (TLR)4 activation. This role extends to the brain, where SIGIRR deficiency increases the synaptic and cognitive dysfunction associated with IL-1R- and TLR4-mediated signalling. The current study set out to investigate the interaction between SIGIRR and TLR2 in brain tissue and the data demonstrate that the response to the TLR2 agonist, Pam3CysSK4 (Pam3Cys4), is enhanced in glial cells from SIGIRR(-/-) animals. Consistent with the view that β-amyloid peptide (Aβ) signals through activation of TLR2, the data also show that Aβ-induced changes are exaggerated in glia from SIGIRR(-/-) animals. We report that microglia, rather than astrocytes, are the primary glial cell expressing both TLR2 and SIGIRR. While Aβ increased TLR2 expression, it decreased SIGIRR expression in microglia. This was mimicked by direct activation of TLR2 with Pam3Cys4. We investigated the effect of an anti-TLR2 antibody (αTLR2) on the Aβ-induced inflammatory responses and demonstrate that it prevented the expression and release of the pro-inflammatory cytokines TNFα and IL-6 from microglia. In addition, application of αTLR2 alleviated the Aβ-mediated impairment in long-term potentiation (LTP) of hippocampal synaptic activity. The protective effects of αTLR2 were accompanied by an up-regulation in SIGIRR expression. We propose that a mechanism involving activation of PI3 kinase/Akt and the transcription factor peroxisome proliferator-activated receptor (PPAR)γ may facilitate this increase in SIGIRR. These findings highlight a novel role of SIGIRR as a negative regulator of TLR2-mediated inflammation in the brain.
Collapse
|
30
|
Nakagawa Y, Chiba K. Role of microglial m1/m2 polarization in relapse and remission of psychiatric disorders and diseases. Pharmaceuticals (Basel) 2014; 7:1028-48. [PMID: 25429645 PMCID: PMC4276905 DOI: 10.3390/ph7121028] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/27/2022] Open
Abstract
Psychiatric disorders such as schizophrenia and major depressive disorder were thought to be caused by neurotransmitter abnormalities. Patients with these disorders often experience relapse and remission; however the underlying molecular mechanisms of relapse and remission still remain unclear. Recent advanced immunological analyses have revealed that M1/M2 polarization of macrophages plays an important role in controlling the balance between promotion and suppression in inflammation. Microglial cells share certain characteristics with macrophages and contribute to immune-surveillance in the central nervous system (CNS). In this review, we summarize immunoregulatory functions of microglia and discuss a possible role of microglial M1/M2 polarization in relapse and remission of psychiatric disorders and diseases. M1 polarized microglia can produce pro-inflammatory cytokines, reactive oxygen species, and nitric oxide, suggesting that these molecules contribute to dysfunction of neural network in the CNS. Alternatively, M2 polarized microglia express cytokines and receptors that are implicated in inhibiting inflammation and restoring homeostasis. Based on these aspects, we propose a possibility that M1 and M2 microglia are related to relapse and remission, respectively in psychiatric disorders and diseases. Consequently, a target molecule skewing M2 polarization of microglia may provide beneficial therapies for these disorders and diseases in the CNS.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Research Strategy and Planning Department, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| | - Kenji Chiba
- Advanced Medical Research Laboratories, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| |
Collapse
|
31
|
Ciurleo R, Bramanti P, Marino S. Role of statins in the treatment of multiple sclerosis. Pharmacol Res 2014; 87:133-43. [PMID: 24657241 DOI: 10.1016/j.phrs.2014.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/10/2014] [Accepted: 03/11/2014] [Indexed: 01/22/2023]
Abstract
Statins as inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase are widely prescribed for hypercholesterolemia treatment. In the last years, statins have also been shown to exert immunomodulatory and anti-inflammatory effects which appear to be related to inhibition of isoprenylation of small GTP-binding proteins and, at least in part, independent of their cholesterol-lowering effects. These "pleiotropic" effects make statins an attractive treatment option for immune-mediated disorders such as multiple sclerosis. Studies in vitro and in experimental autoimmune encephalomyelitis animal model seem to support not only the efficacy of statins as immunomodulatory agents but also their potential neuroprotective properties, although the exact mechanism with which statins exert these effects has not yet been fully understood. The immunomodulatory, anti-inflammatory and neuroprotective properties of statins provided the incentive for several clinical trials in multiple sclerosis, in which they were tested not only as mono-therapy but also in combination with interferon-β. However, the attempt to translate the results of animal model studies in humans produced conflicting results. Further large, prospective, randomized, double-blind, placebo-controlled trials, designed to evaluate the long-term effects of statins alone or in add-on to other disease-modifying therapies, are needed to support their routine clinical use in multiple sclerosis.
