Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Brain Behav Immun ; 110: 237-244, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36893922

RESUMEN

Frailty and a failing immune system lead to significant morbidities in the final years of life and bring along a significant burden on healthcare systems. The good news is that regular exercise provides an effective countermeasure for losing muscle tissue when we age while supporting proper immune system functioning. For a long time, it was assumed that exercise-induced immune responses are predominantly mediated by myeloid cells, but it has become evident that they receive important help from T lymphocytes. Skeletal muscles and T cells interact, not only in muscle pathology but also during exercise. In this review article, we provide an overview of the most important aspects of T cell senescence and discuss how these are modulated by exercise. In addition, we describe how T cells are involved in muscle regeneration and growth. A better understanding of the complex interactions between myocytes and T cells throughout all stages of life provides important insights needed to design strategies that effectively combat the wave of age-related diseases the world is currently faced with.


Asunto(s)
Músculo Esquelético , Linfocitos T , Músculo Esquelético/fisiología , Ejercicio Físico/fisiología
2.
Proc Natl Acad Sci U S A ; 117(9): 5028-5038, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32071226

RESUMEN

The brain's endogenous capacity to restore damaged myelin deteriorates during the course of demyelinating disorders. Currently, no treatment options are available to establish remyelination. Chronic demyelination leads to damaged axons and irreversible destruction of the central nervous system (CNS). We identified two promising therapeutic candidates which enhance remyelination: oncostatin M (OSM), a member of the interleukin-6 family, and downstream mediator tissue inhibitor of metalloproteinases-1 (TIMP-1). While remyelination was completely abrogated in OSMRß knockout (KO) mice, OSM overexpression in the chronically demyelinated CNS established remyelination. Astrocytic TIMP-1 was demonstrated to play a pivotal role in OSM-mediated remyelination. Astrocyte-derived TIMP-1 drove differentiation of oligodendrocyte precursor cells into mature oligodendrocytes in vitro. In vivo, TIMP-1 deficiency completely abolished spontaneous remyelination, phenocopying OSMRß KO mice. Finally, TIMP-1 was expressed by human astrocytes in demyelinated multiple sclerosis lesions, confirming the human value of our findings. Taken together, OSM and its downstream mediator TIMP-1 have the therapeutic potential to boost remyelination in demyelinating disorders.


Asunto(s)
Astrocitos/metabolismo , Oncostatina M/metabolismo , Remielinización/fisiología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Astrocitos/patología , Axones , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Humanos , Interleucina-6/metabolismo , Ratones , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina , Células Precursoras de Oligodendrocitos , Inhibidor Tisular de Metaloproteinasa-1/genética
3.
Acta Neuropathol ; 144(2): 259-281, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35666306

RESUMEN

Oncostatin M (OSM) is an IL-6 family member which exerts neuroprotective and remyelination-promoting effects after damage to the central nervous system (CNS). However, the role of OSM in neuro-inflammation is poorly understood. Here, we investigated OSM's role in pathological events important for the neuro-inflammatory disorder multiple sclerosis (MS). We show that OSM receptor (OSMRß) expression is increased on circulating lymphocytes of MS patients, indicating their elevated responsiveness to OSM signalling. In addition, OSM production by activated myeloid cells and astrocytes is increased in MS brain lesions. In experimental autoimmune encephalomyelitis (EAE), a preclinical model of MS, OSMRß-deficient mice exhibit milder clinical symptoms, accompanied by diminished T helper 17 (Th17) cell infiltration into the CNS and reduced BBB leakage. In vitro, OSM reduces BBB integrity by downregulating the junctional molecules claudin-5 and VE-cadherin, while promoting secretion of the Th17-attracting chemokine CCL20 by inflamed BBB-endothelial cells and reactive astrocytes. Using flow cytometric fluorescence resonance energy transfer (FRET) quantification, we found that OSM-induced endothelial CCL20 promotes activation of lymphocyte function-associated antigen 1 (LFA-1) on Th17 cells. Moreover, CCL20 enhances Th17 cell adhesion to OSM-treated inflamed endothelial cells, which is at least in part ICAM-1 mediated. Together, these data identify an OSM-CCL20 axis, in which OSM contributes significantly to BBB impairment during neuro-inflammation by inducing permeability while recruiting Th17 cells via enhanced endothelial CCL20 secretion and integrin activation. Therefore, care should be taken when considering OSM as a therapeutic agent for treatment of neuro-inflammatory diseases such as MS.


