1
|
García-Salas R, Cilleros-Holgado P, Di Spirito A, Gómez-Fernández D, Piñero-Pérez R, Romero-Domínguez JM, Álvarez-Córdoba M, Reche-López D, Romero-González A, López-Cabrera A, Sánchez-Alcázar JA. Mitochondrial dysfunction, iron accumulation, lipid peroxidation, and inflammasome activation in cellular models derived from patients with multiple sclerosis. Aging (Albany NY) 2025; 17:365-392. [PMID: 39918890 PMCID: PMC11892916 DOI: 10.18632/aging.206198] [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: 09/10/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Despite advancements in managing relapsing active illness, effective treatments for the irreversible progressive decline in MS remain limited. Research employing skin fibroblasts obtained from patients with neurological disorders revealed modifications in cellular stress pathways and bioenergetics. However, research using MS patient-derived cellular models is scarce. In this study, we collected fibroblasts from two MS patients to investigate cellular pathological alterations. We observed that MS fibroblasts showed a senescent morphology associated with iron/lipofuscin accumulation and altered expression of iron metabolism proteins. In addition, we found increased lipid peroxidation and downregulation of antioxidant enzymes expression levels in MS fibroblasts. When challenged against erastin, a ferroptosis inducer, MS fibroblasts showed decreased viability, suggesting increased sensitivity to ferroptosis. Furthermore, MS fibroblasts presented alterations in the expression levels of autophagy-related proteins. Interestingly, these alterations were associated with mitochondrial dysfunction and inflammasome activation. These findings were validated in 7 additional patient-derived cell lines. Our findings suggest that the underlying stress phenotype of MS fibroblasts may be disease-specific and recapitulate the main cellular pathological alterations found in the disease such as mitochondrial dysfunction, iron accumulation, lipid peroxidation, inflammasome activation, and pro-inflammatory cytokine production.
Collapse
Affiliation(s)
- Raquel García-Salas
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Anna Di Spirito
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | | | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Alejandra López-Cabrera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | | |
Collapse
|
2
|
Otálora-Alcaraz A, Reilly T, Oró-Nolla M, Sun MC, Costelloe L, Kearney H, Patra PH, Downer EJ. The NLRP3 inflammasome: A central player in multiple sclerosis. Biochem Pharmacol 2025; 232:116667. [PMID: 39647604 DOI: 10.1016/j.bcp.2024.116667] [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: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune condition associated with many symptoms including spasticity, pain, limb numbness and weakness. It is characterised by inflammatory demyelination and axonal degeneration of the brain and spinal cord. A range of disease-modifying therapies (DMTs) are available to suppress inflammatory disease activity in MS, however, there is a pressing need for new therapeutic avenues as DMTs have a limited ability to suppress confirmed disability progression. A body of literature indicates that innate immune inflammation is linked to MS progression. The nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome has a well-established function in innate immunity which is closely associated with the pathogenesis of neuroinflammatory conditions. Evidence suggests that the inflammasome may be a therapeutic target in disorders such as MS and at present, inhibitors of the NLRP3 inflammasome are in pre-clinical development. Therefore, this review systematically highlights the pathogenic role of inflammasomes in MS, presenting an overview of research evidence linking inflammasome-related polymorphisms to MS susceptibility, and gathering evidence investigating NLRP3 biomarkers in MS. The role of the NLRP3 inflammasome in murine models of MS is furthermore discussed. Finally, a significant component of this review focuses on evidence that NLRP3 signalling components are novel drug targets in MS. Overall this review defines the role of the inflammasome in MS pathogenesis and identifies inflammasome inhibitor targets that warrant full investigation in MS and related disorders.