Collapse
Affiliation(s)
| | | | - Silvia Marino
- IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy; Department of Biomedical Sciences and Morphological and Functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
32
|
Szalardy L, Zadori D, Tanczos E, Simu M, Bencsik K, Vecsei L, Klivenyi P. Elevated levels of PPAR-gamma in the cerebrospinal fluid of patients with multiple sclerosis. Neurosci Lett 2013; 554:131-4. [PMID: 24021801 DOI: 10.1016/j.neulet.2013.08.069] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/21/2013] [Accepted: 08/28/2013] [Indexed: 11/19/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), a ligand-activated transcriptional factor involved in the regulation of glucose and lipid metabolism, has gained interest as a potential therapeutic target in multiple sclerosis (MS) due to its potent immunoregulatory properties and the therapeutic efficacy of its ligands in experimental autoimmune encephalitis (EAE). Elevated expression of PPARγ has been observed in the spinal cord of EAE mice and in an in vitro model of antigen-induced demyelination; however, no reports have yet been available on the PPARγ status in the central nervous system of human individuals with MS. Aiming to identify a possible alteration, the present study assessed the levels of PPARγ protein in the cerebrospinal fluid (CSF) of MS patients via ELISA technique. We report a pronounced elevation in the CSF levels of PPARγ in MS patients (n=35) compared to non-inflammatory controls (n=22). This elevation was independent of blood-CSF barrier integrity, but correlated with CSF white blood cell count and IgG index, associating the observed elevation with neuroinflammation. Controlling for potential confounders, the CSF levels of PPARγ further displayed a moderate but significant association with clinical severity. Corroborating with prior experimental findings, these results may contribute to our understanding about the role of PPARγ in MS, and may implicate this protein as a potential CSF biomarker of the disease.
Collapse
Affiliation(s)
- Levente Szalardy
- Department of Neurology, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | | | | | | | | | | | | |
Collapse
|
33
|
D'Aversa TG, Eugenin EA, Lopez L, Berman JW. Myelin basic protein induces inflammatory mediators from primary human endothelial cells and blood-brain barrier disruption: implications for the pathogenesis of multiple sclerosis. Neuropathol Appl Neurobiol 2013; 39:270-83. [PMID: 22524708 DOI: 10.1111/j.1365-2990.2012.01279.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIM Multiple sclerosis (MS) is an autoimmune disease of the central nervous system, characterized by demyelination of white matter, loss of myelin forming oligodendrocytes, changes in the blood-brain barrier (BBB) and leucocyte infiltration. Myelin basic protein (MBP) is a component of the myelin sheath. Degradation of myelin is believed to be an important step that leads to MS pathology. Transmigration of leucocytes across the vasculature, and a compromised BBB participate in the neuroinflammation of MS. We examined the expression and regulation of the chemokine (C-C motif) ligand 2 (CCL2) and the cytokine interleukin-6 (IL-6) in human endothelial cells (EC), a component of the BBB, after treatment with MBP. METHODS EC were treated with full-length MBP. CCL2 and IL-6 protein were determined by ELISA. Western blot analysis was used to determine signalling pathways. A BBB model was treated with MBP and permeability was assayed using albumin conjugated to Evan's blue dye. The levels of the tight junction proteins occludin and claudin-1, and matrix metalloprotease (MMP)-2 were assayed by Western blot. RESULTS MBP significantly induced CCL2 and IL-6 protein from EC. This induction was partially mediated by the p38 MAPK pathway as there was phosphorylation after MBP treatment. MBP treatment of a BBB model caused an increase in permeability that correlated with a decrease in occludin and claudin-1, and an induction of MMP2. CONCLUSION These data demonstrate that MBP induces chemotactic and inflammatory mediators. MBP also alters BBB permeability and tight junction expression, indicating additional factors that may contribute to the BBB breakdown characteristic of MS.