Asunto(s)
Barrera Hematoencefálica , Encefalomielitis Autoinmune Experimental , Oncostatina M , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Oncostatina M/metabolismo , Oncostatina M/farmacología , Subunidad beta del Receptor de Oncostatina M/biosíntesis , Subunidad beta del Receptor de Oncostatina M/genética , Células Th17/metabolismo , Células Th17/patología
4.
Glia ; 63(10): 1729-37, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25921393

RESUMEN

Multiple sclerosis (MS) is a chronic disabling disease of the central nervous system (CNS), in which destruction of myelin sheaths leads to disturbed axonal conduction. Available MS therapies modulate the immune response, but are unable to prevent neurological decline. Therefore, great efforts are made to develop therapies that limit demyelination and axonal degeneration. Oncostatin M (OSM), a member of the interleukin (IL)-6 cytokine family, is produced in demyelinating lesions of MS patients and stimulates neuronal survival. In this study, we reveal that the OSM receptor (OSMR) was robustly upregulated on microglia/macrophages and astrocytes in the cuprizone-induced demyelination model. While OSMR deficiency led to aggravated demyelination, CNS-targeted OSM treatment largely prevented demyelination. OSM treatment increased IL-4 expression and induced polarization of myeloid cells towards an anti-inflammatory M2 phenotype in vivo. This study reveals a previously uncharacterized and protective role for OSM during demyelination, and indicates that OSM is a promising therapeutic candidate to limit CNS damage in demyelinating diseases including MS.


Asunto(s)
Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/prevención & control , Microglía/metabolismo , Oncostatina M/farmacología , Regulación hacia Arriba/fisiología , Animales , Proteínas de Unión al Calcio/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Quelantes/toxicidad , Cuprizona/toxicidad , Citocinas/genética , Citocinas/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Inhibidores de Crecimiento/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Fenotipo , Factores de Tiempo , Transducción Genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
5.
Brain Behav Immun ; 45: 180-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25514345

RESUMEN

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS), for which current treatments are unable to prevent disease progression. Based on its neuroprotective and neuroregenerating properties, leukemia inhibitory factor (LIF), a member of the interleukin-6 (IL-6) cytokine family, is proposed as a novel candidate for MS therapy. However, its effect on the autoimmune response remains unclear. In this study, we determined how LIF modulates T cell responses that play a crucial role in the pathogenesis of MS. We demonstrate that expression of the LIF receptor was strongly increased on immune cells of MS patients. LIF treatment potently boosted the number of regulatory T cells (Tregs) in CD4(+) T cells isolated from healthy controls and MS patients with low serum levels of IL-6. Moreover, IL-6 signaling was reduced in the donors that responded to LIF treatment in vitro. Our data together with previous findings revealing that IL-6 inhibits Treg development, suggest an opposing function of LIF and IL-6. In a preclinical animal model of MS we shifted the LIF/IL-6 balance in favor of LIF by CNS-targeted overexpression. This increased the number of Tregs in the CNS during active autoimmune responses and reduced disease symptoms. In conclusion, our data show that LIF downregulates the autoimmune response by enhancing Treg numbers, providing further impetus for the use of LIF as a novel treatment for MS and other autoimmune diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-6/inmunología , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/inmunología , Factor Inhibidor de Leucemia/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Estudios de Casos y Controles , Femenino , Humanos , Técnicas In Vitro , Interleucina-6/metabolismo , Factor Inhibidor de Leucemia/farmacología , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Linfocitos T Reguladores/efectos de los fármacos
6.
Mech Ageing Dev ; 220: 111959, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38950628

