1
|
Mallardo M, Mazzeo F, Lus G, Signoriello E, Daniele A, Nigro E. Impact of Lifestyle Interventions on Multiple Sclerosis: Focus on Adipose Tissue. Nutrients 2024; 16:3100. [PMID: 39339700 PMCID: PMC11434938 DOI: 10.3390/nu16183100] [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: 07/25/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by demyelination in the central nervous system (CNS), affecting individuals globally. The pathological mechanisms underlying MS remain unclear, but current evidence suggests that inflammation and immune dysfunction play a critical role in the pathogenesis of MS disease. Adipose tissue (AT) is a dynamic multifunctional organ involved in various immune diseases, including MS, due to its endocrine function and the secretion of adipokines, which can influence inflammation and immune responses. Physical activity represents an efficacious non-pharmacological strategy for the management of a spectrum of conditions that not only improves inflammatory and immune functions but also directly affects the status and function of AT. Additionally, the exploration of nutritional supplementation represents an important field of MS research aimed at enhancing clinical symptoms and is closely tied to the regulation of metabolic responses, including adipokine secretion. This review, therefore, aims to elucidate the intricate relationship between lifestyle and MS by providing an overview of the latest published data about the involvement of AT and the main adipokines, such as adiponectin, leptin, and tumor necrosis factor α (TNFα) in the pathogenesis of MS. Furthermore, we explore whether physical activity and dietary management could serve as useful strategies to improve the quality of life of MS patients.
Collapse
Affiliation(s)
- Marta Mallardo
- Department of Molecular and Biotechnological Medicine, University of Naples "Federico II", 80138 Naples, Italy
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Filomena Mazzeo
- Department of Economics, Law, Cybersecurity and Sports Sciences (DiSEGIM), University of Naples "Parthenope", 80035 Naples, Italy
| | - Giacomo Lus
- Multiple Sclerosis Center, II Neurological Clinic, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
- Department of Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Elisabetta Signoriello
- Multiple Sclerosis Center, II Neurological Clinic, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
- Department of Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Aurora Daniele
- Department of Molecular and Biotechnological Medicine, University of Naples "Federico II", 80138 Naples, Italy
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Ersilia Nigro
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy
- Department of Pharmaceutical, Biological, Environmental Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via G. Vivaldi 42, 81100 Caserta, Italy
| |
Collapse
|
2
|
Neto A, Fernandes A, Barateiro A. The complex relationship between obesity and neurodegenerative diseases: an updated review. Front Cell Neurosci 2023; 17:1294420. [PMID: 38026693 PMCID: PMC10665538 DOI: 10.3389/fncel.2023.1294420] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is a global epidemic, affecting roughly 30% of the world's population and predicted to rise. This disease results from genetic, behavioral, societal, and environmental factors, leading to excessive fat accumulation, due to insufficient energy expenditure. The adipose tissue, once seen as a simple storage depot, is now recognized as a complex organ with various functions, including hormone regulation and modulation of metabolism, inflammation, and homeostasis. Obesity is associated with a low-grade inflammatory state and has been linked to neurodegenerative diseases like multiple sclerosis (MS), Alzheimer's (AD), and Parkinson's (PD). Mechanistically, reduced adipose expandability leads to hypertrophic adipocytes, triggering inflammation, insulin and leptin resistance, blood-brain barrier disruption, altered brain metabolism, neuronal inflammation, brain atrophy, and cognitive decline. Obesity impacts neurodegenerative disorders through shared underlying mechanisms, underscoring its potential as a modifiable risk factor for these diseases. Nevertheless, further research is needed to fully grasp the intricate connections between obesity and neurodegeneration. Collaborative efforts in this field hold promise for innovative strategies to address this complex relationship and develop effective prevention and treatment methods, which also includes specific diets and physical activities, ultimately improving quality of life and health.
Collapse
Affiliation(s)
- Alexandre Neto
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Adelaide Fernandes
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
3
|
UCHL1, besides leptin and fibronectin, also could be a sensitive marker of the relapsing-remitting type of multiple sclerosis. Sci Rep 2023; 13:3423. [PMID: 36854961 PMCID: PMC9974955 DOI: 10.1038/s41598-023-30237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
Research on the markers of immunoregulatory response in multiple sclerosis (MS) is still of great importance. The aim of our study was the evaluation of leptin, fibronectin, and UCHL1 concentrations as potential biomarkers of a relapsing-remitting type of MS (RRMS). Surface Plasmon Resonance Imaging (SPRI) biosensors were used for the evaluation of proteins concentrations in 100 RRMS patients and 46 healthy volunteers. Plasma leptin, fibronectin, and UCHL1 concentrations were significantly higher in RRMS patients compared to the control group (p < 0.001, respectively). UCHL1 concentration evaluation revealed the highest diagnostic sensitivity (100%) and negative predictive value (100%) in differentiating MS patients from healthy individuals. There was no significant difference in the UCHL1 concentrations depending on the patient's sex, the presence of relapse within the last 24 months, and the EDSS value (p > 0.05, respectively). In RRMS patients UCHL1 concentration positively correlated with fibronectin levels (r = 0.3928; p < 0.001). In the current cohort of patients plasma UCHL1 concentration was independent of the time of MS relapse and the severity of neurological symptoms. Thus current study may indicate that plasma UCHL1, besides leptin and fibronectin, also could be a promising high-sensitive potential biomarker of relapsing-remitting type of MS. However, these results should be validated with a larger group of patients, taking into account neuroimaging and cerebrospinal fluid analysis data, and by comparing them to patients with other neurological diseases as a control group.
Collapse
|
4
|
Correale J, Marrodan M. Multiple sclerosis and obesity: The role of adipokines. Front Immunol 2022; 13:1038393. [PMID: 36457996 PMCID: PMC9705772 DOI: 10.3389/fimmu.2022.1038393] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/28/2022] [Indexed: 11/24/2023] Open
Abstract
Multiple Sclerosis (MS), a chronic inflammatory disease of the central nervous system that leads to demyelination and neurodegeneration has been associated with various environmental and lifestyle factors. Population-based studies have provided evidence showing the prevalence of MS is increasing worldwide. Because a similar trend has been observed for obesity and metabolic syndrome, interest has grown in possible underlying biological mechanisms shared by both conditions. Adipokines, a family of soluble factors produced by adipose tissue that participate in a wide range of biological functions, contribute to a low state of chronic inflammation observed in obesity, and influence immune function, metabolism, and nutritional state. In this review, we aim to describe epidemiological and biological factors common to MS and obesity, as well as provide an update on current knowledge of how different pro- and anti-inflammatory adipokines participate as immune response mediators in MS, as well as in the animal model for MS, namely, experimental autoimmune encephalomyelitis (EAE). Multiple Sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination, and neurodegeneration. Although its pathogenesis is not yet fully understood, there is considerable evidence to suggest MS arises from complex interactions between individual genetic susceptibility and external environmental factors. In recent decades, population-based studies have provided evidence indicating the prevalence of MS is increasing worldwide, in parallel with the rise in obesity and metabolic syndrome. This synchronous increment in the incidence of both MS and obesity has led to a search for potential biological mechanisms linking both conditions. Notably, a large number of studies have established significant correlation between obesity and higher prevalence, or worse prognosis, of several immune-mediated conditions. Fat tissue has been found to produce a variety of soluble factors named adipokines. These mediators, secreted by both adipocytes as well as diverse immune cells, participate in a wide range of biological functions, further strengthening the concept of a link between immune function, metabolism, and nutritional state. Because obesity causes overproduction of pro-inflammatory adipokines (namely leptin, resistin and visfatin) and reduction of anti-inflammatory adipokines (adiponectin and apelin), adipose tissue dysregulation would appear to contribute to a state of chronic, low-grade inflammation favoring the development of disease. In this review, we present a summary of current knowledge related to the pathological effects of different adipokines, prevalent in obese MS patients.
Collapse
Affiliation(s)
- Jorge Correale
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | |
Collapse
|
5
|
Low SOCS3 expression in CD4 + T cells from pemphigus vulgaris patients enhanced Th1- and Th17-cell differentiation and exacerbated acantholysis via STAT activation. Mol Immunol 2022; 150:114-125. [PMID: 36030709 DOI: 10.1016/j.molimm.2022.08.007] [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: 03/29/2022] [Revised: 07/08/2022] [Accepted: 08/14/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Pemphigus vulgaris (PV) is a chronic inflammatory autoimmune blistering disease. Aberrant SOCS3/STAT pathway activation is associated with many autoimmune diseases. This study explored the relationship between activation of the SOCS3/STAT pathway and abnormally increased proportions of Th1 and Th17 cells in the peripheral blood of PV patients as well as the effect of CD4+ T cells with abnormal SOCS3/STAT pathway activation on acantholysis. METHODS In PV patients, the proportions of Th1 and Th17 cells in peripheral blood, the levels of IFN-γ and IL-17 in serum and the mRNA levels of SOCS3 and STAT1/3 in CD4+ T cells were detected. Then, SOCS3-knockdown primary CD4+ T cells were prepared, and cocultured with HaCaT cells. Finally, after SOCS3 knockdown and coculture, CD4+ T cells were collected, and the proportions of Th1 and Th17 cells, the protein levels of STAT1/3 and p-STAT1/3, and the levels of IFN-γ and IL-17 were measured. After 2 days of coculture, HaCaT cells were collected, inflammatory factors mRNA expression and acantholysis were assessed. RESULTS In PV patients, the proportions of Th1 (P = 0.016) and Th17 (P = 0.045) cells and the levels of IFN-γ (P = 0.010) were significantly increased. SOCS3 mRNA in CD4+ T cells was significantly decreased (P = 0.008), whereas STAT1 (P = 0.043) and STAT3 (P = 0.004) mRNA were significantly increased. After SOCS3 knockdown, the proportions of Th1 (P < 0.001) and Th17 (P = 0.006) cells, the levels of IFN-γ (P < 0.001) and IL-17 (P = 0.001), and the protein levels of p-STAT1 (P = 0.001) and p-STAT3 (P = 0.003) were significantly increased in the CD4+ T-shSOCS3-1 group. In the coculture system, the proportions of Th1 (P < 0.001) and Th17 (P < 0.001) cells, the levels of IFN-γ (P < 0.001) and IL-17 (P < 0.001), and the number of cell fragments (P < 0.001) were significantly increased in the CD4+ T-shSOCS3-1+HaCaT-PV-IgG group, whereas the protein level of desmoglein3 (Dsg3) was significantly decreased. In addition, PV-IgG significantly increased IFN-γ and IL-6 mRNA in HaCaT cells. CONCLUSION Low SOCS3 expression in CD4+ T cells from PV patients leads to overactivation of STAT, which causes CD4+ T cells to overdifferentiate into Th1 and Th17 cells. Additionally, PV-IgG-induced local inflammation in skin lesions, which is mediated by IFN-γ and IL-6, can aggravate this phenomenon. Furthermore, low SOCS3 expression in CD4+ T cells further exacerbates PV-IgG-induced acantholysis. Therefore, upregulating the expression of SOCS3 in CD4+ T cells of PV patients and maintaining the balance of the IFN-γ/STAT1/SOCS3 and IL-6/STAT3/SOCS3 pathways can alleviate acantholysis in patients with PV.