Collapse
Affiliation(s)
- T G D'Aversa
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | |
Collapse
|
34
|
α-Lipoic acid enhances endogenous peroxisome-proliferator-activated receptor-γ to ameliorate experimental autoimmune encephalomyelitis in mice. Clin Sci (Lond) 2013; 125:329-40. [PMID: 23550596 DOI: 10.1042/cs20120560] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
ALA (α-lipoic acid) is a natural, endogenous antioxidant that acts as a PPAR-γ (peroxisome-proliferator-activated receptor-γ) agonist to counteract oxidative stress. Thus far, the antioxidative and immunomodulatory effects of ALA on EAE (experimental autoimmune encephalomyelitis) are not well understood. In this study, we found that ALA restricts the infiltration of inflammatory cells into the CNS (central nervous system) in MOG (myelin oligodendrocyte glycoprotein)-EAE mice, thus reducing the disease severity. In addition, we revealed that ALA significantly suppresses the number and percentage of encephalitogenic Th1 and Th17 cells and increases splenic Treg-cells (regulatory T-cells). Strikingly, we further demonstrated that ALA induces endogenous PPAR-γ centrally and peripherally but has no effect on HO-1 (haem oxygenase 1). Together, these data suggest that ALA can up-regulate endogenous systemic and central PPAR-γ and enhance systemic Treg-cells to inhibit the inflammatory response and ameliorate MOG-EAE. In conclusion, our data provide the first evidence that ALA can augment the production of PPAR-γ in vivo and modulate adaptive immunity both centrally and peripherally in EAE and may reveal further antioxidative and immunomodulatory mechanisms for the application of ALA in human MS (multiple sclerosis).
Collapse
|
35
|
Paintlia AS, Paintlia MK, Singh AK, Singh I. Modulation of Rho-Rock signaling pathway protects oligodendrocytes against cytokine toxicity via PPAR-α-dependent mechanism. Glia 2013; 61:1500-1517. [PMID: 23839981 DOI: 10.1002/glia.22537] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/06/2013] [Accepted: 05/07/2013] [Indexed: 12/13/2022]
Abstract
We earlier documented that lovastatin (LOV)-mediated inhibition of small Rho GTPases activity protects vulnerable oligodendrocytes (OLs) in mixed glial cell cultures stimulated with Th1 cytokines and in a murine model of multiple sclerosis (MS). However, the precise mechanism of OL protection remains unclear. We here employed genetic and biochemical approaches to elucidate the underlying mechanism that protects LOV treated OLs from Th1 (tumor necrosis factor-α) and Th17 (interleukin-17) cytokines toxicity in in vitro. Cytokines enhanced the reactive oxygen species (ROS) generation and mitochondrial membrane depolarization with corresponding lowering of glutathione (reduced) level in OLs and that were reverted by LOV. In addition, the expression of ROS detoxifying enzymes (catalase and superoxide-dismutase 2) and the transactivation of peroxisome proliferators-activated receptor (PPAR)-α/-β/-γ including PPAR-γ coactivator-1α were enhanced by LOV in similarly treated OLs. Interestingly, LOV-mediated inhibition of small Rho GTPases, i.e., RhoA and cdc42, and Rho-associated kinase (ROCK) activity enhanced the levels of PPAR ligands in OLs via extracellular signal regulated kinase (1/2)/p38 mitogen-activated protein kinase/cytoplasmic phospholipase 2/cyclooxygenase-2 signaling cascade activation. Small hairpin RNA transfection-based studies established that LOV mainly enhances PPAR-α and less so of PPAR-β and PPAR-γ transactivation that enhances ROS detoxifying defense in OLs. In support of this, the observed LOV-mediated protection was lacking in PPAR-α-deficient OLs exposed to cytokines. Collectively, these data provide unprecedented evidence that LOV-mediated inhibition of the Rho-ROCK signaling pathway boosts ROS detoxifying defense in OLs via PPAR-α-dependent mechanism that has implication in neurodegenerative disorders including MS.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina
| | - Manjeet K Paintlia
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Ralph H. Johnson VA Medical Center, Charleston, South Carolina
| | - Inderjit Singh
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina
| |
Collapse
|
36
|
Paintlia AS, Paintlia MK, Mohan S, Singh AK, Singh I. AMP-activated protein kinase signaling protects oligodendrocytes that restore central nervous system functions in an experimental autoimmune encephalomyelitis model. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:526-41. [PMID: 23759513 DOI: 10.1016/j.ajpath.2013.04.030] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 04/22/2013] [Accepted: 04/29/2013] [Indexed: 12/12/2022]
Abstract
AMP-activated protein kinase (AMPK) signaling is reported to protect neurons under pathologic conditions; however, its effect on oligodendrocytes (OLs) remains to be elucidated. We investigated whether AMPK signaling protects OLs to restore central nervous system (CNS) functions in an experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis. Increased inflammation and demyelination in the CNS and peripheral immune responses were consistent with the observed clinical impairments in EAE animals, which were attenuated by treatment with metformin compared with vehicle. In addition, expressions of neurotrophic factors and of signatory genes of OL lineages were increased in the CNS of metformin-treated EAE animals. Likewise, metformin attenuated inflammatory response and enhanced expressions of neurotrophic factors, thereby protecting OLs via AMPK activation in mixed glial cultures stimulated with lipopolysaccharide/interferon γ in vitro, as evidenced by analysis of the expression of signatory genes of O1(+)/MBP(+) OLs and their cellular populations. Metformin also attenuated oxidative stress and malondialdehyde-containing protein levels, with corresponding induction of antioxidative defenses in OLs exposed to cytokines via AMPK activation. These effects of metformin were evident in the CNS of EAE animals. These data provide evidence that AMPK signaling is crucial to protect OLs and, thus, CNS functions in EAE animals. We conclude that AMPK activators, including metformin, have the potential to limit neurologic deficits in multiple sclerosis and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
37
|
Cao L, He C. Polarization of macrophages and microglia in inflammatory demyelination. Neurosci Bull 2013; 29:189-98. [PMID: 23558588 DOI: 10.1007/s12264-013-1324-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 02/06/2013] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system, and microglia and macrophages play important roles in its pathogenesis. The activation of microglia and macrophages accompanies disease development, whereas depletion of these cells significantly decreases disease severity. Microglia and macrophages usually have diverse and plastic phenotypes. Both pro-inflammatory and antiinflammatory microglia and macrophages exist in MS and its animal model, experimental autoimmune encephalomyelitis. The polarization of microglia and macrophages may underlie the differing functional properties that have been reported. in this review, we discuss the responses and polarization of microglia and macrophages in MS, and their effects on its pathogenesis and repair. Harnessing their beneficial effects by modulating their polarization states holds great promise for the treatment of inflammatory demyelinating diseases.
Collapse
Affiliation(s)
- Li Cao
- Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of the Ministry of Education, Neuroscience Research Center of Changzheng Hospital, Second Military Medical University, Shanghai 200433, China.