RESUMEN

Oligodendrocyte precursor cells (OPCs) comprise 5-8 % of the adult glial cell population and stand out as the most proliferative cell type in the central nervous system (CNS). OPCs are responsible for generating oligodendrocytes (OLs), the myelinating cells of the CNS. However, OPC functions decline as we age, resulting in impaired differentiation and inadequate remyelination. This review explores the cellular and molecular changes associated with OPC aging, and their impact on OPC differentiation and functionality. Furthermore, it examines the impact of OPC aging within the context of multiple sclerosis and Alzheimer's disease, both neurodegenerative conditions wherein aged OPCs exacerbate disease progression by impeding remyelination. Moreover, various pharmacological interventions targeting pathways related to senescence and differentiation are discussed as potential strategies to rejuvenate aged OPCs. Enhancing our understanding of OPC aging mechanisms holds promise for developing new therapies to improve remyelination and repair in age-related neurodegenerative disorders.

7.
Mediators Inflamm ; 2013: 685317, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23818742

RESUMEN

Demyelination is one of the pathological hallmarks of multiple sclerosis (MS). To date, no therapy is available which directly potentiates endogenous remyelination. Interleukin-11 (IL-11), a member of the gp130 family of cytokines, is upregulated in MS lesions. Systemic IL-11 treatment was shown to ameliorate clinical symptoms in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. IL-11 modulates immune cells and protects oligodendrocytes in vitro. In this study, the cuprizone-induced demyelination mouse model was used to elucidate effects of IL-11 on de- and remyelination, independent of the immune response. Prophylactic-lentiviral- (LV-) mediated overexpression of IL-11 in mouse brain significantly limited acute demyelination, which was accompanied with the preservation of CC1(+) mature oligodendrocytes (OLs) and a decrease in microglial activation (Mac-2(+)). We further demonstrated that IL-11 directly reduces myelin phagocytosis in vitro. When IL-11 expressing LV was therapeutically applied in animals with extensive demyelination, a significant enhancement of remyelination was observed as demonstrated by Luxol Fast Blue staining and electron microscopy imaging. Our results indicate that IL-11 promotes maturation of NG2(+) OPCs into myelinating CC1(+) OLs and may thus explain the enhanced remyelination. Overall, we demonstrate that IL-11 is of therapeutic interest for MS and other demyelinating diseases by limiting demyelination and promoting remyelination.


Asunto(s)
Sistema Nervioso Central/metabolismo , Enfermedades Desmielinizantes/metabolismo , Interleucina-11/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Sistema Nervioso Central/ultraestructura , Cuprizona/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Humanos , Inmunohistoquímica , Interleucina-11/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Vaina de Mielina/metabolismo
8.
Mol Ther ; 18(4): 684-91, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20068552

RESUMEN

Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS) with an inflammatory and a neurodegenerative component. The neuropoietic cytokine leukemia inhibitory factor (LIF) is expressed in MS lesions, but its effect on lesion development is far from understood. LIF is an interesting candidate for MS therapy, as it has neuroprotective properties and may also promote the survival of myelinating oligodendrocytes (OLGs). However, therapeutic administration of LIF is complicated by its limited ability to cross the blood-brain barrier and its pleiotropic actions outside the CNS. In this study, lentiviral vectors (LVs) were used to achieve stable expression and secretion of LIF in the CNS of adult mice. CNS-targeted expression of LIF significantly reduced demyelination in a murine model of MS. In addition, local expression of LIF ameliorated clinical symptoms with enhanced efficacy compared to systemic treatment with recombinant protein. These findings demonstrate that gene therapeutic administration of LIF is a promising approach to limit lesion burden and clinical symptoms in neuroinflammatory disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Terapia Genética/métodos , Factor Inhibidor de Leucemia/genética , Esclerosis Múltiple/terapia , Animales , Barrera Hematoencefálica/metabolismo , Enfermedades Autoinmunes Desmielinizantes SNC/terapia , Modelos Animales de Enfermedad , Vectores Genéticos , Lentivirus , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Oligodendroglía/patología , Linfocitos T/trasplante
9.
Lancet Gastroenterol Hepatol ; 6(10): 784-792, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34358486