Collapse
|
6
|
Fahmi RM, Kamel AE, Elsayed DA, Zidan AA, Sarhan NT. Serum levels of leptin and adiponectin in patients with multiple sclerosis. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2021. [DOI: 10.1186/s41983-021-00369-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The role of adipokines such as leptin and adiponectin in regulating the immunity has been documented, however data concerning their consequence on multiple sclerosis (MS) Egyptian patients are deficient. The aim of this study is to demonstrate the serum levels of leptin and adiponectin in MS patients and to assess their association with disease disability and severity. A case–control study including 60 subjects (30 MS patients and 30 age, sex and body mass index-matched healthy controls) was performed.
Results
Serum leptin level was significantly higher among MS patients than controls (P < 0.001) while adiponectin was not significantly elevated in MS patients (P = 0.24). There was a significant positive correlation between leptin levels with MS disability (Expanded Disability Status Scale) (r = 0.678; P < 0.001), severity (Multiple Sclerosis Severity Score) (r = 0.631; P < 0.001) and progression (progression index) (r = 0.461; P = 0.01). There was no statistically significant correlation between adiponectin with disease disability, severity or progression.
Conclusions
MS patients had significantly higher serum leptin levels and insignificant adiponectin levels compared to controls. Leptin has a potential role in multiple sclerosis disability and severity. However, adiponectin is not useful as a biomarker of MS disease, disability and severity.
Collapse
|
7
|
Bechara R, Gaffen SL. '(m 6)A' stands for 'autoimmunity': reading, writing, and erasing RNA modifications during inflammation. Trends Immunol 2021; 42:1073-1076. [PMID: 34728144 DOI: 10.1016/j.it.2021.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/16/2022]
Abstract
Covalent RNA modifications that regulate gene expression post transcriptionally, in particular N6-methyladenosine (m6A), are emerging as important regulators of autoimmune responses. Here, we highlight new findings describing the functional diversity and specificity of m6A modifications and their regulation in the context of autoimmunity.
Collapse
Affiliation(s)
- Rami Bechara
- Université Paris-Saclay, CEA, INSERM UMR 1184, Centre de Recherche en Immunologie des Infections Virales et des Maladies Auto-Immunes, Le Kremlin Bicêtre, France.
| | - Sarah L Gaffen
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Adipokines as Immune Cell Modulators in Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms221910845. [PMID: 34639186 PMCID: PMC8509121 DOI: 10.3390/ijms221910845] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS), a chronic inflammatory and demyelinating disease of the central nervous system (CNS), is a major clinical and societal problem, which has a tremendous impact on the life of patients and their proxies. Current immunomodulatory and anti-inflammatory therapies prove to be relatively effective; however, they fail to concomitantly stop ongoing neurological deterioration and do not reverse acquired disability. The proportion to which genetic and environmental factors contribute to the etiology of MS is still incompletely understood; however, a recent association between MS etiology and obesity was shown, with obesity greatly increasing the risk of developing MS. An altered balance of adipokines, which are white adipose tissue (WAT) hormones, plays an important role in the low-grade chronic inflammation during obesity by their pervasive modification of local and systemic inflammation. Vice versa, inflammatory factors secreted by immune cells affect adipokine function. To explore the role of adipokines in MS pathology, we will here review the reciprocal effects of adipokines and immune cells and summarize alterations in adipokine levels in MS patient cohorts. Finally, we will discuss proof-of-concept studies demonstrating the therapeutic potential of adipokines to target both neuroinflammation and neurodegeneration processes in MS.
Collapse
|
9
|
Alhazzani K, Ahmad SF, Al-Harbi NO, Attia SM, Bakheet SA, Sarawi W, Alqarni SA, Algahtani M, Nadeem A. Pharmacological Inhibition of STAT3 by Stattic Ameliorates Clinical Symptoms and Reduces Autoinflammation in Myeloid, Lymphoid, and Neuronal Tissue Compartments in Relapsing-Remitting Model of Experimental Autoimmune Encephalomyelitis in SJL/J Mice. Pharmaceutics 2021; 13:pharmaceutics13070925. [PMID: 34206429 PMCID: PMC8308768 DOI: 10.3390/pharmaceutics13070925] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/17/2021] [Indexed: 11/22/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated inflammatory disease that leads to demyelination and neuronal loss in the central nervous system. Immune cells of lymphoid and myeloid origin play a significant role in the initiation and amplification of neuronal inflammation in MS. STAT3 signaling plays a pivotal role in both myeloid and lymphoid immune cells, such as neutrophils and CD4+ T cells, through regulation of their inflammatory potential. Dysregulation in STAT3 signaling in myeloid and lymphoid cell compartments has been reported in MS. In this report, we attempted to investigate the effect of a small molecular inhibitor of STAT3, i.e., Stattic, in a relapsing–remitting (RR) model of experimental autoimmune encephalomyelitis (EAE). The effect of Stattic was investigated for clinical features, oxidative stress parameters, and Th17-related signaling in both the periphery and brain of SJL/J mice. Our data report that p-STAT3 expression is elevated in granulocytes, CD4+ T cells, and brain tissue in myelin proteolipid protein (PLP)-immunized SJL/J mice, which is associated with the presence of clinical symptoms and upregulation of inflammatory markers in these cells/tissues. Treatment with Stattic leads to the amelioration of disease symptoms and attenuation of inflammatory markers in neutrophils (iNOS/nitrotyrosine/IL-1β), CD4+ T cells (IL-17A/IL-23R), and brain tissue (IL-17A/iNOS/IL-1β/MPO activity/lipid peroxides) in mice with EAE. These data suggest that the blockade of STAT3 signaling in cells of lymphoid and myeloid origin may cause the attenuation of systemic and neuronal inflammation, which could be responsible for the amelioration of disease symptoms in an RR model of EAE. Therefore, pharmacological inhibition of STAT3 in RRMS could be a potential therapeutic strategy.
Collapse
|
10
|
Najafi S, Saadat P, Beladi Moghadam N, Manoucherinia A, Aghazadeh Z, Vali Mohammadi A, Pashaiefar H, Hosseini M, Mirshafiey A. Evaluation of the Effect of Mannuronic Acid as a Novel NSAID With Immunosuppressive Properties on Expression of SOCS1, SOCS3, SHIP1, and TRAF6 Genes and Serum Levels of IL-6 and TNF-α in Patients With Multiple Sclerosis. J Clin Pharmacol 2021; 61:1303-1310. [PMID: 33908653 DOI: 10.1002/jcph.1879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/18/2021] [Indexed: 11/12/2022]
Abstract
Multiple sclerosis (MS) is described as a chronic inflammatory, demyelinating disease of the central nervous system on an autoimmune basis, which is the most frequent reason for nontraumatic disability in youth. The efficacy and safety of β-D-nannuronic acid (M2000) as a novel immunosuppressive drug (patented PCT/EP2017/067920) has been shown in an experimental model of MS and also in a phase 2 clinical trial. The effects of M2000 on SOCS1, SOCS3, TRAF6, and SHIP1 gene expression and also serum levels of IL-6 and TNF-α in secondary progressive multiple sclerosis patients have been assessed in this study. In this study, 14 secondary progressive multiple sclerosis patients and 14 healthy subjects (as the control group) were recruited from the phase 2 clinical trial (Clinical Trial identifier, IRCT2016111313739N6). Gene expression of SOCS1, SOCS3, TRAF6, and SHIP1 was measured at baseline and after 6 months of therapy with M2000 using a quantitative real-time polymerase chain reaction method. Furthermore, the serum levels of IL-6 and TNF-α were assessed by the enzyme-linked immunosorbent assay method. Our results showed that the gene expression of SOCS1, SOCS3, and SHIP1 was increased after 6 months of therapy with M2000 in MS patients. Moreover, the serum levels of IL-6 and TNF-α of patients declined compared with baseline, but this was not statistically significant. The results of this study demonstrated that M2000, with immunosuppressive properties, could upregulate SOCS1, SOCS3, and SHIP1 genes in patients with secondary progressive multiple sclerosis.