Collapse
Affiliation(s)
- Almudena Otálora-Alcaraz
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Thomas Reilly
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Martí Oró-Nolla
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Melody Cui Sun
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Lisa Costelloe
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Hugh Kearney
- MS Unit, Department of Neurology, St. James's Hospital, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Pabitra H Patra
- Transpharmation Ltd., London Biosciences Innovation Centre, London, United Kingdom
| | - Eric J Downer
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
3
|
Üremiş N, Üremiş MM. Oxidative/Nitrosative Stress, Apoptosis, and Redox Signaling: Key Players in Neurodegenerative Diseases. J Biochem Mol Toxicol 2025; 39:e70133. [PMID: 39799559 PMCID: PMC11725306 DOI: 10.1002/jbt.70133] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
Neurodegenerative diseases are significant health concerns that have a profound impact on the quality and duration of life for millions of individuals. These diseases are characterized by pathological changes in various brain regions, specific genetic mutations associated with the disease, deposits of abnormal proteins, and the degeneration of neurological cells. As neurodegenerative disorders vary in their epidemiological characteristics and vulnerability of neurons, treatment of these diseases is usually aimed at slowing disease progression. The heterogeneity of genetic and environmental factors involved in the process of neurodegeneration makes current treatment methods inadequate. However, the existence of common molecular mechanisms in the pathogenesis of these diseases may allow the development of new targeted therapeutic strategies. Oxidative and nitrosative stress damages membrane components by accumulating ROS and RNS and disrupting redox balance. This process results in the induction of apoptosis, which is important in the pathogenesis of neurodegenerative diseases through oxidative stress. Studies conducted using postmortem human samples, animal models, and cell cultures have demonstrated that oxidative stress, nitrosative stress, and apoptosis are crucial factors in the development of diseases such as Alzheimer's, Parkinson's, Multiple Sclerosis, amyotrophic lateral sclerosis, and Huntington's disease. The excessive production of reactive oxygen and nitrogen species, elevated levels of free radicals, heightened mitochondrial stress, disturbances in energy metabolism, and the oxidation and nitrosylation of cellular macromolecules are recognized as triggers for neuronal cell death. Challenges in managing and treating neurodegenerative diseases require a better understanding of this field at the molecular level. Therefore, this review elaborates on the molecular mechanisms by which oxidative and nitrosative stress are involved in neuronal apoptosis.
Collapse
Affiliation(s)
- Nuray Üremiş
- Department of Medical BiochemistryFaculty of Medicine, Kahramanmaraş Sütçü İmam UniversityKahramanmaraşTurkey
| | - Muhammed Mehdi Üremiş
- Department of Medical BiochemistryFaculty of Medicine, Kahramanmaraş Sütçü İmam UniversityKahramanmaraşTurkey
| |
Collapse
|
4
|
Buga AM, Padureanu V, Riza AL, Oancea CN, Albu CV, Nica AD. The Gut-Brain Axis as a Therapeutic Target in Multiple Sclerosis. Cells 2023; 12:1872. [PMID: 37508537 PMCID: PMC10378521 DOI: 10.3390/cells12141872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The CNS is very susceptible to oxidative stress; the gut microbiota plays an important role as a trigger of oxidative damage that promotes mitochondrial dysfunction, neuroinflammation, and neurodegeneration. In the current review, we discuss recent findings on oxidative-stress-related inflammation mediated by the gut-brain axis in multiple sclerosis (MS). Growing evidence suggests targeting gut microbiota can be a promising strategy for MS management. Intricate interaction between multiple factors leads to increased intra- and inter-individual heterogeneity, frequently painting a different picture in vivo from that obtained under controlled conditions. Following an evidence-based approach, all proposed interventions should be validated in clinical trials with cohorts large enough to reach significance. Our review summarizes existing clinical trials focused on identifying suitable interventions, the suitable combinations, and appropriate timings to target microbiota-related oxidative stress. Most studies assessed relapsing-remitting MS (RRMS); only a few studies with very limited cohorts were carried out in other MS stages (e.g., secondary progressive MS-SPMS). Future trials must consider an extended time frame, perhaps starting with the perinatal period and lasting until the young adult period, aiming to capture as many complex intersystem interactions as possible.
Collapse
Affiliation(s)
- Ana Maria Buga
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.M.B.); (C.N.O.)
| | - Vlad Padureanu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania;
| | - Anca-Lelia Riza
- Laboratory of Human Genomics, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania;
- Regional Center for Medical Genetics Dolj, Emergency County Hospital Craiova, 200638 Craiova, Romania
| | - Carmen Nicoleta Oancea
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.M.B.); (C.N.O.)