| | | |
Collapse
|
38
|
Poulet B, Ulici V, Stone TC, Pead M, Gburcik V, Constantinou E, Palmer DB, Beier F, Timmons JA, Pitsillides AA. Time-series transcriptional profiling yields new perspectives on susceptibility to murine osteoarthritis. ACTA ACUST UNITED AC 2013; 64:3256-66. [PMID: 22833266 DOI: 10.1002/art.34572] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Chronological age is a powerful epidemiologic risk factor for osteoarthritis (OA), a multifactorial disease that is characterized by articular cartilage (AC) degradation. It is unclear from a molecular perspective how aging interacts with OA to produce this risk to AC integrity. To address this key question, we used in vivo time-course analysis of OA development and murine interstrain variability in natural susceptibility to OA to examine changes in non-OA-prone CBA mice versus OA-prone STR/Ort mice, which develop disease that bears significant histologic resemblance to human OA. Through global transcriptome profiling, we attempted to discover the molecular signature linked with both OA vulnerability and progression. METHODS Affymetrix Mouse Gene 1.0 ST Array profiles were generated from AC samples derived from CBA and STR/Ort mice at 3 different ages, corresponding to the stages prior to, at, and late after the natural onset of OA in the STR/Ort mice. RESULTS We found that the OA in STR/Ort mice exhibited a molecular phenotype resembling human OA, and we pinpointed a central role of NF-κB signaling and the emergence of an immune-related signature in OA cartilage over time. We discovered that, strikingly, young healthy AC has a highly expressed skeletal muscle gene expression program, which is switched off during maturation, but is intriguingly retained in AC during OA development in STR/Ort mice. CONCLUSION This study is the first to show that AC chondrocytes share a high-abundance gene-expression program with skeletal muscle. We show that failure to switch this program off, as well as the restoration of this program, is associated with inappropriate expression of NF-κB signaling pathways, skeletal muscle-related genes, and induction and/or progression of OA.
Collapse
|
39
|
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
|
40
|
Paintlia MK, Paintlia AS, Singh AK, Singh I. S-nitrosoglutathione induces ciliary neurotrophic factor expression in astrocytes, which has implications to protect the central nervous system under pathological conditions. J Biol Chem 2012; 288:3831-43. [PMID: 23264628 DOI: 10.1074/jbc.m112.405654] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence suggests that reactive astrogliosis has beneficial and detrimental outcomes in various CNS disorders, but the mechanism behind this dichotomy is unclear. Recent advances in this direction suggested that NO signaling is critical to regulate the outcomes of reactive astrogliosis in vivo. Using biochemical and genetic approaches, we here investigated the effect of S-nitrosoglutathione (GSNO; a physiological NO donor) in astrocytes in vitro settings. GSNO enhanced the expressions of glial fibrillary acidic protein and neurotrophic factors including ciliary neurotrophic factor (CNTF) in astrocytes in a dose-dependent manner. The enhanced CNTF expression in GSNO-treated astrocytes was ascribed to NO-mediated sGC/cGMP/PKG signaling. It was associated with p38 MAPK-dependent increased peroxisome proliferator-activated receptor-γ transactivation. In addition, the chromatin accessibility of peroxisome proliferator-activated receptor-γ accompanied with ATF2 and CREB (cAMP-response element-binding protein) was enhanced across the CNTF gene promoter in GSNO treated astrocytes. Interestingly, secreted CNTF was responsible for increased expression of glial fibrillary acidic protein in GSNO-treated astrocytes in an autocrine manner via a JAK2- and STAT3-dependent mechanism. In addition, CNTF secreted by GSNO-treated astrocytes enhanced the differentiation of immature oligodendrocytes in vitro. These effects of GSNO were consistent with an endogenously produced NO in astrocytes stimulated with proinflammatory cytokines in vitro. We conclude that NO signaling induces CNTF expression in astrocytes that favors the beneficial outcomes of reactive astrogliosis in vivo. Our data suggest that the endogenously produced NO or its exogenous source has potential to modulate the outcomes of reactive astrogliosis to protect CNS under pathological conditions.