Collapse
Affiliation(s)
- Soheil Najafi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Payam Saadat
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Nahid Beladi Moghadam
- Department of Neurology, Imam Hossein Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ali Manoucherinia
- Department of Clinical Neuroscience (CNS), Karolinska Institutet, Stockholm, Sweden
| | - Zahra Aghazadeh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Pashaiefar
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Kolić I, Stojković L, Stankovic A, Stefanović M, Dinčić E, Zivkovic M. Association study of rs7799039, rs1137101 and rs8192678 gene variants with disease susceptibility/severity and corresponding LEP, LEPR and PGC1A gene expression in multiple sclerosis. Gene 2021; 774:145422. [PMID: 33450350 DOI: 10.1016/j.gene.2021.145422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/22/2020] [Accepted: 01/05/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Leptin (LEP), leptin receptor (LEPR) and peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC1A) are involved in the pathogenesis of multiple sclerosis (MS) by affecting the inflammatory response and reactive oxygen species production. LEP rs7799039 and LEPR rs1137101 genetic variants modify the serum LEP levels and PGC1A rs8192678 alters the PGC1A activity. The study objective was to explore the associations of these variants with susceptibility to MS, disease course/clinical parameters and also with peripheral blood mononuclear cell expression of the target genes and plasma LEP concentrations, in the study subjects. METHODS The study groups included 528 patients with MS and 429 controls. TaqMan® assays were used for genotyping and gene expression quantification. The Chi-square, parametric and nonparametric tests and simple/multiple logistic regression were performed for the statistical analysis of data. RESULTS A multiple logistic regression model including all three investigated variants, applied to patients (RRMS + SPMS) and controls, showed that PGC1A rs8192678 minor allele had an increased risk for the occurrence of disease, with OR (95%CI) = 1,32 (1,01-1,73), P = 0,04. Between-effect of gender and LEPR variant on the multiple sclerosis severity score (MSSS) was identified (P = 0,005). In male patients (relapsing-remitting and secondary progressive), LEPR minor allele carriers had increased MSSS (GG + AG vs AA, median (minimum-maximum) = 5,38 (0,64-9,88) vs 4,27 (0,78-9,63); P = 0,01, Padj = 0,03). In relapsing-remitting patients LEP rs7799039 affected the LEP gene expression (P = 0,006; Padj = 0,04). CONCLUSION The current findings implicate an impact of investigated genetic variants on the pathogenesis of MS.
Collapse
Affiliation(s)
- Ivana Kolić
- "Vinča" Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, Laboratory for Radiobiology and Molecular Genetics, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, Serbia
| | - Ljiljana Stojković
- "Vinča" Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, Laboratory for Radiobiology and Molecular Genetics, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, Serbia.
| | - Aleksandra Stankovic
- "Vinča" Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, Laboratory for Radiobiology and Molecular Genetics, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, Serbia
| | - Milan Stefanović
- "Vinča" Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, Laboratory for Radiobiology and Molecular Genetics, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, Serbia
| | - Evica Dinčić
- Military Medical Academy, Clinic for Neurology, Crnotravska 17, Belgrade, Serbia
| | - Maja Zivkovic
- "Vinča" Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, Laboratory for Radiobiology and Molecular Genetics, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, Serbia
| |
Collapse
|
12
|
Hamilton K, Harvey J. The Neuronal Actions of Leptin and the Implications for Treating Alzheimer's Disease. Pharmaceuticals (Basel) 2021; 14:ph14010052. [PMID: 33440796 PMCID: PMC7827292 DOI: 10.3390/ph14010052] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
It is widely accepted that the endocrine hormone leptin controls food intake and energy homeostasis via activation of leptin receptors expressed on hypothalamic arcuate neurons. The hippocampal formation also displays raised levels of leptin receptor expression and accumulating evidence indicates that leptin has a significant impact on hippocampal synaptic function. Thus, cellular and behavioural studies support a cognitive enhancing role for leptin as excitatory synaptic transmission, synaptic plasticity and glutamate receptor trafficking at hippocampal Schaffer collateral (SC)-CA1 synapses are regulated by leptin, and treatment with leptin enhances performance in hippocampus-dependent memory tasks. Recent studies indicate that hippocampal temporoammonic (TA)-CA1 synapses are also a key target for leptin. The ability of leptin to regulate TA-CA1 synapses has important functional consequences as TA-CA1 synapses are implicated in spatial and episodic memory processes. Moreover, degeneration is initiated in the TA pathway at very early stages of Alzheimer's disease, and recent clinical evidence has revealed links between plasma leptin levels and the incidence of Alzheimer's disease (AD). Additionally, accumulating evidence indicates that leptin has neuroprotective actions in various AD models, whereas dysfunctions in the leptin system accelerate AD pathogenesis. Here, we review the data implicating the leptin system as a potential novel target for AD, and the evidence that boosting the hippocampal actions of leptin may be beneficial.
Collapse
|
13
|
Rezaeepoor M, Hoseini-Aghdam M, Sheikh V, Eftekharian MM, Behzad M. Evaluation of Interleukin-23 and JAKs/STATs/SOCSs/ROR-γt Expression in Type 2 Diabetes Mellitus Patients Treated With or Without Sitagliptin. J Interferon Cytokine Res 2020; 40:515-523. [PMID: 33136467 DOI: 10.1089/jir.2020.0113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The production of interleukin-23 (IL-23) and the expression levels of related genes were evaluated in type 2 diabetes mellitus patients. The correlations between them were also determined. Thirty patients without sitagliptin (sitagliptin negative; SN), 30 patients with sitagliptin (sitagliptin positive; SP), and 30 healthy controls (HCs) were recruited. The level of IL-23 in the supernatant of anti CD3-activated peripheral blood mononuclear cells (PBMCs) was assessed using enzyme-linked immunosorbent assay. The expressions of IL-23, JAK1/JAK2/TYK2, STAT1/STAT3, ROR-γt, and SOCS1/SOCS3 in PBMCs were evaluated by real-time polymerase chain reaction. The production of IL-23 and the expressions of IL-23, JAK2, STAT3, and ROR-γt were observed to be enhanced in SN patients versus HCs, while the levels were decreased in SP patients versus SN patients (P < 0.05). SOCS1 and SOCS3 expressions were lower in SN patients than HCs, and their expressions were elevated in SP patients versus SN patients (P < 0.05). In SN patients, positive correlations between the IL-23 with fasting plasma glucose and HbA1c were observed, and JAK2/STAT3/ROR-γt were positively correlated with IL-23. JAK2, STAT3, and ROR-γt were positively related to each other and were negatively related to SOCS3. Enhanced IL-23/JAK2/STAT3/ROR-γt and reduced SOCS1/SOCS3 were found in SN patients. Sitagliptin may regulate the IL-23 and related gene expression.
Collapse
Affiliation(s)
- Mahsa Rezaeepoor
- Department of Immunology and School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mirhamed Hoseini-Aghdam
- Department of Immunology and School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Vida Sheikh
- Department of Internal Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mahdi Behzad
- Department of Immunology and School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
14
|
Pastor-Fernández G, Mariblanca IR, Navarro MN. Decoding IL-23 Signaling Cascade for New Therapeutic Opportunities. Cells 2020; 9:cells9092044. [PMID: 32906785 PMCID: PMC7563346 DOI: 10.3390/cells9092044] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/24/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
The interleukin 23 (IL-23) is a key pro-inflammatory cytokine in the development of chronic inflammatory diseases, such as psoriasis, inflammatory bowel diseases, multiple sclerosis, or rheumatoid arthritis. The pathological consequences of excessive IL-23 signaling have been linked to its ability to promote the production of inflammatory mediators, such as IL-17, IL-22, granulocyte-macrophage colony-stimulating (GM-CSF), or the tumor necrosis factor (TNFα) by target populations, mainly Th17 and IL-17-secreting TCRγδ cells (Tγδ17). Due to their pivotal role in inflammatory diseases, IL-23 and its downstream effector molecules have emerged as attractive therapeutic targets, leading to the development of neutralizing antibodies against IL-23 and IL-17 that have shown efficacy in different inflammatory diseases. Despite the success of monoclonal antibodies, there are patients that show no response or partial response to these treatments. Thus, effective therapies for inflammatory diseases may require the combination of multiple immune-modulatory drugs to prevent disease progression and to improve quality of life. Alternative strategies aimed at inhibiting intracellular signaling cascades using small molecule inhibitors or interfering peptides have not been fully exploited in the context of IL-23-mediated diseases. In this review, we discuss the current knowledge about proximal signaling events triggered by IL-23 upon binding to its membrane receptor to bring to the spotlight new opportunities for therapeutic intervention in IL-23-mediated pathologies.
Collapse
|
15
|
Li ZH, Wang YF, He DD, Zhang XM, Zhou YL, Yue H, Huang S, Fu Z, Zhang LY, Mao ZQ, Li S, Zhang CY, Chen X, Fu J. Let-7f-5p suppresses Th17 differentiation via targeting STAT3 in multiple sclerosis. Aging (Albany NY) 2020; 11:4463-4477. [PMID: 31326963 PMCID: PMC6660039 DOI: 10.18632/aging.102093] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022]
Abstract
T helper 17 (Th17) cells are regarded as key factors in the pathogenesis of multiple sclerosis (MS). Although the involvement of certain microRNAs (miRNAs) in the development of MS has been reported, their roles in Th17 cell differentiation and MS pathogenesis remain elusive. In this study, we identified that let-7f-5p expression is significantly downregulated in CD4+ T cells from MS patients and during the process of Th17 differentiation. The overexpression of let-7f-5p suppressed Th17 differentiation, whereas the knockdown of let-7f-5p expression enhanced this progress. We then explored the molecular mechanism through which let-7f-5p suppressed Th17 differentiation and identified signal transducer and activator of transcription 3 (STAT3), a pivotal transcription factor of Th17 cells, as a direct target of let-7f-5p. In contrast to the downregulated expression of let-7f-5p, STAT3 and p-STAT3 protein levels were dramatically upregulated and inversely correlated with let-7f-5p in peripheral blood CD4+ T cells from MS patients. In conclusion, let-7f-5p functions as a potential inhibitor of Th17 differentiation in the pathogenesis of MS by targeting STAT3 and may serve as a new therapeutic target.