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alexandru Dan Nica
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.M.B.); (C.N.O.)
| |
Collapse
|
5
|
The Possible Role of Neural Cell Apoptosis in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23147584. [PMID: 35886931 PMCID: PMC9316123 DOI: 10.3390/ijms23147584] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
The etiology of multiple sclerosis (MS), a demyelinating disease affecting the central nervous system (CNS), remains obscure. Although apoptosis of oligodendrocytes and neurons has been observed in MS lesions, the contribution of this cell death process to disease pathogenesis remains controversial. It is usually considered that MS-associated demyelination and axonal degeneration result from neuroinflammation and an autoimmune process targeting myelin proteins. However, experimental data indicate that oligodendrocyte and/or neuronal cell death may indeed precede the development of inflammation and autoimmunity. These findings raise the question as to whether neural cell apoptosis is the key event initiating and/or driving the pathological cascade, leading to clinical functional deficits in MS. Similarly, regarding axonal damage, a key pathological feature of MS lesions, the roles of inflammation-independent and cell autonomous neuronal processes need to be further explored. While oligodendrocyte and neuronal loss in MS may not necessarily be mutually exclusive, particular attention should be given to the role of neuronal apoptosis in the development of axonal loss. If proven, MS could be viewed primarily as a neurodegenerative disease accompanied by a secondary neuroinflammatory and autoimmune process.
Collapse
|
6
|
Cui Y, Yu H, Bu Z, Wen L, Yan L, Feng J. Focus on the Role of the NLRP3 Inflammasome in Multiple Sclerosis: Pathogenesis, Diagnosis, and Therapeutics. Front Mol Neurosci 2022; 15:894298. [PMID: 35694441 PMCID: PMC9175009 DOI: 10.3389/fnmol.2022.894298] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is initiated with an aberrant innate immune response in the central nervous system (CNS) and is involved in many neurological diseases. Inflammasomes are intracellular multiprotein complexes that can be used as platforms to induce the maturation and secretion of proinflammatory cytokines and pyroptosis, thus playing a pivotal role in neuroinflammation. Among the inflammasomes, the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasome is well-characterized and contributes to many neurological diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), and ischemic stroke. MS is a chronic autoimmune disease of the CNS, and its hallmarks include chronic inflammation, demyelination, and neurodegeneration. Studies have demonstrated a relationship between MS and the NLRP3 inflammasome. To date, the pathogenesis of MS is not fully understood, and clinical studies on novel therapies are still underway. Here, we review the activation mechanism of the NLRP3 inflammasome, its role in MS, and therapies targeting related molecules, which may be beneficial in MS.
Collapse
|
7
|
Neuroprotective Effect of Glatiramer Acetate on Neurofilament Light Chain Leakage and Glutamate Excess in an Animal Model of Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms222413419. [PMID: 34948217 PMCID: PMC8707261 DOI: 10.3390/ijms222413419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/29/2022] Open
Abstract
Axonal and neuronal pathologies are a central constituent of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), induced by the myelin oligodendrocyte glycoprotein (MOG) 35–55 peptide. In this study, we investigated neurodegenerative manifestations in chronic MOG 35–55 induced EAE and the effect of glatiramer acetate (GA) treatment on these manifestations. We report that the neuronal loss seen in this model is not attributed to apoptotic neuronal cell death. In EAE-affected mice, axonal damage prevails from the early disease phase, as revealed by analysis of neurofilament light (NFL) leakage into the sera along the disease duration, as well as by immunohistological examination. Elevation of interstitial glutamate concentrations measured in the cerebrospinal fluid (CSF) implies that glutamate excess plays a role in the damage processes inflicted by this disease. GA applied as a therapeutic regimen to mice with apparent clinical symptoms significantly reduces the pathological manifestations, namely apoptotic cell death, NFL leakage, histological tissue damage, and glutamate excess, thus corroborating the neuroprotective consequences of this treatment.
Collapse
|
8
|
Govindarajan V, de Rivero Vaccari JP, Keane RW. Role of inflammasomes in multiple sclerosis and their potential as therapeutic targets. J Neuroinflammation 2020; 17:260. [PMID: 32878648 PMCID: PMC7469327 DOI: 10.1186/s12974-020-01944-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS), and it remains the most common immune-mediated disorder affecting the CNS. While the cause of MS is unclear, the underlying pathomechanisms are thought to be either destruction by autoimmune T cells or dysfunction of myelin-producing cells. Recent advances have indicated that inflammasomes contribute the etiology of MS. Inflammasomes are multiprotein complexes of the innate immune response involved in the processing of caspase-1, the activation of pro-inflammatory cytokines interleukin (IL)-1β and IL-18 as well as the cell death-mediated mechanism of pyroptosis and the activation of the adaptive immune response. Here we review the literature to date on the role of different inflammasome signaling pathways in the pathogenesis of MS and how these pathways may be targeted to reduce deleterious inflammatory processes and improve outcomes in this patient population.