Collapse
Affiliation(s)
- Manjeet K Paintlia
- Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | |
Collapse
|
41
|
Regulation of avoidant behaviors and pain by the anti-inflammatory tyrosine phosphatase SHP-1. ACTA ACUST UNITED AC 2012; 2:235-46. [PMID: 18250891 DOI: 10.1017/s1740925x07000476] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The protein tyrosine phosphatase SHP-1 is a critical regulator of cytokine signaling and inflammation. Mice homozygous for a null allele at the SHP-1 locus have a phenotype of severe inflammation and are hyper-responsive to the TLR4 ligand LPS. TLR4 stimulation in the CNS has been linked to both neuropathic pain and sickness behaviors. To determine if reduction in SHP-1 expression affects LPS-induced behaviors, responses of heterozygous SHP-1-deficient (me/+) and wild-type (+/+) mice to LPS were measured. Chronic (4-week) treatment with LPS induced avoidant behaviors indicative of fear/anxiety in me/+, but not +/+, mice. These behaviors were correlated with a LPS-induced type 2 cytokine, cytokine receptor, and immune effector arginase profile in the brains of me/+ mice not found in +/+ mice. Me/+ mice also had a constitutively greater level of TLR4 in the CNS than +/+ mice. Additionally, me/+ mice displayed constitutively increased thermal sensitivity compared to +/+ mice, measured by the tail-flick test. Moreover, me/+ glial cultures were more responsive to LPS than +/+ glia. Therefore, the reduced expression of SHP-1 in me/+ imparts haploinsufficiency with respect to the control of CNS TLR4 and pain signaling. Furthermore, type 2 cytokines become prevalent during chronic TLR4 hyperstimulation in the CNS and are associated positively with behaviors that are usually linked to type 1 pro-inflammatory cytokines. These findings question the notion that type 2 immunity is solely anti-inflammatory in the CNS and indicate that type 2 immunity induces/potentiates CNS inflammatory processes.
Collapse
|
42
|
Uderhardt S, Krönke G. 12/15-lipoxygenase during the regulation of inflammation, immunity, and self-tolerance. J Mol Med (Berl) 2012; 90:1247-56. [PMID: 22983484 DOI: 10.1007/s00109-012-0954-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 08/22/2012] [Accepted: 08/27/2012] [Indexed: 12/20/2022]
Abstract
12/15-Lipoxygenase (12/15-LO) catalyzes the oxidation of free and esterified fatty acids thereby generating a whole spectrum of bioactive lipid mediators. This enzyme is involved in the regulation of various homeostatic processes as well as in the pathogenesis of multiple diseases. During the innate and adaptive immune response, 12/15-LO and its products exert both pro- and anti-inflammatory effects. Likewise, this enzyme has been implicated in the pathogenesis of autoimmune disease as well as in the maintenance of self-tolerance. This review will summarize our current knowledge about the role of 12/15-LO and will try to examine the two faces of this enzyme within the context of inflammation and immunity.
Collapse
Affiliation(s)
- Stefan Uderhardt
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | | |
Collapse
|
43
|
Zuo X, Peng Z, Wu Y, Moussalli MJ, Yang XL, Wang Y, Parker-Thornburg J, Morris JS, Broaddus RR, Fischer SM, Shureiqi I. Effects of gut-targeted 15-LOX-1 transgene expression on colonic tumorigenesis in mice. J Natl Cancer Inst 2012; 104:709-16. [PMID: 22472308 DOI: 10.1093/jnci/djs187] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Expression of 15-lipoxygenase-1 (15-LOX-1) is decreased in many human cancers; however, the mechanistic significance of its decreased expression has been difficult to determine because its mouse homolog 12/15-LOX has opposing functions. We generated a mouse model in which expression of a human 15-LOX-1 transgene was targeted to the intestinal epithelium via the villin promoter. Targeted expression was confirmed by real-time reverse transcription-polymerase chain reaction and immunoblotting. When the 15-LOX-1 transgene was expressed in colonic epithelial cells of two independent mouse lines (B6 and FVB), azoxymethane-inducible colonic tumorigenesis was suppressed (mean number of tumors: wild type [WT] = 8.2, 15-LOX-1(+/-) = 4.91, 15-LOX-1(+/+) = 3.57; WT vs 15-LOX-1(+/-) two-sided P = .003, WT vs 15-LOX-1(+/+) two-sided P < .001; n = 10-14 mice per group). 15-LOX-1 transgene expression was always decreased in the tumors that did develop. In the presence of expression of the 15-LOX-1 transgene, expression of tumor necrosis factor alpha and its target inducible nitric oxide synthase were decreased and activation of nuclear factor-kappa B in colonic epithelial cells was inhibited.
Collapse
Affiliation(s)
- Xiangsheng Zuo
- Department of Clinical Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Corthals AP. Multiple sclerosis is not a disease of the immune system. QUARTERLY REVIEW OF BIOLOGY 2012; 86:287-321. [PMID: 22384749 DOI: 10.1086/662453] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Multiple sclerosis is a complex neurodegenerative disease, thought to arise through autoimmunity against antigens of the central nervous system. The autoimmunity hypothesis fails to explain why genetic and environmental risk factors linked to the disease in one population tend to be unimportant in other populations. Despite great advances in documenting the cell and molecular mechanisms underlying MS pathophysiology, the autoimmunity framework has also been unable to develop a comprehensive explanation of the etiology of the disease. I propose a new framework for understanding MS as a dysfunction of the metabolism of lipids. Specifically, the homeostasis of lipid metabolism collapses during acute-phase inflammatory response triggered by a pathogen, trauma, or stress, starting a feedback loop of increased oxidative stress, inflammatory response, and proliferation of cytoxic foam cells that cross the blood brain barrier and both catabolize myelin and prevent remyelination. Understanding MS as a chronic metabolic disorder illuminates four aspects of disease onset and progression: 1) its pathophysiology; 2) genetic susceptibility; 3) environmental and pathogen triggers; and 4) the skewed sex ratio of patients. It also suggests new avenues for treatment.
Collapse
Affiliation(s)
- Angelique P Corthals
- Department of Sciences, John Jay College of Criminal Justice, City University of New York New York, New York 10019, USA.
| |
Collapse
|
45
|
Regulation of Glial Cell Functions by PPAR-gamma Natural and Synthetic Agonists. PPAR Res 2011; 2008:864140. [PMID: 18464925 PMCID: PMC2367430 DOI: 10.1155/2008/864140] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 03/12/2008] [Indexed: 11/18/2022] Open
Abstract
In the recent years, the peroxisome proliferator-activated receptor-γ (PPAR-γ), a well known target for type II diabetes treatment, has received an increasing attention for its therapeutic potential in inflammatory and degenerative brain disorders. PPAR-γ agonists, which include naturally occurring compounds (such as long chain fatty acids and the cyclopentenone prostaglandin 15-deoxy Δ12,14 prostaglandin J2), and synthetic agonists (among which the thiazolidinediones and few nonsteroidal anti-inflammatory drugs) have shown anti-inflammatory and protective effects in several experimental models of Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, multiple sclerosis and stroke, as well as in few clinical studies. The pleiotropic effects of PPAR-γ agonists are likely to be mediated by several mechanisms involving anti-inflammatory activities on peripheral immune cells (macrophages and lymphocytes), as well as direct effects on neural cells including cerebral vascular endothelial cells, neurons, and glia. In the present article, we will review the recent findings supporting a major role for PPAR-γ agonists in controlling neuroinflammation and neurodegeneration through their activities on glial cells, with a particular emphasis on microglial cells as major macrophage population of the brain parenchyma and main actors in brain inflammation.
Collapse
|
46
|
PPAR-gamma: Therapeutic Potential for Multiple Sclerosis. PPAR Res 2011; 2008:627463. [PMID: 18604287 PMCID: PMC2441778 DOI: 10.1155/2008/627463] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Accepted: 06/02/2008] [Indexed: 01/04/2023] Open
Abstract
The role of peroxisome proliferator-activated receptors (PPARs) in altering lipid and glucose metabolism is well established. More recent studies indicate that PPARs also play critical roles in controlling immune responses. We and others have previously demonstrated that PPAR-γ agonists modulate the development of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). This review will discuss the cellular and molecular mechanisms by which these agonists are believed to modulate disease. The therapeutic potential of PPAR-γ agonists in the treatment of multiple sclerosis will also be considered.
Collapse
|
47
|
Abstract
Mononuclear phagocytes often function as control switches of the immune system, securing the balance between pro- and anti-inflammatory reactions. For this purpose and depending on the activating stimuli, these cells can develop into different subsets: proinflammatory classically activated (M1) or anti-inflammatory alternatively activated (M2) macrophages. The expression of the nuclear peroxisome proliferator-activated receptors (PPARs) is regulated by M1- or M2-inducing stimuli, and these receptors are generally considered to counteract inflammatory M1 macrophages, while actively promoting M2 activation. This is of importance in a tumor context, where M1 are important initiators of inflammation-driven cancers. As a consequence, PPAR agonists are potentially usefull for inhibiting the early phases of tumorigenesis through their antagonistic effect on M1. In more established tumors, the macrophage phenotype is more diverse, making it more difficult to predict the outcome of PPAR agonism. Overall, in our view current knowledge provides a sound basis for the clinical evaluation of PPAR ligands as chemopreventive agents in chronic inflammation-associated cancer development, while cautioning against the unthoughtful application of these agents as cancer therapeutics.