RESUMEN

BACKGROUND: Current treatments for functional dyspepsia have limited efficacy or present safety issues. We aimed to assess spore-forming probiotics in functional dyspepsia as monotherapy or add-on therapy to long-term treatment with proton-pump inhibitors. METHODS: In this single-centre, randomised, double-blind, placebo-controlled pilot trial that took place at University Hospitals Leuven (Leuven, Belgium), adult patients (≥18 years) with functional dyspepsia (as defined by Rome IV criteria, on proton-pump inhibitors or off proton-pump inhibitors) were randomly assigned (1:1) via computer-generated blocked lists, stratified by proton-pump inhibitor status, to receive 8 weeks of treatment with probiotics (Bacillus coagulans MY01 and Bacillus subtilis MY02, 2·5 × 109 colony-forming units per capsule) or placebo consumed twice per day, followed by an open-label extension phase of 8 weeks. Individuals with a history of abdominal surgery, diabetes, coeliac or inflammatory bowel disease, active psychiatric conditions, and use of immunosuppressant drugs, antibiotics, or probiotics in the past 3 months were excluded. All patients and on-site study personnel were masked to treatment allocation in the first 8 weeks. Symptoms, immune activation, and faecal microbiota were assessed and recorded. The primary endpoint was a decrease of at least 0·7 in the postprandial distress syndrome (PDS) score of the Leuven Postprandial Distress Scale in patients with a baseline PDS score of 1 or greater (at least mild symptoms), assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT04030780. FINDINGS: Between June 3, 2019, and March 11, 2020, of 93 individuals assessed for eligibility, we included 68 patients with functional dyspepsia (51 [75%] women, mean age 40·1 years [SD 14·4], 34 [50%] on proton-pump inhibitors). We randomly assigned 32 participants to probiotics and 36 to placebo. The proportion of clinical responders was higher with probiotics (12 [48%] of 25) than placebo (six [20%] of 30; relative risk 1·95 [95% CI 1·07-4·11]; p=0·028). The number of patients with adverse events was similar with probiotics (five [16%] of 32) and placebo (12 [33%] of 36). Two serious adverse events occurring during the open-label phase (appendicitis and syncope in two separate patients) were assessed as unlikely to be related to the study product. INTERPRETATION: In this exploratory study, B coagulans MY01 and B subtilis MY02 were efficacious and safe in the treatment of functional dyspepsia. Participants had potentially beneficial immune and microbial changes, which could provide insights into possible underlying mechanisms as future predictors or treatment targets. FUNDING: MY HEALTH.


Asunto(s)
Suplementos Dietéticos/efectos adversos , Dispepsia/dietoterapia , Dispepsia/fisiopatología , Probióticos/uso terapéutico , Adulto , Bacillus coagulans , Bacillus subtilis , Bélgica/epidemiología , Estudios de Casos y Controles , Método Doble Ciego , Dispepsia/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Placebos/administración & dosificación , Prevalencia , Probióticos/administración & dosificación , Probióticos/efectos adversos , Inhibidores de la Bomba de Protones/uso terapéutico , Seguridad , Esporas/química , Resultado del Tratamiento
10.
Proteomics ; 8(6): 1237-47, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18338825

RESUMEN

Leukemia inhibitory factor (LIF) promotes the survival of oligodendrocytes (OLG) both in vitro and in an animal model of multiple sclerosis. Here, we show that LIF protects mature rat OLG cultures selectively against the combined insult of the proinflammatory cytokines interferon-gamma and tumor necrosis factor-alpha, but it does not protect against oxidative stress nor against staurosporine induced apoptosis. We further demonstrate that LIF activates the janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) and the phosphatidylinositol 3 kinase/Akt pathway in mature OLG. We show that LIF protection is independent of suppressors of cytokine signaling and Bcl-2 mRNA expression levels. To gain further insight into the protective mechanism, a quantitative proteomic approach (DIGE) was applied to identify differentially expressed proteins in LIF-treated OLG. Our results indicate that LIF induces a shift in the cellular machinery toward a prosurvival execution program, illustrated by an enhanced expression of isoforms of the antiapoptotic molecule 14-3-3. These data provide further insight into the mechanisms of LIF-mediated protection of mature OLGs.