Collapse
Affiliation(s)
- Vaidya Govindarajan
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB 5058, Miami, FL, 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Robert W Keane
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB 5058, Miami, FL, 33136, USA. .,Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
9
|
Serum levels of inflammasome pathway factors in clinically isolated syndrome and multiple sclerosis patients: a pilot study. Cent Eur J Immunol 2020; 45:237-240. [PMID: 33456338 PMCID: PMC7792443 DOI: 10.5114/ceji.2020.96877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 04/18/2018] [Indexed: 12/30/2022] Open
Abstract
Pathogenic roles of nuclear factor κB (NF-κB) pathway and NLRP3 inflammasome complex factors are involved in multiple sclerosis (MS) development. Activation of the NF-κB, NLRP3, and caspase-1 cascade results in production of proinflammatory cytokines that lead to stimulation of macrophages, lymphocytes, and glial cells. Although increased levels of inflammasome complex factors are observed in MS, contribution of inflammasome pathway to conversion from clinically isolated syndrome (CIS) to relapsing remitting MS (RRMS) has been scarcely investigated. To examine predictive value of inflammasome factors in CIS-MS conversion, levels of NLRP3, caspase-1, and NFκB are measured by ELISA in sera of age-gender matched CIS (n = 18; 8 converting, 10 non-converting) and RRMS (n = 23) patients. CIS and RRMS patients have comparable serum levels of NLRP3, caspase-1, and NFκB. Similarly, no statistically significant difference can be found among converting and non-converting CIS patients by means of inflammasome complex factor levels. Inflammasome factors are presumably overexpressed at early stages of MS. Therefore, they are unlikely to be used as biomarkers to predict CIS-MS conversion.
Collapse
|
10
|
Song Q, Zhao F, Yao J, Dai H, Hu L, Yu S. Protective effect of microRNA-134-3p on multiple sclerosis through inhibiting PRSS57 and promotion of CD34 + cell proliferation in rats. J Cell Biochem 2020; 121:4347-4363. [PMID: 32619071 DOI: 10.1002/jcb.29643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/19/2019] [Indexed: 12/23/2022]
Abstract
MicroRNAs (miRs) have been extensively studied for their involvement in multiple sclerosis (MS). We investigated the involvement of miR-134-3p on MS. The MS rat model was established, and positive expression of interleukin-17 (IL-17) was detected using the immunohistochemical method while the expression of miR-134-3p and serine protease 57 (PRSS57) was determined by means of reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Second, the miR-134-3p overexpression or short hairpin RNA against PRSS57 was introduced into the CD34+ cells to investigate the levels of proliferation and apoptosis-related genes by RT-qPCR and Western blot analysis. In addition, analysis of the targeting relations of miR-134-3p and PRSS57 was conducted using online software and dual-luciferase reporter gene assay. Furthermore, neuronal functions, inflammatory response, proliferation, and apoptosis of CD34+ cells were assayed by flow cytometry, enzyme-linked immunosorbent assay, and methyl thiazolyl tetrazolium. IL-17 and PRSS57 expression increased while miR-134-3p expression decreased in the spinal cord from MS rats. miR-134-3p could target PRSS57. miR-134-3p overexpression or PRSS57 silencing enhanced mitochondrial activity of neurons, mitochondrial membrane potential content, CD34+ cell proliferation, while decreasing Cyt C content, inflammatory response, and cell apoptosis. Collectively, overexpression of miR-134-3p promotes CD34+ cell proliferation via inhibition of PRSS57 in MS, which may serve as a promising target for MS intervention.
Collapse
Affiliation(s)
- Qihan Song
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Fengli Zhao
- Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Jingfan Yao
- Department of Neurology, Beijing Tiantan Hospital of Capital Medical University, Beijing, China
| | - Hailin Dai
- Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Lei Hu
- Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Shun Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Basnyat P, Sumelahti ML, Lehtimäki T, Elovaara I, Hagman S. Gene expression profiles of TNF-like cytokine 1A (TL1A) and its receptors death receptor 3 (DR3) and decoy receptor 3 (DcR3) in multiple sclerosis. J Neuroimmunol 2019; 335:577020. [PMID: 31445379 DOI: 10.1016/j.jneuroim.2019.577020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/02/2019] [Accepted: 08/13/2019] [Indexed: 01/12/2023]
Abstract
TL1A/DR3/DcR3 pathway is an important mediator of inflammatory responses and contributes to the pathogenesis of several chronic inflammatory diseases. Therefore, we analysed PBMC gene expression of these molecules in 30 relapsing-remitting multiple sclerosis (RRMS) patients, 8 secondary progressive MS (SPMS), 9 primary progressive MS (PPMS), 11 clinically isolated syndrome (CIS) patients, and 16 healthy controls (HCs), to evaluate their biomarker potential in MS. The results showed significant decrease in TL1A expression in RRMS compared to other study groups. TL1A as a marker of inflammation, we found its higher expression among treatment näive RRMS patients as compared to HCs and among patients who were treated with DMTs. Moreover, TL1A expression was found to be associated with the clinical and MRI findings of MS patients suggesting its possible involvement in the establishment or preservation of immune system homeostasis or in the regulation of inflammatory activity. Taken together, these findings suggest the TL1A should be evaluated further for its potential as a candidate biomarker of inflammatory activity and the marker of therapeutic response to immunomodulatory treatments in MS.
Collapse
Affiliation(s)
- Pabitra Basnyat
- Neuroimmunology Unit, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.
| | - Marja-Liisa Sumelahti
- Neuroimmunology Unit, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland; Department of Neurology, Tampere University Hospital, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Irina Elovaara
- Neuroimmunology Unit, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland; Department of Neurology, Tampere University Hospital, Tampere, Finland
| | - Sanna Hagman
- Neuroimmunology Unit, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland; Neuro Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| |
Collapse
|
12
|
Pi Z, Lin H, Yang J. Isoflurane reduces pain and inhibits apoptosis of myocardial cells through the phosphoinositide 3-kinase/protein kinase B signaling pathway in mice during cardiac surgery. Mol Med Rep 2018; 17:6497-6505. [PMID: 29488606 PMCID: PMC5928630 DOI: 10.3892/mmr.2018.8642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/28/2017] [Indexed: 01/27/2023] Open
Abstract
Heart bypass surgery is the most common treatment for myocardial ischemia. Clinical investigations have revealed that isoflurane anesthesia is efficient to alleviate pain during cardiac surgery, including heart bypass surgery. Previous studies have revealed the protective effects of isoflurane on myocardial cells of patients with myocardial ischemia during the perioperative period. The present study aimed to investigate the mechanism underlying the protective effects of isoflurane on myocardial cells in mice with myocardial ischemia. ELISA, flow cytometry, immunofluorescence and western blotting were used to analyze the effects of isoflurane anesthesia on myocardial cells. Briefly, myocardial cell apoptosis and viability, pain, phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signaling pathway expression and the pharmacodynamics of isoflurane were studied in mice treated with isoflurane for heart bypass surgery. The results demonstrated that isoflurane anesthesia efficiently attenuated pain in mice during surgery. Viability and apoptosis of myocardial cells was also improved by isoflurane in vitro and in vivo. The PI3K/AKT pathway was upregulated in myocardial cells on day 3 post-operation. Mechanistically, isoflurane promoted PI3K/AKT activation, upregulated B-cell lymphoma 2 (Bcl-2)-associated X protein and Bcl-2 expression levels, and reduced the expression levels of caspase-3 and caspase-8 in myocardial cells. In conclusion, the findings indicated that isoflurane is beneficial for pain attenuation and inhibits apoptosis of myocardial cells via the PI3K/AKT signaling pathway in mice during cardiac surgery.