Collapse
|
48
|
Paintlia AS, Paintlia MK, Singh AK, Orak JK, Singh I. Activation of PPAR-γ and PTEN cascade participates in lovastatin-mediated accelerated differentiation of oligodendrocyte progenitor cells. Glia 2011; 58:1669-85. [PMID: 20578043 DOI: 10.1002/glia.21039] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previously, we and others documented that statins including-lovastatin (LOV) promote the differentiation of oligodendrocyte progenitor cells (OPCs) and remyelination in experimental autoimmune encephalomyelitis (EAE), an multiple sclerosis (MS) model. Conversely, some recent studies demonstrated that statins negatively influence oligodendrocyte (OL) differentiation in vitro and remyelination in a cuprizone-CNS demyelinating model. Therefore, herein, we first investigated the cause of impaired differentiation of OLs by statins in vitro settings. Our observations indicated that the depletion of cholesterol was detrimental to LOV treated OPCs under cholesterol/serum-deprived culture conditions similar to that were used in conflicting studies. However, the depletion of geranylgeranyl-pp under normal cholesterol homeostasis conditions enhanced the phenotypic commitment and differentiation of LOV-treated OPCs ascribed to inhibition of RhoA-Rho kinase. Interestingly, this effect of LOV was associated with increased activation and expression of both PPAR-γ and PTEN in OPCs as confirmed by various pharmacological and molecular based approaches. Furthermore, PTEN was involved in an inhibition of OPCs proliferation via PI3K-Akt inhibition and induction of cell cycle arrest at G1 phase, but without affecting their cell survival. These effects of LOV on OPCs in vitro were absent in the CNS of normal rats chronically treated with LOV concentrations used in EAE indicating that PPAR-γ induction in normal brain may be tightly regulated-providing evidences that statins are therapeutically safe for humans. Collectively, these data provide initial evidence that statin-mediated activation of the PPAR-γ-PTEN cascade participates in OL differentiation, thus suggesting new therapeutic-interventions for MS or related CNS-demyelinating diseases.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina, USA
| | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Rosales-Corral S, Reiter RJ, Tan DX, Ortiz GG, Lopez-Armas G. Functional aspects of redox control during neuroinflammation. Antioxid Redox Signal 2010; 13:193-247. [PMID: 19951033 DOI: 10.1089/ars.2009.2629] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neuroinflammation is a CNS reaction to injury in which some severe pathologies, regardless of their origin, converge. The phenomenon emphasizes crosstalk between neurons and glia and reveals a complex interaction with oxidizing agents through redox sensors localized in enzymes, receptors, and transcription factors. When oxidizing pressures cause reversible molecular changes, such as minimal or transitory proinflammatory cytokine overproduction, redox couples provide a means of translating the presence of reactive oxygen or nitrogen species into useful signals in the cell. Additionally, thiol-based redox sensors convey information about localized changes in redox potential induced by physiologic or pathologic situations. They are susceptible to oxidative changes and become key events during neuroinflammation, altering the course of a signaling response or the behavior of specific transcription factors. When oxidative stress augments the pressure on the intracellular environment, the effective reduction potential of redox pairs diminishes, and cell signaling shifts toward proinflammatory and proapoptotic signals, creating a vicious cycle between oxidative stress and neuroinflammation. In addition, electrophilic compounds derived from the oxidative cascade react with key protein thiols and interfere with redox signaling. This article reviews the relevant functional aspects of redox control during the neuroinflammatory process.
Collapse
Affiliation(s)
- Sergio Rosales-Corral
- Lab. Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO) del Instituto Mexicano del Seguro Social (IMSS) , Guadalajara, Jalisco. Mexico.
| | | | | | | | | |
Collapse
|