Asunto(s)
Proteínas 14-3-3/metabolismo , Apoptosis/efectos de los fármacos , Factor Inhibidor de Leucemia/farmacología , Oligodendroglía/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Oligodendroglía/citología , Oligodendroglía/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Espectrometría de Masa por Ionización de Electrospray , Proteínas Supresoras de la Señalización de Citocinas/genética , Espectrometría de Masas en Tándem , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
11.
J Neuroimmunol ; 204(1-2): 52-7, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18771807

RESUMEN

Leukemia inhibitory factor (LIF) promotes survival of glial cells and neurons during autoimmune and injury responses in the central nervous system (CNS). While various studies indicate that LIF also modulates ongoing inflammatory responses, data on underlying events are lacking. In this study we demonstrate that LIF modulates macrophage function. LIF inhibits the production of oxygen radicals and TNFalpha and stimulates myelin uptake by macrophages. These effects of LIF are accompanied by activation of the JAK/STAT3 signalling pathway. Our findings demonstrate that LIF has anti-inflammatory properties and enhances myelin clearance, implicating that LIF may be an important factor in CNS inflammatory disease.


Asunto(s)
Factor Inhibidor de Leucemia/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Vaina de Mielina/metabolismo , Fagocitosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Receptor gp130 de Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo
12.
Cell Transplant ; 25(6): 1207-18, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26688298

RESUMEN

The aim of this study is to examine whether the histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), can generate dendritic cells (DCs) with a stable tolerogenic phenotype to counteract autoimmune responses in an animal model of multiple sclerosis. We investigated if the tolerogenic potency of DCs could be increased by continuous treatment during in vitro differentiation toward DCs compared to standard 24-h in vitro treatment of already terminally differentiated DCs. We show that in vitro treatment with SAHA reduces the generation of new CD11c(+) DCs out of mouse bone marrow. SAHA-generated DCs show reduced antigen-presenting function as evidenced by a reduction in myelin endocytosis, a decreased MHC II expression, and a failure to upregulate costimulatory molecules upon LPS challenge. In addition, SAHA-generated DCs display a reduction in proinflammatory cytokines and molecules involved in apoptosis induction, inflammatory migration, and TLR signaling, and they are less immunostimulatory compared to untreated DCs. We demonstrated that the underlying mechanism involves a diminished STAT1 phosphorylation and was independent of STAT6 activation. Although in vitro results were promising, SAHA-generated DCs were not able to alleviate the development of experimental autoimmune encephalomyelitis in mice. In vitro washout experiments demonstrated that the tolerogenic phenotype of SAHA-treated DCs is reversible. Taken together, while SAHA potently boosts tolerogenic properties in DCs during the differentiation process in vitro, SAHA-generated DCs were unable to reduce autoimmunity in vivo. Our results imply that caution needs to be taken when developing DC-based therapies to induce tolerance in the context of autoimmune disease.


Asunto(s)
Células Dendríticas/inmunología , Ácidos Hidroxámicos/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Antígeno CD11c/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Endocitosis/efectos de los fármacos , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Fenotipo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Vorinostat
13.
Ann N Y Acad Sci ; 1351: 52-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26100315

RESUMEN

Multiple sclerosis (MS) is a chronic disabling autoimmune disease of the central nervous system. The interleukin (IL)-6 cytokine family plays a crucial role in regulating the immune response in MS. All members of the IL-6 family share the common signal-transducing receptor protein, glycoprotein 130. Although the intracellular signaling of these cytokines seems to be largely overlapping, they have diverse and contrasting effects on the immune response. This review focuses on the effects of the family members IL-6, leukemia inhibitory factor, oncostatin M, and IL-11 on immune cell subsets and how these effects relate to the pathogenesis of MS. Finally, we propose possible avenues to modulate these family members for future MS therapy.