Collapse
Affiliation(s)
- Zhibing Pi
- Department of Anesthesiology of The First Affiliated Hospital of Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hai Lin
- Department of Anesthesiology of The First Affiliated Hospital of Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jianping Yang
- Department of Anesthesiology of The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
13
|
Yang T, Wang S, Yang X, Zheng Q, Wang L, Li Q, Wei M, Du Z, Fan Y. Upregulation of Bcl-2 and Its Promoter Signals in CD4+ T Cells during Neuromyelitis Optica Remission. Front Neurosci 2017; 11:11. [PMID: 28174515 PMCID: PMC5258721 DOI: 10.3389/fnins.2017.00011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 01/09/2017] [Indexed: 12/27/2022] Open
Abstract
The homeostatic balance between production and elimination of CD4+ T cells in peripheral blood plays an important role in patients with neuromyelitis optica (NMO). The objective of the present study was to evaluate the anti-apoptosis genes Bcl-2 and its promoter signal (nuclear factor kappa-light-chain-enhancer of activated B cells, NFκB) in CD4+ T cells. Healthy subjects (HS, n = 25) and patients with multiple sclerosis (MS) (n = 25) and NMO (n = 30) in remission were consecutively enrolled in this prospective study between May and December 2015. CD4+ T cells were isolated using magnetic beads coated with anti-CD4 monoclonal antibodies, and gene expression of Bcl-2, NFκB, phosphatidylinositol-4, 5-bisphosphate 3-kinase/protein kinase B (PI3K/Akt), and MAP kinase kinase kinase 7 (MAP3K7) was measured by real-time reverse transcription-polymerase chain reaction (rt-PCR). Cytokines of tumor necrosis factor (TNF)-α and interleukin (IL)-1β were detected using human cytokine multiplex assay. Bcl-2 and NFκB gene expressions were elevated in NMO patients (1.63 ± 0.25; 2.35 ± 0.25) compared with those of HS (0.90 ± 0.11; 1.42 ± 0.22) and/or MS patients (1.03 ± 0.18; 1.55 ± 0.20) (P < 0.05). MAP3K7, but not Akt, was increased in NMO patients (1.23 ± 0.18; 1.56 ± 0.22) (P < 0.01) and was a significant factor related to elevated NFκB gene expressions (P < 0.001). On the other hand, IL-1β and TNF-α were also detected in the study and the results showed that both were elevated in NMO patients (23.84 ± 1.81; 56.40 ± 2.45) (P < 0.01; P < 0.05, respectively). We propose that MAP3K7 induced by IL-1β and TNF-α but not Akt promotes NFκB expression and, in turn, prolongs Bcl-2-mediated survival of CD4+ T cells in NMO patients.
Collapse
Affiliation(s)
- Tao Yang
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Su Wang
- Department of Tumor Radiotherapy, Hiser Medical Center of Qingdao Qingdao, China
| | - Xiao Yang
- School of Management Science and Engineering, Shandong University of Finance and Economics Jinan, China
| | - Qi Zheng
- Department of Oncology, Guang An Men Hospital of China Academy of Chinese Medical SciencesBeijing, China; School of Traditional Chinese Medicine, Capital Medical UniversityBeijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University Beijing, China
| | - Qian Li
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Mingyan Wei
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Zongpan Du
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Yongping Fan
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| |
Collapse
|
14
|
Macchi B, Mastino A. Programmed cell death and natural killer cells in multiple sclerosis: new potential therapeutic targets? Neural Regen Res 2016; 11:733-4. [PMID: 27335552 PMCID: PMC4904459 DOI: 10.4103/1673-5374.182695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Beatrice Macchi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Antonio Mastino
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy; The Institute of Translational Pharmacology, CNR, Rome, Italy
| |
Collapse
|
15
|
Macchi B, Marino-Merlo F, Nocentini U, Pisani V, Cuzzocrea S, Grelli S, Mastino A. Role of inflammation and apoptosis in multiple sclerosis: Comparative analysis between the periphery and the central nervous system. J Neuroimmunol 2015; 287:80-7. [PMID: 26439966 DOI: 10.1016/j.jneuroim.2015.08.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 01/23/2023]
Abstract
Multiple sclerosis (MS) is a complex, multifactorial disease associated with damage to the axonal myelin sheaths and neuronal degeneration. The pathognomonic event in MS is oligodendrocyte loss accompanied by axonal damage, blood-brain barrier leakage, inflammation and infiltration of immune cells. The etiopathogenesis of MS is far from being elucidated. However, increasing evidence suggests that the inflammatory and apoptotic responses, occurring in patients either at the peripheral level or the central nervous system (CNS), can play a role. In this review, we give a comprehensive picture of general aspects of inflammation and apoptosis in MS, with special emphasis on the until now not well highlighted possible links between phenomena relevant to these aspects occurring in either the periphery or in the CNS during MS.
Collapse
Affiliation(s)
- Beatrice Macchi
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Francesca Marino-Merlo
- Department of Biological and Environmental Sciences, University of Messina, Via F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| | - Ugo Nocentini
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; I.R.C.C.S. "Santa Lucia" Foundation, Via Ardeatina 306, 00179 Rome, Italy.
| | - Valerio Pisani
- I.R.C.C.S. "Santa Lucia" Foundation, Via Ardeatina 306, 00179 Rome, Italy.
| | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Via F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| | - Sandro Grelli
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Antonio Mastino
- Department of Biological and Environmental Sciences, University of Messina, Via F. Stagno d'Alcontres 31, 98166 Messina, Italy; The Institute of Translational Pharmacology, CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| |
Collapse
|