Collapse
Affiliation(s)
- Zhi-Hui Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Yi-Fei Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Dan-Dan He
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin Heilongjiang 150086, China
| | - Xue-Mei Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Ying-Lian Zhou
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Hui Yue
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Shan Huang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Zheng Fu
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210093,China
| | - Ling-Yu Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Zhu-Qing Mao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Shuang Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Chen-Yu Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210093,China
| | - Xi Chen
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210093,China
| | - Jin Fu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150086, China
| |
Collapse
|
16
|
STAT3 signaling in myeloid cells promotes pathogenic myelin-specific T cell differentiation and autoimmune demyelination. Proc Natl Acad Sci U S A 2020; 117:5430-5441. [PMID: 32094172 PMCID: PMC7071888 DOI: 10.1073/pnas.1913997117] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Myeloid lineage cells are suspected of having an integral role in pathophysiological processes of multiple sclerosis (MS), but the molecular mechanism(s) governing their effector function remains incompletely understood. We show that STAT3 is activated in myeloid cells near active MS lesions. Conditional deletion of Stat3 in myeloid cells abolished symptoms of experimental autoimmune encephalomyelitis (EAE) by suppressing the generation of pathogenic T cells. Functional and transcriptomic analyses of myeloid cells from wild-type and conditional knockout mice implicated antigen processing/presentation and inflammatory cytokine production as the mechanism underlying the effects of STAT3 deletion on impaired T cell activation. Our data indicate that targeting STAT3 in myeloid cells might be a viable treatment option for autoimmune demyelinating disease. Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the central nervous system. Dysregulation of STAT3, a transcription factor pivotal to various cellular processes including Th17 cell differentiation, has been implicated in MS. Here, we report that STAT3 is activated in infiltrating monocytic cells near active MS lesions and that activation of STAT3 in myeloid cells is essential for leukocyte infiltration, neuroinflammation, and demyelination in experimental autoimmune encephalomyelitis (EAE). Genetic disruption of Stat3 in peripheral myeloid lineage cells abrogated EAE, which was associated with decreased antigen-specific T helper cell responses. Myeloid cells from immunized Stat3 mutant mice exhibited impaired antigen-presenting functions and were ineffective in driving encephalitogenic T cell differentiation. Single-cell transcriptome analyses of myeloid lineage cells from preclinical wild-type and mutant mice revealed that loss of myeloid STAT3 signaling disrupted antigen-dependent cross-activation of myeloid cells and T helper cells. This study identifies a previously unrecognized requisite for myeloid cell STAT3 in the activation of myelin-reactive T cells and suggests myeloid STAT3 as a potential therapeutic target for autoimmune demyelinating disease.
Collapse
|
17
|
Kolić I, Stojković L, Dinčić E, Jovanović I, Stanković A, Živković M. Expression of LEP, LEPR and PGC1A genes is altered in peripheral blood mononuclear cells of patients with relapsing-remitting multiple sclerosis. J Neuroimmunol 2020; 338:577090. [DOI: 10.1016/j.jneuroim.2019.577090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/01/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023]
|
18
|
Yan Z, Yang W, Parkitny L, Gibson SA, Lee KS, Collins F, Deshane JS, Cheng W, Weinmann AS, Wei H, Qin H, Benveniste EN. Deficiency of Socs3 leads to brain-targeted EAE via enhanced neutrophil activation and ROS production. JCI Insight 2019; 5:126520. [PMID: 30939124 DOI: 10.1172/jci.insight.126520] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dysregulation of the JAK/STAT signaling pathway is associated with Multiple Sclerosis (MS) and its mouse model, Experimental Autoimmune Encephalomyelitis (EAE). Suppressors Of Cytokine Signaling (SOCS) negatively regulate the JAK/STAT pathway. We previously reported a severe, brain-targeted, atypical form of EAE in mice lacking Socs3 in myeloid cells (Socs3ΔLysM), which is associated with cerebellar neutrophil infiltration. There is emerging evidence that neutrophils are detrimental in the pathology of MS/EAE, however, their exact function is unclear. Here we demonstrate that neutrophils from the cerebellum of Socs3ΔLysM mice show a hyper-activated phenotype with excessive production of reactive oxygen species (ROS) at the peak of EAE. Neutralization of ROS in vivo delayed the onset and reduced severity of atypical EAE. Mechanistically, Socs3-deficient neutrophils exhibit enhanced STAT3 activation, a hyper-activated phenotype in response to G-CSF, and upon G-CSF priming, increased ROS production. Neutralization of G-CSF in vivo significantly reduced the incidence and severity of the atypical EAE phenotype. Overall, our work elucidates that hypersensitivity of G-CSF/STAT3 signaling in Socs3ΔLysM mice leads to atypical EAE by enhanced neutrophil activation and increased oxidative stress, which may explain the detrimental role of G-CSF in MS patients.
Collapse
Affiliation(s)
- Zhaoqi Yan
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei Yang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Luke Parkitny
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sara A Gibson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kevin S Lee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Forrest Collins
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Wayne Cheng
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amy S Weinmann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hairong Wei
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
19
|
Kvistad SS, Myhr KM, Holmøy T, Benth JŠ, Wergeland S, Beiske AG, Bjerve KS, Hovdal H, Midgard R, Sagen JV, Torkildsen Ø. Serum levels of leptin and adiponectin are not associated with disease activity or treatment response in multiple sclerosis. J Neuroimmunol 2018; 323:73-77. [PMID: 30196837 DOI: 10.1016/j.jneuroim.2018.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 06/29/2018] [Accepted: 07/23/2018] [Indexed: 01/07/2023]
Abstract
Adipokines secreted by fatty tissue have inflammatory properties and are suggested biomarkers of MS disease activity. To assess this, 88 MS patients were followed with nine repeated measurements of leptin and adiponectin and 12 magnetic resonance imaging (MRI) scans for two years; six months without any immunomodulatory treatment followed by 18 months during interferon-beta (IFNB) treatment. Serum levels of leptin dropped and adiponectin increased upon initiation of IFNB-therapy, but were not associated with clinical or MRI disease activity or with treatment response. Our findings indicate that leptin and adiponectin are not useful as biomarkers of MS disease activity.
Collapse
Affiliation(s)
- Silje Stokke Kvistad
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Kjell-Morten Myhr
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Norwegian Multiple Sclerosis Registry and Biobank, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jūratė Šaltytė Benth
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
| | - Stig Wergeland
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Norwegian Multiple Sclerosis Registry and Biobank, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | | | - Kristian S Bjerve
- Department of Medical Biochemistry, St. Olav's Hospital, Trondheim University Hospital, Norway; Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Harald Hovdal
- Department of Neurology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Rune Midgard
- Department of Neurology, Molde Hospital, Norway; Unit for Applied Clinical Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jørn V Sagen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Hormone Laboratory, Department of Laboratory Medicine and Pathology, Haukeland University Hospital, Bergen, Norway
| | - Øivind Torkildsen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
20
|
Alwarawrah Y, Kiernan K, MacIver NJ. Changes in Nutritional Status Impact Immune Cell Metabolism and Function. Front Immunol 2018; 9:1055. [PMID: 29868016 PMCID: PMC5968375 DOI: 10.3389/fimmu.2018.01055] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Immune cell function and metabolism are closely linked. Many studies have now clearly demonstrated that alterations in cellular metabolism influence immune cell function and that, conversely, immune cell function determines the cellular metabolic state. Less well understood, however, are the effects of systemic metabolism or whole organism nutritional status on immune cell function and metabolism. Several studies have demonstrated that undernutrition is associated with immunosuppression, which leads to both increased susceptibility to infection and protection against several types of autoimmune disease, whereas overnutrition is associated with low-grade, chronic inflammation that increases the risk of metabolic and cardiovascular disease, promotes autoreactivity, and disrupts protective immunity. Here, we review the effects of nutritional status on immunity and highlight the effects of nutrition on circulating cytokines and immune cell populations in both human studies and mouse models. As T cells are critical members of the immune system, which direct overall immune response, we will focus this review on the influence of systemic nutritional status on T cell metabolism and function. Several cytokines and hormones have been identified which mediate the effects of nutrition on T cell metabolism and function through the expression and action of key regulatory signaling proteins. Understanding how T cells are sensitive to both inadequate and overabundant nutrients may enhance our ability to target immune cell metabolism and alter immunity in both malnutrition and obesity.
Collapse
Affiliation(s)
- Yazan Alwarawrah
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Kaitlin Kiernan
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Nancie J MacIver
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States.,Department of Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
21
|
Xie XF, Huang XH, Shen AZ, Li J, Sun YH. Association between circulating leptin levels and multiple sclerosis: a systematic review and meta-analysis. Postgrad Med J 2018; 94:278-283. [PMID: 29581230 DOI: 10.1136/postgradmedj-2017-135397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/06/2018] [Accepted: 03/03/2018] [Indexed: 01/11/2023]
Abstract
AimLeptin, synthesised by adipocytes, has been identified as a hormone that can influence inflammatory activity. Several studies have investigated leptin levels in patients with multiple sclerosis (MS), but the results are not consistent. This study aims to derive a more precise evaluation on the relationship between circulating leptin levels and MS.DesignA comprehensive literature searched up to July 2017 was conducted to evaluate the association of circulating leptin levels and MS. The random-effect model was applied to calculate pooled standardised mean difference (SMD) and its 95% CI.Main outcome measuresCirculating leptin levels of patients with MS and healthy controls.ResultsOf 2155 studies identified, 33 met eligibility criteria and 9 studies with 645 patients with MS and 586 controls were finally included in the meta-analysis. Meta-analysis revealed that, compared with the healthy control group, the MS group had significantly higher plasma/serum leptin levels, with the SMD of 0.70% and 95% CI (0.24 to 1.15). Subgroup analyses suggested that the leptin levels of patients with MS were associated with region, age, study sample size, measurement type, gender and blood sample type.ConclusionOverall, our study suggests that patients with MS have a significantly higher leptin level than in healthy controls. Further mechanism studies and longitudinal large cohort studies are still needed to further reveal the role of leptin in the pathogenesis of MS.
Collapse
Affiliation(s)
- Xue-Feng Xie
- Department of Basic and Clinical Pharmacology, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Hui Huang
- Department of Basic and Clinical Pharmacology, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Ai-Zong Shen
- Department of Pharmacy, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Jun Li
- Department of Basic and Clinical Pharmacology, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Ye-Huan Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
22
|
Huitema MJD, Schenk GJ. Insights into the Mechanisms That May Clarify Obesity as a Risk Factor for Multiple Sclerosis. Curr Neurol Neurosci Rep 2018; 18:18. [PMID: 29525910 PMCID: PMC5845596 DOI: 10.1007/s11910-018-0827-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The proportion to which genetic and environmental factors contribute to the etiology of multiple sclerosis (MS) is still incompletely understood. An interesting association between MS etiology and obesity has recently been shown although the mechanisms underlying this association are still unknown. We propose deregulated gut microbiota and increased leptin levels as possible mechanisms underlying MS etiology in obese individuals. RECENT FINDINGS Alterations in the human gut microbiota and leptin levels have recently been established as immune modulators in both MS patients and obese individuals. A resemblance between pro-inflammatory bacterial profiles in MS and obese individuals was observed. Furthermore, elevated leptin levels push the immune system towards a more pro-inflammatory state and inhibit the regulatory immune response. Deregulated gut microbiota and elevated leptin levels may explain the increased risk of developing MS in obese individuals. Further research to confirm causality is warranted.
Collapse
Affiliation(s)
- Marije J D Huitema
- Faculty of medicine, VU University, Van der Boechorststraat 7, 1081 BT, Amsterdam, Netherlands
| | - Geert J Schenk
- Department of Anatomy and Neurosciences, Neuroscience Amsterdam, VUmc MS Center Amsterdam, VU University Medical Center, De Boelelaan 1108, 1081 HV, Amsterdam, Netherlands.
| |
Collapse
|
23
|
Ozkul C, Guclu-Gunduz A, Irkec C, Fidan I, Aydin Y, Ozkan T, Yazici G. Effect of combined exercise training on serum brain-derived neurotrophic factor, suppressors of cytokine signaling 1 and 3 in patients with multiple sclerosis. J Neuroimmunol 2018; 316:121-129. [DOI: 10.1016/j.jneuroim.2018.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/29/2017] [Accepted: 01/02/2018] [Indexed: 01/23/2023]
|
24
|
Cardiovascular Autonomic Dysfunction: Link Between Multiple Sclerosis Osteoporosis and Neurodegeneration. Neuromolecular Med 2018; 20:37-53. [DOI: 10.1007/s12017-018-8481-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/06/2018] [Indexed: 12/19/2022]
|
25
|
Liu Y, Gibson SA, Benveniste EN, Qin H. Opportunities for Translation from the Bench: Therapeutic Intervention of the JAK/STAT Pathway in Neuroinflammatory Diseases. Crit Rev Immunol 2018; 35:505-27. [PMID: 27279046 DOI: 10.1615/critrevimmunol.2016015517] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathogenic CD4+ T cells and myeloid cells play critical roles in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. These immune cells secrete aberrantly high levels of pro-inflammatory cytokines that pathogenically bridge the innate and adaptive immune systems and damage neurons and oligodendrocytes. These cytokines include interleukin-2 (IL-2), IL-6, IL-12, IL-21, IL-23, granulocyte macrophage-colony stimulating factor (GM-CSF), and interferon-γ (IFN-γ). It is, therefore, not surprising that both the dysregulated expression of these cytokines and the subsequent activation of their downstream signaling cascades is a common feature in MS/EAE. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is utilized by numerous cytokines for signal transduction and is essential for the development and regulation of immune responses. Unbridled activation of the JAK/STAT pathway by pro-inflammatory cytokines has been demonstrated to be critically involved in the pathogenesis of MS/EAE. In this review, we discuss recent advancements in our understanding of the involvement of the JAK/STAT signaling pathway in the pathogenesis of MS/EAE, with a particular focus on therapeutic approaches to target the JAK/STAT pathway.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294; Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sara A Gibson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| |
Collapse
|
26
|
Mokhtarzade M, Ranjbar R, Majdinasab N, Patel D, Molanouri Shamsi M. Effect of aerobic interval training on serum IL-10, TNFα, and adipokines levels in women with multiple sclerosis: possible relations with fatigue and quality of life. Endocrine 2017; 57:262-271. [PMID: 28616851 DOI: 10.1007/s12020-017-1337-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 05/29/2017] [Indexed: 01/13/2023]
Abstract
PURPOSE Multiple sclerosis is associated with immune system dysfunction and chronic inflammation; however, possible relations between immunologic and metabolic factors and some psychological indexes such as fatigue and quality of life, especially in relation to exercise training, have not yet been investigated. The present study was designed to investigate the effect of aerobic interval training on interleukin-10/tumor necrosis factor ratio and adipokine (leptin and adiponectin) concentrations in women with multiple sclerosis. Furthermore, the relationship between these factors with fatigue and quality of life were assessed. METHODS Forty women with multiple sclerosis (Expanded Disability Status Scale ≤3) were randomized into either a non-exercising control or training group. The training group performed 8-weeks of upper and lower limb aerobic interval training. Serum concentrations of tumor necrosis factorα, interleukin-10, leptin, and adiponectin were measured before and after the 8-week intervention. Moreover, antropometric measures and measures for fatigue and quality of life were determined at the onset of and after exercise training. RESULTS The results revealed that leptin and tumor necrosis factorα levels significantly decreased subsequent to the aerobic interval training. Although blood adiponectin levels considerably increased in the training group, interleukin-10 and interleukin-10/tumor necrosis factorα ratio underwent no substantial change after the exercise training. In addition, the aerobic interval training was associated with improvement in fatigue, quality of life, and maximal oxygen consumption. CONCLUSIONS Our findings suggested that aerobic interval training can be an effective strategy for managing the immune system at least by its significant impact on inflammatory cytokines and adipokines levels in women with multiple sclerosis. Additionally, this positive impact improved fatigue and adipose tissue indicators.
Collapse
Affiliation(s)
| | - Rouholah Ranjbar
- Department of Sport Physiology, Shahid Chamran University, Ahvaz, Iran
| | - Nastaran Majdinasab
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Darpan Patel
- School of Nursing, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mehdieh Molanouri Shamsi
- Department of Physical Education & Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
27
|
Esposito S, Bonavita S, Sparaco M, Gallo A, Tedeschi G. The role of diet in multiple sclerosis: A review. Nutr Neurosci 2017; 21:377-390. [PMID: 28338444 DOI: 10.1080/1028415x.2017.1303016] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is a multifactorial, inflammatory, and neurodegenerative disease of the central nervous system, where environmental factors interact with genetic susceptibility. The role of diet on MS has not been comprehensively elucidated; therefore, through an extensive search of relevant literature, this review reports the most significant evidence regarding nutrition as a possible co-factor influencing the inflammatory cascade by acting on both its molecular pathways and gut microbiota. Since nutritional status and dietary habits in MS patients have not been extensively reported, the lack of a scientific-based consensus on dietary recommendation in MS could encourage many patients to experiment alternative dietetic regimens, increasing the risk of malnutrition. This work investigates the health implications of an unbalanced diet in MS, and collects recent findings on nutrients of great interest among MS patients and physicians. The aim of this review is to elucidate the role of an accurate nutritional counseling in MS to move toward a multidisciplinary management of the disease and to encourage future studies demonstrating the role of a healthy diet on the onset and course of MS.
Collapse
Affiliation(s)
- Sabrina Esposito
- a I Clinic of Neurology, Second University of Naples , 80138 , Italy.,b Department of Neuroscience, Psychology, Drug Research and Child Health , University of Florence , Italy
| | - Simona Bonavita
- a I Clinic of Neurology, Second University of Naples , 80138 , Italy.,c MRI Research Center SUN-FISM, Second University of Naples , Italy.,d Institute for Diagnosis and Care "Hermitage Capodimonte" , Naples , Italy
| | - Maddalena Sparaco
- a I Clinic of Neurology, Second University of Naples , 80138 , Italy
| | - Antonio Gallo
- a I Clinic of Neurology, Second University of Naples , 80138 , Italy.,c MRI Research Center SUN-FISM, Second University of Naples , Italy.,d Institute for Diagnosis and Care "Hermitage Capodimonte" , Naples , Italy
| | - Gioacchino Tedeschi
- a I Clinic of Neurology, Second University of Naples , 80138 , Italy.,c MRI Research Center SUN-FISM, Second University of Naples , Italy.,d Institute for Diagnosis and Care "Hermitage Capodimonte" , Naples , Italy
| |
Collapse
|
28
|
Novo AM, Batista S. Multiple Sclerosis: Implications of Obesity in Neuroinflammation. ADVANCES IN NEUROBIOLOGY 2017; 19:191-210. [PMID: 28933066 DOI: 10.1007/978-3-319-63260-5_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Since the discovery of the remarkable properties of adipose tissue as a metabolically active organ, several evidences on the possible link between obesity and the pathogenesis of multiple sclerosis (MS) have been gathered. Obesity in early life, mainly during adolescence, has been proposed as a relevant risk factor for late MS development. Moreover, once MS is initiated, obesity can contribute to increase disease severity by negatively influencing disease progress. Despite the fact that clinical data are not yet conclusive, many biochemical links have been recently disclosed. The "low-grade inflammation" that characterizes obesity can lead to neuroinflammation through different mechanisms, including choroid plexus and blood-brain barrier disruption. Furthermore, it is well known that resident immune cells of central nervous system and peripheral immune cells are involved in the pathogenesis of MS, and adipokines and neuropeptides such as neuropeptide Y may mediate the cross talk between them.
Collapse
Affiliation(s)
- Ana Margarida Novo
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sónia Batista
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal. .,Faculty of Medicine, University of Coimbra, Coimbra, Portugal. .,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
29
|
Jörg S, Grohme DA, Erzler M, Binsfeld M, Haghikia A, Müller DN, Linker RA, Kleinewietfeld M. Environmental factors in autoimmune diseases and their role in multiple sclerosis. Cell Mol Life Sci 2016; 73:4611-4622. [PMID: 27491297 PMCID: PMC5097114 DOI: 10.1007/s00018-016-2311-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/04/2016] [Accepted: 07/18/2016] [Indexed: 12/19/2022]
Abstract
An increase in autoimmune diseases poses a socioeconomic challenge worldwide. Predisposing genetic risk has been identified, yet environmental factors make up a significant part of the risk in disease initiation and propagation. Next to improved hygiene and a gross reduction of infections, changes in dietary habits are one of the most evident Western lifestyle factors potentially associated with the increase in autoimmune diseases. Growing evidence suggests that particularly a typical 'Western diet', rich in saturated fat and salt and related pathologies can have a profound impact on local and systemic immune responses under physiologic and autoimmune conditions such as in multiple sclerosis (MS). In this review, we discuss recent findings on environmental factors influencing autoimmunity with an emphasis on the impact of 'Western diet' on immune homeostasis and gut microbiota in MS.
Collapse
Affiliation(s)
- Stefanie Jörg
- University Hospital Erlangen at the Friedrich-Alexander-University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Diana A Grohme
- Translational Immunology, Department of Clinical Pathobiochemistry, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Melanie Erzler
- Translational Immunology, Department of Clinical Pathobiochemistry, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Marilene Binsfeld
- VIB Laboratory of Translational Immunomodulation & Hasselt University, Diepenbeek, Belgium
| | - Aiden Haghikia
- Department of Neurology, Ruhr-University Bochum, Bochum, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center, An Institutional Cooperation Between the Charité Medical Faculty and the Max-Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Ralf A Linker
- University Hospital Erlangen at the Friedrich-Alexander-University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Markus Kleinewietfeld
- Translational Immunology, Department of Clinical Pathobiochemistry, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany.
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany.
- VIB Laboratory of Translational Immunomodulation & Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
30
|
Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediators Inflamm 2016; 2016:4036232. [PMID: 27721574 PMCID: PMC5046034 DOI: 10.1155/2016/4036232] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/22/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disorder of the Central Nervous System that has been associated with several environmental factors, such as diet and obesity. The possible link between MS and obesity has become more interesting in recent years since the discovery of the remarkable properties of adipose tissue. Once MS is initiated, obesity can contribute to increased disease severity by negatively influencing disease progress and treatment response, but, also, obesity in early life is highly relevant as a susceptibility factor and causally related risk for late MS development. The aim of this review was to discuss recent evidence about the link between obesity, as a chronic inflammatory state, and the pathogenesis of MS as a chronic autoimmune and inflammatory disease. First, we describe the main cells involved in MS pathogenesis, both from neural tissue and from the immune system, and including a new participant, the adipocyte, focusing on their roles in MS. Second, we concentrate on the role of several adipokines that are able to participate in the mediation of the immune response in MS and on the possible cross talk between the latter. Finally, we explore recent therapy that involves the transplantation of adipocyte precursor cells for the treatment of MS.
Collapse
|
31
|
Procaccini C, Santopaolo M, Faicchia D, Colamatteo A, Formisano L, de Candia P, Galgani M, De Rosa V, Matarese G. Role of metabolism in neurodegenerative disorders. Metabolism 2016; 65:1376-90. [PMID: 27506744 DOI: 10.1016/j.metabol.2016.05.018] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 01/12/2023]
Abstract
Along with the increase in life expectancy over the last century, the prevalence of age-related disorders, such as neurodegenerative diseases continues to rise. This is the case of Alzheimer's, Parkinson's, Huntington's diseases and Multiple sclerosis, which are chronic disorders characterized by neuronal loss in motor, sensory or cognitive systems. Accumulating evidence has suggested the presence of a strong correlation between metabolic changes and neurodegeneration. Indeed epidemiologic studies have shown strong associations between obesity, metabolic dysfunction, and neurodegeneration, while animal models have provided insights into the complex relationships between these conditions. In this context, hormones such as leptin, ghrelin, insulin and IGF-1 seem to play a key role in the regulation of neuronal damage, toxic insults and several other neurodegenerative processes. This review aims to presenting the most recent evidence supporting the crosstalk linking energy metabolism and neurodegeneration, and will focus on metabolic manipulation as a possible therapeutic tool in the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy
| | - Marianna Santopaolo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy
| | - Deriggio Faicchia
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II", 80131, Napoli, Italy
| | - Alessandra Colamatteo
- Unità di NeuroImmunologia, IRCCS Fondazione Santa Lucia, 00143, Roma, Italy; Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Baronissi Campus, 84081, Baronissi, Salerno, Italy
| | - Luigi Formisano
- Divisione di Farmacologia, Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, 82100, Benevento, Italy
| | | | - Mario Galgani
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy; Unità di NeuroImmunologia, IRCCS Fondazione Santa Lucia, 00143, Roma, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy.
| |
Collapse
|
32
|
Talaei F. Pathophysiological Concepts in Multiple Sclerosis and the Therapeutic Effects of Hydrogen Sulfide. Basic Clin Neurosci 2016; 7:121-36. [PMID: 27303607 PMCID: PMC4892317 DOI: 10.15412/j.bcn.03070206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Introduction: Multiple sclerosis (MS) is generally known as a manageable but not yet curable autoimmune disease affecting central nervous system. A potential therapeutic approach should possess several properties: Prevent immune system from damaging the brain and spinal cord, promote differentiation of oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes to produce myelin, prevent the formation of fibronectin aggregates by astrocytes to inhibit scar formation, and enhance function of healthy endothelial cells (ECs). Methods: To determine if an increase in sulfur contents through H2S, a potent antioxidant known to induce protective autophagy in cells, could provide the above desired outcomes, peripheral blood mononuclear cells (PBMNCs), OCPs, astrocytes, and ECs were treated with NaHS (50 μM) in vitro. Results: Transmigration assay using EC monolayer showed that serotonin increased migration of PBMNC while pretreatment of EC with NaHS inhibited the migration induced by serotonin treatment. NaHS upregulated proteins involved in immune system response and downregulated PBMNCs- and EC-related adhesion molecules (LFA-1 and VCAM-1). Furthermore, it had a cell expansion inducing effect, altering EC morphology. The effects of NaHS on OPCs and astrocytes were studied compared to mTOR inhibitor rapamycin. In NaHS treated astrocytes the induced fibronectin production was partially inhibited while rapamycin almost fully inhibited fibronectin production. NaHS slowed but did not inhibit the differentiation of OCPs or the production of myelin compared to rapamycin. Conclusion: The in vitro results point to the potential therapeutic application of hydrogen sulfide releasing molecules or health-promoting sulfur compounds in MS.
Collapse
Affiliation(s)
- Fatemeh Talaei
- Novel Drug Delivery Systems Lab, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Agirrezabal I, Palacios R, Moreno B, Sepulcre J, Abernathy A, Saiz A, Llufriu S, Comabella M, Montalban X, Martinez A, Arteta D, Villoslada P. Increased expression of dedicator-cytokinesis-10, caspase-2 and Synaptotagmin-like 2 is associated with clinical disease activity in multiple sclerosis. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s40893-016-0009-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
34
|
Farrokhi M, Dabirzadeh M, Fadaee E, Beni AA, Saadatpour Z, Rezaei A, Heidari Z. Polymorphism in Leptin and Leptin Receptor Genes May Modify Leptin Levels and Represent Risk Factors for Multiple Sclerosis. Immunol Invest 2016; 45:328-35. [DOI: 10.3109/08820139.2016.1157811] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
35
|
Palavra F, Almeida L, Ambrósio AF, Reis F. Obesity and brain inflammation: a focus on multiple sclerosis. Obes Rev 2016; 17:211-24. [PMID: 26783119 DOI: 10.1111/obr.12363] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 10/25/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
The increase in prevalence of obesity in industrialized societies is an indisputable fact. However, the apparent passive role played by adipocytes, in pathophysiological terms, has been gradually substituted by a metabolically active performance, relevant to many biochemical mechanisms that may contribute to a chronic low-grade inflammatory status, which increasingly imposes itself as a key feature of obesity. This chronic inflammatory status will have to be integrated into the complex equation of many diseases in which inflammation plays a crucial role. Multiple sclerosis (MS) is a chronic inflammatory condition typically confined to the central nervous system, and many work has been produced to find possible points of contact between the biology of this immune-mediated disease and obesity. So far, clinical data are not conclusive, but many biochemical features have been recently disclosed. Brain inflammation has been implicated in some of the mechanisms that lead to obesity, which has also been recognized as an important player in inducing some degree of immune dysfunction. In this review, we collected evidence that allows establishing bridges between obesity and MS. After considering epidemiological controversies, we will focus on possible shared mechanisms, as well as on the potential contributions that disease-modifying drugs may have on this apparent relationship of mutual interference.
Collapse
Affiliation(s)
- F Palavra
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal
| | - L Almeida
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - A F Ambrósio
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| | - F Reis
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
36
|
McCormick SM, Heller NM. Regulation of Macrophage, Dendritic Cell, and Microglial Phenotype and Function by the SOCS Proteins. Front Immunol 2015; 6:549. [PMID: 26579124 PMCID: PMC4621458 DOI: 10.3389/fimmu.2015.00549] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022] Open
Abstract
Macrophages are innate immune cells of dynamic phenotype that rapidly respond to external stimuli in the microenvironment by altering their phenotype to respond to and to direct the immune response. The ability to dynamically change phenotype must be carefully regulated to prevent uncontrolled inflammatory responses and subsequently to promote resolution of inflammation. The suppressor of cytokine signaling (SOCS) proteins play a key role in regulating macrophage phenotype. In this review, we summarize research to date from mouse and human studies on the role of the SOCS proteins in determining the phenotype and function of macrophages. We will also touch on the influence of the SOCS on dendritic cell (DC) and microglial phenotype and function. The molecular mechanisms of SOCS function in macrophages and DCs are discussed, along with how dysregulation of SOCS expression or function can lead to alterations in macrophage/DC/microglial phenotype and function and to disease. Regulation of SOCS expression by microRNA is discussed. Novel therapies and unanswered questions with regard to SOCS regulation of monocyte-macrophage phenotype and function are highlighted.
Collapse
Affiliation(s)
- Sarah M McCormick
- Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA
| | - Nicola M Heller
- Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA ; Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|
37
|
Wieghofer P, Prinz M. Genetic manipulation of microglia during brain development and disease. Biochim Biophys Acta Mol Basis Dis 2015; 1862:299-309. [PMID: 26432479 DOI: 10.1016/j.bbadis.2015.09.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Peter Wieghofer
- Institute of Neuropathology, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Germany.
| |
Collapse
|
38
|
Leptin as a Neuroprotector and a Central Nervous System Functional Stability Factor. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s11055-015-0120-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
|
40
|
Aguilar-Valles A, Inoue W, Rummel C, Luheshi GN. Obesity, adipokines and neuroinflammation. Neuropharmacology 2015; 96:124-34. [PMID: 25582291 DOI: 10.1016/j.neuropharm.2014.12.023] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 12/14/2022]
Abstract
Global levels of obesity are reaching epidemic proportions, leading to a dramatic increase in incidence of secondary diseases and the significant economic burden associated with their treatment. These comorbidities include diabetes, cardiovascular disease, and some psychopathologies, which have been linked to a low-grade inflammatory state. Obese individuals exhibit an increase in circulating inflammatory mediators implicated as the underlying cause of these comorbidities. A number of these molecules are also manufactured and released by white adipose tissue (WAT), in direct proportion to tissue mass and are collectively known as adipokines. In the current review we focused on the role of two of the better-studied members of this family namely, leptin and adiponectin, with particular emphasis on their role in neuro-immune interactions, neuroinflammation and subsequent brain diseases. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Argel Aguilar-Valles
- Department of Neuroscience, Université de Montréal and Goodman Cancer Centre, Department of Biochemistry, McGill University, Montréal, Canada
| | - Wataru Inoue
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Christoph Rummel
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Giamal N Luheshi
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec H4H 1R3, Canada.
| |
Collapse
|
41
|
Procaccini C, Pucino V, Mantzoros CS, Matarese G. Leptin in autoimmune diseases. Metabolism 2015; 64:92-104. [PMID: 25467840 DOI: 10.1016/j.metabol.2014.10.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022]
Abstract
The past twenty years of research on leptin has provided crucial information on the link between metabolic state and immune system function. Adipocytes influence not only the endocrine system but also the immune response, through several cytokine-like mediators known as adipokines, which include leptin. Initially described as an antiobesity hormone, leptin has subsequently been shown also to influence hematopoiesis, thermogenesis, reproduction, angiogenesis, and more importantly immune homeostasis. As a cytokine, leptin can affect thymic homeostasis and the secretion of acute-phase reactants such as interleukin-1 (IL-1) and tumor-necrosis factor-alpha (TNF-α). Leptin links nutritional status and proinflammatory T helper 1 (Th1) immune responses and the decrease in leptin plasma concentration during food deprivation leads to impaired immune function. Conversely, elevated circulating leptin levels in obesity appear to contribute to the low-grade inflammatory background which makes obese individuals more susceptible to increased risk of developing cardiovascular diseases, diabetes, or degenerative disease including autoimmunity and cancer. In this review, we provide an overview of recent advances on the role of leptin in the pathogenesis of several autoimmune disorders that may be of particular relevance in the modulation of the autoimmune attack through metabolic-based therapeutic approaches.
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy
| | - Valentina Pucino
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy
| | - Christos S Mantzoros
- Section of Endocrinology, Boston VA Healthcare System, Jamaica Plain, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine, Boston Medical Center, Boston University, 72 Evans Street, Boston, MA 02217, USA
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Facoltà di Medicina e Chirurgia, Università di Salerno, Baronissi Campus, 84081 Baronissi, Salerno, Italy; IRCCS-MultiMedica, 20138 Milano, Italy.
| |
Collapse
|
42
|
Serum leptin levels in treatment-naive patients with clinically isolated syndrome or relapsing-remitting multiple sclerosis. Autoimmune Dis 2014; 2014:486282. [PMID: 25505980 PMCID: PMC4254075 DOI: 10.1155/2014/486282] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 11/25/2022] Open
Abstract
Several studies have investigated leptin levels in patients with multiple sclerosis (MS) with somewhat conflicting results. They have all focused on patients with established relapsing-remitting (RR) MS but have not specifically looked at patients with clinically isolated syndrome (CIS) suggestive of MS, in the early stages of disease. In this study, serum leptin levels were measured in 89 treatment-naïve patients with CIS (53 patients) or RRMS (36 patients) and 73 controls searching for differences between the groups and for associations with several disease parameters. The expected significant sexual dimorphism in leptin levels (higher levels in females) was observed in both MS patients and controls. Increased leptin levels were found in female patients with RRMS compared to female controls (P = .003) and female CIS patients (P = .001). Female CIS patients had comparable levels to controls. Leptin levels correlated positively to disease duration, but not to EDSS, in female patients with RRMS. The results of the present study do not indicate involvement of leptin in the early stages of MS. Normal leptin levels in patients with CIS suggest that leptin does not have a pathogenic role. The ratio leptin/BMI increases during disease course in female MS patients in a time-dependent and disability-independent manner.
Collapse
|
43
|
Walker DG, Whetzel AM, Lue LF. Expression of suppressor of cytokine signaling genes in human elderly and Alzheimer's disease brains and human microglia. Neuroscience 2014; 302:121-37. [PMID: 25286386 DOI: 10.1016/j.neuroscience.2014.09.052] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/23/2014] [Accepted: 09/23/2014] [Indexed: 12/16/2022]
Abstract
Multiple cellular systems exist to prevent uncontrolled inflammation in brain tissues; the suppressor of cytokine signaling (SOCS) proteins have key roles in these processes. SOCS proteins are involved in restricting cellular signaling pathways by enhancing the degradation of activated receptors and removing the stimuli for continued activation. There are eight separate SOCS genes that code for proteins with similar structures and properties. All SOCS proteins can reduce signaling of activated transcription factors Janus kinase (JAK) and signal transducer and activator of transcription (STAT), but they also regulate many other signaling pathways. SOCS-1 and SOCS-3 have particular roles in regulating inflammatory processes. Chronic inflammation is a key feature of the pathology present in Alzheimer's disease (AD)-affected brains resulting from responses to amyloid plaques or neurofibrillary tangles, the pathological hallmarks of AD. The goal of this study was to examine SOCS gene expression in human non-demented (ND) and AD brains and in human brain-derived microglia to determine if AD-related pathology resulted in a deficit of these critical molecules. We demonstrated that SOCS-1, SOCS-2, SOCS-3 and cytokine-inducible SH2 containing protein (CIS) mRNA expression was increased in amyloid beta peptide (Aβ)- and inflammatory-stimulated microglia, while SOCS-6 mRNA expression was decreased by both types of treatments. Using human brain samples from the temporal cortex from ND and AD cases, SOCS-1 through SOCS-7 and CIS mRNA and SOCS-1 through SOCS-7 protein could be detected constitutively in ND and AD human brain samples. Although, the expression of key SOCS genes did not change to a large extent as a result of AD pathology, there were significantly increased levels of SOCS-2, SOCS-3 and CIS mRNA and increased protein levels of SOCS-4 and SOCS-7 in AD brains. In summary, there was no evidence of a deficit of these key inflammatory regulating proteins in aged or AD brains.
Collapse
Affiliation(s)
- D G Walker
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - A M Whetzel
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - L-F Lue
- Laboratory of NeuroRegeneration, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| |
Collapse
|
44
|
Bogie JFJ, Stinissen P, Hendriks JJA. Macrophage subsets and microglia in multiple sclerosis. Acta Neuropathol 2014; 128:191-213. [PMID: 24952885 DOI: 10.1007/s00401-014-1310-2] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/10/2014] [Accepted: 06/15/2014] [Indexed: 12/11/2022]
Abstract
Along with microglia and monocyte-derived macrophages, macrophages in the perivascular space, choroid plexus, and meninges are the principal effector cells in neuroinflammatory and neurodegenerative disorders. These phagocytes are highly heterogeneous cells displaying spatial- and temporal-dependent identities in the healthy, injured, and inflamed CNS. In the last decade, researchers have debated on whether phagocytes subtypes and phenotypes are pathogenic or protective in CNS pathologies. In the context of this dichotomy, we summarize and discuss the current knowledge on the spatiotemporal physiology of macrophage subsets and microglia in the healthy and diseased CNS, and elaborate on factors regulating their behavior. In addition, the impact of macrophages present in lymphoid organs on CNS pathologies is defined. The prime focus of this review is on multiple sclerosis (MS), which is characterized by inflammation, demyelination, neurodegeneration, and CNS repair, and in which microglia and macrophages have been extensively scrutinized. On one hand, microglia and macrophages promote neuroinflammatory and neurodegenerative events in MS by releasing inflammatory mediators and stimulating leukocyte activity and infiltration into the CNS. On the other hand, microglia and macrophages assist in CNS repair through the production of neurotrophic factors and clearance of inhibitory myelin debris. Finally, we define how microglia and macrophage physiology can be harnessed for new therapeutics aimed at suppressing neuroinflammatory and cytodegenerative events, as well as promoting CNS repair. We conclude that microglia and macrophages are highly dynamic cells displaying disease stage and location-specific fates in neurological disorders. Changing the physiology of divergent phagocyte subsets at particular disease stages holds promise for future therapeutics for CNS pathologies.
Collapse
Affiliation(s)
- Jeroen F J Bogie
- Hasselt University, Biomedisch Onderzoeksinstituut and Transnationale Universiteit Limburg, School of Life Sciences, Diepenbeek, Belgium
| | | | | |
Collapse
|
45
|
Sedeño-Monge V, Arcega-Revilla R, Rojas-Morales E, Santos-López G, Perez-García JC, Sosa-Jurado F, Vallejo-Ruiz V, Solis-Morales CL, Aguilar-Rosas S, Reyes-Leyva J. Quantitative analysis of the suppressors of cytokine signaling 1 and 3 in peripheral blood leukocytes of patients with multiple sclerosis. J Neuroimmunol 2014; 273:117-9. [PMID: 24951315 DOI: 10.1016/j.jneuroim.2014.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/20/2014] [Accepted: 05/27/2014] [Indexed: 01/22/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by a triad of inflammation, demyelination and gliosis. Because the suppressors of cytokine signaling (Socs) regulate the immune response, we quantified SOCS1 and SOCS3 transcription in peripheral blood leukocytes of patients with MS. SOCS1 transcription decreased significantly in MS patients compared with neurologically healthy persons (0.08±0.02 vs. 1.02±0.23; p=0.0001); while SOCS3 transcription increased in MS patients compared with controls (2.76±0.66 vs. 1.03±0.27; p=0.0008). Our results showed an imbalance of SOCS1 and SOCS3 transcription in MS patients, and a moderated negative correlation between them (Spearman's r=-0.57; p=0.0003).
Collapse
Affiliation(s)
- Virginia Sedeño-Monge
- Facultad de Medicina, Universidad Popular Autónoma del Estado de Puebla, Puebla, México.
| | - Raúl Arcega-Revilla
- Servicio de Neurología, UMAE, Hospital de Especialidades, Instituto Mexicano del Seguro Social, Puebla, México
| | | | - Gerardo Santos-López
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla, México
| | | | - Francisca Sosa-Jurado
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla, México
| | - Verónica Vallejo-Ruiz
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla, México
| | | | - Salvador Aguilar-Rosas
- Departamento de Ciencias Biológicas, Universidad Popular Autónoma del Estado de Puebla, Puebla, México
| | - Julio Reyes-Leyva
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla, México
| |
Collapse
|
46
|
Procaccini C, Pucino V, De Rosa V, Marone G, Matarese G. Neuro-endocrine networks controlling immune system in health and disease. Front Immunol 2014; 5:143. [PMID: 24778633 PMCID: PMC3985001 DOI: 10.3389/fimmu.2014.00143] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/19/2014] [Indexed: 01/03/2023] Open
Abstract
The nervous and immune systems have long been considered as compartments that perform separate and different functions. However, recent clinical, epidemiological, and experimental data have suggested that the pathogenesis of several immune-mediated disorders, such as multiple sclerosis (MS), might involve factors, hormones, and neural mediators that link the immune and nervous system. These molecules are members of the same superfamily, which allow the mutual and bi-directional neural-immune interaction. More recently, the discovery of leptin, one of the most abundant adipocyte-derived hormones that control food intake and metabolism, has suggested that nutritional/metabolic status, acting at central level, can control immune self-tolerance, since it promotes experimental autoimmune encephalomyelitis, an animal model of MS. Here, we summarize the most recent advances and the key players linking the central nervous system, immune tolerance, and the metabolic status. Understanding this coordinated interaction may pave the way for novel therapeutic approaches to increase host defense and suppress immune-mediated disorders.
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche , Napoli , Italy
| | - Valentina Pucino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II" , Napoli , Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche , Napoli , Italy ; Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia , Roma , Italy
| | - Gianni Marone
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II" , Napoli , Italy ; Centro Interdipartimentale di Ricerca in Scienze Immunologiche di Base e Cliniche, Università di Napoli "Federico II" , Napoli , Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno , Salerno , Italy ; IRCCS Multimedica , Milano , Italy
| |
Collapse
|
47
|
Procaccini C, Pucino V, De Rosa V, Marone G, Matarese G. Neuro-endocrine networks controlling immune system in health and disease. Front Immunol 2014. [PMID: 24778633 DOI: 10.3389/fimmu.2014.00143/abstract] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The nervous and immune systems have long been considered as compartments that perform separate and different functions. However, recent clinical, epidemiological, and experimental data have suggested that the pathogenesis of several immune-mediated disorders, such as multiple sclerosis (MS), might involve factors, hormones, and neural mediators that link the immune and nervous system. These molecules are members of the same superfamily, which allow the mutual and bi-directional neural-immune interaction. More recently, the discovery of leptin, one of the most abundant adipocyte-derived hormones that control food intake and metabolism, has suggested that nutritional/metabolic status, acting at central level, can control immune self-tolerance, since it promotes experimental autoimmune encephalomyelitis, an animal model of MS. Here, we summarize the most recent advances and the key players linking the central nervous system, immune tolerance, and the metabolic status. Understanding this coordinated interaction may pave the way for novel therapeutic approaches to increase host defense and suppress immune-mediated disorders.
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche , Napoli , Italy
| | - Valentina Pucino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II" , Napoli , Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche , Napoli , Italy ; Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia , Roma , Italy
| | - Gianni Marone
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II" , Napoli , Italy ; Centro Interdipartimentale di Ricerca in Scienze Immunologiche di Base e Cliniche, Università di Napoli "Federico II" , Napoli , Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno , Salerno , Italy ; IRCCS Multimedica , Milano , Italy
| |
Collapse
|
48
|
Liang Y, Xu WD, Peng H, Pan HF, Ye DQ. SOCS signaling in autoimmune diseases: molecular mechanisms and therapeutic implications. Eur J Immunol 2014; 44:1265-75. [PMID: 24595859 DOI: 10.1002/eji.201344369] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/08/2014] [Accepted: 02/25/2014] [Indexed: 11/08/2022]
Abstract
Suppressor of cytokine signaling (SOCS) proteins are mainly induced by various cytokines and have been described as classical inhibitors of cytokine signaling. SOCS signaling is involved in the regulation of immune cells, and recent findings suggest that SOCS proteins, especially SOCS1 and SOCS3, are often dysregulated in a wide variety of autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, psoriasis, and multiple sclerosis. Recent studies suggest that SOCS signaling could be therapeutically targeted in various autoimmune diseases. In this review, we discuss recent studies on the role of SOCS proteins in the development and pathogenesis of autoimmune diseases, as well as their clinical implications.
Collapse
Affiliation(s)
- Yan Liang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, People's Republic of China
| | | | | | | | | |
Collapse
|
49
|
The regulation of reactive changes around multiple sclerosis lesions by phosphorylated signal transducer and activator of transcription. J Neuropathol Exp Neurol 2014; 72:1135-44. [PMID: 24226263 DOI: 10.1097/nen.0000000000000011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Activation of signal transducer and activator of transcription 3 (STAT3) by phosphorylation is thought to mediate anti-inflammatory responses to CNS injury. Several studies have reported an increase in phosphorylated STAT3 (pSTAT3) in peripheral T cells and monocytes from patients with multiple sclerosis (MS) during relapses, suggesting that pSTAT3 might represent an inflammatory marker. Here, we examined immunoreactivity for pSTAT3 in brain tissue samples from MS patients and controls. Phosphorylated STAT3 immunoreactivity was sparse within lesions, with no difference between active and inactive lesions. It was, however, significantly greater in white matter (WM) adjacent to active and inactive lesions; moreover, it was significantly greater in WM adjacent to active versus inactive lesions. Phosphorylated STAT3-positive cells were identified as astrocytes and macrophages/microglia. Phosphorylated STAT3 expression was also detected by Western blotting in WM of patients with MS. In comparison, pSTAT3 immunoreactivity was either rare or found focally in brain tissue samples from patients with other neurologic diseases. Our findings show that pSTAT3 does not correlate with inflammatory activity in MS lesions, but that it may play an important role in regulating reactive changes proximal to MS lesions.
Collapse
|
50
|
Lee EB, Mattson MP. The neuropathology of obesity: insights from human disease. Acta Neuropathol 2014; 127:3-28. [PMID: 24096619 DOI: 10.1007/s00401-013-1190-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 02/06/2023]
Abstract
Obesity, a pathologic state defined by excess adipose tissue, is a significant public health problem as it affects a large proportion of individuals and is linked with increased risk for numerous chronic diseases. Obesity is the result of fundamental changes associated with modern society including overnutrition and sedentary lifestyles. Proper energy homeostasis is dependent on normal brain function as the master metabolic regulator, which integrates peripheral signals, modulates autonomic outflow and controls feeding behavior. Therefore, many human brain diseases are associated with obesity. This review explores the neuropathology of obesity by examining brain diseases which either cause or are influenced by obesity. First, several genetic and acquired brain diseases are discussed as a means to understand the central regulation of peripheral metabolism. These diseases range from monogenetic causes of obesity (leptin deficiency, MC4R deficiency, Bardet-Biedl syndrome and others) to complex neurodevelopmental disorders (Prader-Willi syndrome and Sim1 deficiency) and neurodegenerative conditions (frontotemporal dementia and Gourmand's syndrome) and serve to highlight the central regulatory mechanisms which have evolved to maintain energy homeostasis. Next, to examine the effect of obesity on the brain, chronic neuropathologic conditions (epilepsy, multiple sclerosis and Alzheimer's disease) are discussed as examples of obesity leading to maladaptive processes which exacerbate chronic disease. Thus, obesity is associated with multiple pathways including abnormal metabolism, altered hormonal signaling and increased inflammation which act in concert to promote downstream neuropathology. Finally, the effect of anti-obesity interventions is discussed in terms of brain structure and function. Together, understanding human diseases and anti-obesity interventions leads to insights into the bidirectional interaction between peripheral metabolism and central brain function, highlighting the need for continued clinicopathologic and mechanistic studies of the neuropathology of obesity.
Collapse
|