Asunto(s)
Sistema Nervioso Central/inmunología , Inmunomodulación/inmunología , Interleucina-6/inmunología , Esclerosis Múltiple/inmunología , Transducción de Señal/inmunología , Glicoproteínas/metabolismo , Humanos , Interleucina-11/inmunología , Factor Inhibidor de Leucemia/metabolismo , Oncostatina M/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología
14.
Mol Neurobiol ; 50(3): 1142-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24996996

RESUMEN

The family of interleukin (IL)-6 like cytokines plays an important role in the neuroinflammatory response to injury by regulating both neural as well as immune responses. Here, we show that expression of the IL-6 family member oncostatin M (OSM) and its receptor is upregulated after spinal cord injury (SCI). To reveal the relevance of increased OSM signaling in the pathophysiology of SCI, OSM was applied locally after spinal cord hemisection in mice. OSM treatment significantly improved locomotor recovery after mild and severe SCI. Improved recovery in OSM-treated mice was associated with a reduced lesion size. OSM significantly diminished astrogliosis and immune cell infiltration. Thus, OSM limits secondary damage after CNS trauma. In vitro viability assays demonstrated that OSM protects primary neurons in culture from cell death, suggesting that the underlying mechanism involves direct neuroprotective effects of OSM. Furthermore, OSM dose-dependently promoted neurite outgrowth in cultured neurons, indicating that the cytokine plays an additional role in CNS repair. Indeed, our in vivo experiments demonstrate that OSM treatment increases plasticity of serotonergic fibers after SCI. Together, our data show that OSM is produced at the lesion site, where it protects the CNS from further damage and promotes recovery.


Asunto(s)
Neuritas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oncostatina M/farmacología , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Ratones , Neuritas/fisiología , Fármacos Neuroprotectores/uso terapéutico , Oncostatina M/metabolismo , Oncostatina M/uso terapéutico , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Regulación hacia Arriba
15.
J Neuroimmunol ; 255(1-2): 45-53, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23194644

RESUMEN

Transcriptomic and proteomic analyses of multiple sclerosis (MS) lesions indicate alterations in the gamma-aminobutyric acid (GABA) inhibitory system, suggesting its involvement in the disease process. To further elucidate the role of GABA in central nervous system (CNS) inflammation in vivo, the chronic myelin oligodendrocyte glycoprotein (MOG)(35-55) experimental autoimmune encephalomyelitis (EAE) model was used. Daily GABA injections (200mg/kg) from day 3 onwards significantly augmented disease severity, which was associated with increased CNS mRNA expression levels of tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6. GABA-treated mice showed enhanced MOG-dependent proliferation and were skewed towards a T helper 1 phenotype. Moreover, in vitro, the lipopolysaccharide (LPS)-induced increase in interleukin (IL)-6 production by macrophages was enhanced at low GABA concentrations (0.03-0.3mM). In sharp contrast to exogenous GABA administration, endogenous GABA increment by systemic treatment with the GABA-transaminase inhibitor vigabatrin (250mg/kg) had prophylactic as well as therapeutic potential in EAE. Together, these results indicate an immune amplifying role of GABA in neuroinflammatory diseases like MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Inhibición Neural/efectos de los fármacos , Inhibición Neural/inmunología , Ácido gamma-Aminobutírico/administración & dosificación , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , GABAérgicos/administración & dosificación , GABAérgicos/uso terapéutico , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/inmunología , Índice de Severidad de la Enfermedad , Ácido gamma-Aminobutírico/fisiología , Ácido gamma-Aminobutírico/uso terapéutico
16.
Trends Mol Med ; 16(11): 493-500, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20870461

RESUMEN

Therapies for multiple sclerosis (MS) reduce the relapse rate but are unable to stop neurological decline. Here, we evaluate the potential of leukemia inhibitory factor (LIF) as a novel therapeutic in diseases with a neurodegenerative and inflammatory component, such as MS. LIF, which can be a proinflammatory cytokine, can also modulate the immune response in a beneficial way. Recent evidence demonstrates a crucial role of LIF in neuroprotection and axonal regeneration as well as the prevention of demyelination. Finally, LIF is an important survival factor for stem cells and neuronal precursors. Therefore, we propose that LIF is a potential therapeutic candidate for MS.


Asunto(s)
Factor Inhibidor de Leucemia/metabolismo , Esclerosis Múltiple/metabolismo , Animales , Proliferación Celular , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/terapia , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/terapia , Terapia Genética/métodos , Humanos , Factor Inhibidor de Leucemia/genética , Esclerosis Múltiple/genética , Esclerosis Múltiple/terapia , Neuronas/citología , Neuronas/metabolismo , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda