Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 49, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355633

RESUMEN

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) constitute a recently discovered bone-marrow-derived cell type useful for dealing with neuroinflammatory disorders. However, these cells are only formed during inflammatory conditions from immature myeloid cells (IMCs) that acquire immunosuppressive activity, thus being commonly gathered from diseased animals. Then, to obtain a more clinically feasible source, we characterized IMCs directly derived from healthy bone marrow and proved their potential immunosuppressive activity under pathological conditions in vitro. We then explored their neuroprotective potential in a model of human cerebellar ataxia, the Purkinje Cell Degeneration (PCD) mouse, as it displays a well-defined neurodegenerative and neuroinflammatory process that can be also aggravated by invasive surgeries. METHODS: IMCs were obtained from healthy bone marrow and co-cultured with activated T cells. The proliferation and apoptotic rate of the later were analyzed with Tag-it Violet. For in vivo studies, IMCs were transplanted by stereotactic surgery into the cerebellum of PCD mice. We also used sham-operated animals as controls of the surgical effects, as well as their untreated counterparts. Motor behavior of mice was assessed by rotarod test. The Purkinje cell density was measured by immunohistochemistry and cell death assessed with the TUNEL technique. We also analyzed the microglial phenotype by immunofluorescence and the expression pattern of inflammation-related genes by qPCR. Parametric tests were applied depending on the specific experiment: one or two way ANOVA and Student's T test. RESULTS: IMCs were proven to effectively acquire immunosuppressive activity under pathological conditions in vitro, thus acting as MDSCs. Concerning in vivo studios, sham-operated PCD mice suffered detrimental effects in motor coordination, Purkinje cell survival and microglial activation. After intracranial administration of IMCs into the cerebellum of PCD mice, no special benefits were detected in the transplanted animals when compared to untreated mice. Nonetheless, this transplant almost completely prevented the impairments caused by the surgery in PCD mice, probably by the modulation of the inflammatory patterns. CONCLUSIONS: Our work comprise two main translational findings: (1) IMCs can be directly used as they behave as MDSCs under pathological conditions, thus avoiding their gathering from diseased subjects; (2) IMCs are promising adjuvants when performing neurosurgery.


Asunto(s)
Cerebelo , Células Mieloides , Ratones , Humanos , Animales , Células Mieloides/metabolismo , Células de Purkinje/patología , Monocitos , Inmunosupresores
2.
Acta Neuropathol ; 146(2): 263-282, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37243699

RESUMEN

Multiple sclerosis (MS) is a highly heterogeneous demyelinating disease of the central nervous system (CNS) that needs for reliable biomarkers to foresee disease severity. Recently, myeloid-derived suppressor cells (MDSCs) have emerged as an immune cell population with an important role in MS. The monocytic-MDSCs (M-MDSCs) share the phenotype with Ly-6Chi-cells in the MS animal model, experimental autoimmune encephalomyelitis (EAE), and have been retrospectively related to the severity of the clinical course in the EAE. However, no data are available about the presence of M-MDSCs in the CNS of MS patients or its relation with the future disease aggressiveness. In this work, we show for the first time cells exhibiting all the bona-fide phenotypical markers of M-MDSCs associated with MS lesions, whose abundance in these areas appears to be directly correlated with longer disease duration in primary progressive MS patients. Moreover, we show that blood immunosuppressive Ly-6Chi-cells are strongly related to the future severity of EAE disease course. We found that a higher abundance of Ly-6Chi-cells at the onset of the EAE clinical course is associated with a milder disease course and less tissue damage. In parallel, we determined that the abundance of M-MDSCs in blood samples from untreated MS patients at their first relapse is inversely correlated with the Expanded Disability Status Scale (EDSS) at baseline and after a 1-year follow-up. In summary, our data point to M-MDSC load as a factor to be considered for future studies focused on the prediction of disease severity in EAE and MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Células Supresoras de Origen Mieloide , Animales , Ratones , Esclerosis Múltiple/patología , Células Supresoras de Origen Mieloide/patología , Estudios Retrospectivos , Encefalomielitis Autoinmune Experimental/patología , Progresión de la Enfermedad , Ratones Endogámicos C57BL
3.
J Neuroinflammation ; 19(1): 277, 2022 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-36403026

RESUMEN

BACKGROUND: The increasing number of treatments that are now available to manage patients with multiple sclerosis (MS) highlights the need to develop biomarkers that can be used within the framework of individualized medicine. Fingolimod is a disease-modifying treatment that belongs to the sphingosine-1-phosphate receptor modulators. In addition to inhibiting T cell egress from lymph nodes, fingolimod promotes the immunosuppressive activity of myeloid-derived suppressor cells (MDSCs), whose monocytic subset (M-MDSCs) can be used as a biomarker of disease severity, as well as the degree of demyelination and extent of axonal damage in the experimental autoimmune encephalomyelitis (EAE) model of MS. In the present study, we have assessed whether the abundance of circulating M-MDSCs may represent a useful biomarker of fingolimod efficacy in EAE and in the clinical context of MS patients. METHODS: Treatment with vehicle or fingolimod was orally administered to EAE mice for 14 days in an individualized manner, starting the day when each mouse began to develop clinical signs. Peripheral blood from EAE mice was collected previous to treatment and human peripheral blood mononuclear cells (PBMCs) were collected from fingolimod to treat MS patients' peripheral blood. In both cases, M-MDSCs abundance was analyzed by flow cytometry and its relationship with the future clinical affectation of each individual animal or patient was assessed. RESULTS: Fingolimod-treated animals presented a milder EAE course with less demyelination and axonal damage, although a few animals did not respond well to treatment and they invariably had fewer M-MDSCs prior to initiating the treatment. Remarkably, M-MDSC abundance was also found to be an important and specific parameter to distinguish EAE mice prone to better fingolimod efficacy. Finally, in a translational effort, M-MDSCs were quantified in MS patients at baseline and correlated with different clinical parameters after 12 months of fingolimod treatment. M-MDSCs at baseline were highly representative of a good therapeutic response to fingolimod, i.e., patients who met at least two of the criteria used to define non-evidence of disease activity-3 (NEDA-3) 12 months after treatment. CONCLUSION: Our data indicate that M-MDSCs might be a useful predictive biomarker of the response of MS patients to fingolimod.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Células Supresoras de Origen Mieloide , Humanos , Animales , Ratones , Clorhidrato de Fingolimod/uso terapéutico , Células Supresoras de Origen Mieloide/patología , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Leucocitos Mononucleares , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Biomarcadores
4.
Cell Immunol ; 371: 104451, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34781155

RESUMEN

The COVID-19 pandemic has once again brought to the forefront the existence of a tight link between the coagulation/fibrinolytic system and the immunologic processes. Tissue-type plasminogen activator (tPA) is a serine protease with a key role in fibrinolysis by converting plasminogen into plasmin that can finally degrade fibrin clots. tPA is released in the blood by endothelial cells and hepatocytes but is also produced by various types of immune cells including T cells and monocytes. Beyond its role on hemostasis, tPA is also a potent modulator of inflammation and is involved in the regulation of several inflammatory diseases. Here, after a brief description of tPA structure, we review its new functions in adaptive immunity focusing on T cells and antigen presenting cells. We intend to synthesize the recent knowledge on proteolysis- and receptor-mediated effects of tPA on immune response in physiological and pathological context.


Asunto(s)
Coagulación Sanguínea/inmunología , COVID-19/inmunología , Fibrinólisis/inmunología , Inmunidad/inmunología , SARS-CoV-2/inmunología , Activador de Tejido Plasminógeno/inmunología , Células Presentadoras de Antígenos/inmunología , COVID-19/epidemiología , COVID-19/virología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Modelos Inmunológicos , Pandemias , SARS-CoV-2/fisiología , Linfocitos T/inmunología , Activador de Tejido Plasminógeno/metabolismo
5.
Glia ; 69(4): 905-924, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33217041

RESUMEN

The most frequent variant of multiple sclerosis (MS) is the relapsing-remitting form, characterized by symptomatic phases followed by periods of total/partial recovery. Hence, it is possible that these patients can benefit from endogenous agents that control the inflammatory process and favor spontaneous remyelination. In this context, there is increasing interest in the role of myeloid-derived suppressor cells (MDSCs) during the clinical course of experimental autoimmune encephalomyelitis (EAE). MDSCs speed up infiltrated T-cell anergy and apoptosis. In different animal models of MS, a milder disease course is related to higher presence/density of MDSCs in the periphery, and smaller demyelinated lesions in the central nervous system (CNS). These observations lead us to wonder whether MDSCs might not only exert an anti-inflammatory effect but might also have direct influence on oligodendrocyte precursor cells (OPCs) and remyelination. In the present work, we reveal for the first time the relationship between OPCs and MDSCs in EAE, relationship that is guided by the distance from the inflammatory core. We describe the effects of MDSCs on survival, proliferation, as well as potent promoters of OPC differentiation toward mature phenotypes. We show for the first time that osteopontin is remarkably present in the analyzed secretome of MDSCs. The ablation of this cue from MDSCs-secretome demonstrates that osteopontin is the main MDSC effector on these oligodendroglial cells. These data highlight a crucial pathogenic interaction between innate immunity and the CNS, opening ways to develop MDSC- and/or osteopontin-based therapies to promote effective myelin preservation and repair in MS patients.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Células Supresoras de Origen Mieloide , Células Precursoras de Oligodendrocitos , Remielinización , Animales , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/terapia , Humanos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/terapia , Vaina de Mielina , Oligodendroglía , Osteopontina , Secretoma
6.
J Neuroinflammation ; 18(1): 52, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33610187

RESUMEN

BACKGROUND: Tissue plasminogen activator (tPA) is a serine protease involved in fibrinolysis. It is released by endothelial cells, but also expressed by neurons and glial cells in the central nervous system (CNS). Interestingly, this enzyme also contributes to pathological processes in the CNS such as neuroinflammation by activating microglia and increasing blood-brain barrier permeability. Nevertheless, its role in the control of adaptive and innate immune response remains poorly understood. METHODS: tPA effects on myeloid and lymphoid cell response were studied in vivo in the mouse model of multiple sclerosis experimental autoimmune encephalomyelitis and in vitro in splenocytes. RESULTS: tPA-/- animals exhibited less severe experimental autoimmune encephalomyelitis than their wild-type counterparts. This was accompanied by a reduction in both lymphoid and myeloid cell populations in the spinal cord parenchyma. In parallel, tPA increased T cell activation and proliferation, as well as cytokine production by a protease-dependent mechanism and via plasmin generation. In addition, tPA directly raised the expression of MHC-II and the co-stimulatory molecules CD80 and CD86 at the surface of dendritic cells and macrophages by a direct action dependent of the activation of epidermal growth factor receptor. CONCLUSIONS: Our study provides new insights into the mechanisms responsible for the harmful functions of tPA in multiple sclerosis and its animal models: tPA promotes the proliferation and activation of both lymphoid and myeloid populations by distinct, though complementary, mechanisms.


Asunto(s)
Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/inducido químicamente , Activación de Linfocitos/efectos de los fármacos , Células Mieloides/efectos de los fármacos , Activador de Tejido Plasminógeno/toxicidad , Animales , Femenino , Humanos , Activación de Linfocitos/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Activador de Tejido Plasminógeno/deficiencia
7.
Brain ; 143(5): 1414-1430, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32282893

RESUMEN

Primary progressive multiple sclerosis is a poorly understood disease entity with no specific prognostic biomarkers and scarce therapeutic options. We aimed to identify disease activity biomarkers in multiple sclerosis by performing an RNA sequencing approach in peripheral blood mononuclear cells from a discovery cohort of 44 untreated patients with multiple sclerosis belonging to different clinical forms and activity phases of the disease, and 12 healthy control subjects. A validation cohort of 58 patients with multiple sclerosis and 26 healthy control subjects was included in the study to replicate the RNA sequencing findings. The RNA sequencing revealed an interleukin 1 beta (IL1B) signature in patients with primary progressive multiple sclerosis. Subsequent immunophenotyping pointed to blood monocytes as responsible for the IL1B signature observed in this group of patients. Functional experiments at baseline measuring apoptosis-associated speck-like protein containing a CARD (ASC) speck formation showed that the NOD-leucine rich repeat and pyrin containing protein 3 (NLRP3) inflammasome was overactive in monocytes from patients with primary progressive multiple sclerosis, and canonical NLRP3 inflammasome activation with a combination of ATP plus lipopolysaccharide was associated with increased IL1B production in this group of patients. Primary progressive multiple sclerosis patients with high IL1B gene expression levels in peripheral blood mononuclear cells progressed significantly faster compared to patients with low IL1B levels based on the time to reach an EDSS of 6.0 and the Multiple Sclerosis Severity Score. In agreement with peripheral blood findings, both NLRP3 and IL1B expression in brain tissue from patients with primary progressive multiple sclerosis was mainly restricted to cells of myeloid lineage. Treatment of mice with a specific NLRP3 inflammasome inhibitor attenuated established experimental autoimmune encephalomyelitis disease severity and improved CNS histopathology. NLRP3 inflammasome-specific inhibition was also effective in reducing axonal damage in a model of lipopolysaccharide-neuroinflammation using organotypic cerebellar cultures. Altogether, these results point to a role of IL1B and the NLRP3 inflammasome as prognostic biomarker and potential therapeutic target, respectively, in patients with primary progressive multiple sclerosis.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1beta/inmunología , Esclerosis Múltiple Crónica Progresiva/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Adulto , Animales , Biomarcadores/análisis , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Pronóstico
8.
Neurobiol Dis ; 140: 104869, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32278882

RESUMEN

Multiple Sclerosis (MS) is the second cause of paraplegia among young adults, after all types of CNS traumatic lesions. In its most frequent relapsing-remitting form, the severity of the disease course is very heterogeneous, and its reliable evaluation remains a key issue for clinicians. Myeloid-Derived sSuppressor Cells (MDSCs) are immature myeloid cells that suppress the inflammatory response, a phenomenon related to the resolution or recovery of the clinical symptoms associated with experimental autoimmune encephalomyelitis (EAE), the most common model for MS. Here, we establish the severity index as a new parameter for the clinical assessment in EAE. It is derived from the relationship between the maximal clinical score and the time elapsed since disease onset. Moreover, we relate this new index with several histopathological hallmarks in EAE and with the peripheral content of MDSCs. Based on this new parameter, we show that the splenic MDSC content is related to the evolution of the clinical course of EAE, ranging from mild to severe. Indeed, when the severity index indicates a severe disease course, EAE mice display more intense lymphocyte infiltration, demyelination and axonal damage. A direct correlation was drawn between the MDSC population in the peripheral immune system, and the preservation of myelin and axons, which was also correlated with T cell apoptosis within the CNS (being these cells the main target for MDSC suppression). The data presented clearly indicated that the severity index is a suitable tool to analyze disease severity in EAE. Moreover, our data suggest a clear relationship between circulating MDSC enrichment and disease outcome, opening new perspectives for the future targeting of this population as an indicator of MS severity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Esclerosis Múltiple/patología , Células Supresoras de Origen Mieloide/patología , Bazo/patología , Animales , Apoptosis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/patología , Células Mieloides/inmunología
9.
Neurobiol Dis ; 127: 13-31, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30798007

RESUMEN

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the human central nervous system (CNS), mainly affecting young adults. Among the immunomodulatory disease modifying treatments approved up to date to treat MS, IFN-ß remains to be one of the most widely prescribed for the Relapsing-Remitting (RR) variant of the disease, although its mechanism of action is still partially understood. RR-MS variant is characterized by phases with increasing neurological symptoms (relapses) followed by periods of total or partial recovery (remissions), which implies the existence of immunomodulatory agents to promote the relapsing-to-remitting transition. Among these agents, it has been described the immunosuppressive role of a heterogeneous population of immature myeloid cells, namely the myeloid-derived suppressor cells (MDSCs) during the clinical course of the experimental autoimmune encephalomyelitis (EAE), the most used MS model to study RRMS. However, it is still unknown how the current MS disease modifying treatments, e.g. IFN- ß, affects to MDSCs number or activity. Our present results show that a single injection of IFN-ß at the onset of the clinical course reduces the severity of the EAE, enhancing the presence of MDSCs within the smaller demyelinated areas. Moreover, the single dose of IFN-ß promotes MDSC immunosuppressive activity both in vivo and in vitro, augmenting T cell apoptosis. Finally, we show that IFN-ß preserves MDSC immaturity, preventing their differentiation to mature and less suppressive myeloid cell subsets. Taking together, all these data add new insights into the mechanism of IFN-ß treatment in EAE and point to MDSCs as a putative endogenous mediator of its beneficial role in this animal model of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Interferón beta/uso terapéutico , Células Supresoras de Origen Mieloide/efectos de los fármacos , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Inmunosupresores/farmacología , Interferón beta/farmacología , Ratones , Células Supresoras de Origen Mieloide/inmunología
10.
Glia ; 66(7): 1447-1463, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29484707

RESUMEN

The innate immune response is mediated by primary immune modulators such as cytokines and chemokines that together with immune cells and resident glia orchestrate CNS immunity and inflammation. Growing evidence supports that the endocannabinoid 2-arachidonoylglycerol (2-AG) exerts protective actions in CNS injury models. Here, we used the acute phase of Theiler's virus induced demyelination disease (TMEV-IDD) as a model of acute neuroinflammation to investigate whether 2-AG modifies the brain innate immune responses to TMEV and CNS leukocyte trafficking. 2-AG or the inhibition of its hydrolysis diminished the reactivity and number of microglia at the TMEV injection site reducing their morphological complexity and modulating them towards an anti-inflammatory state via CB2 receptors. Indeed, 2-AG dampened the infiltration of immune cells into the CNS and inhibited their egress from the spleen, resulting in long-term beneficial effects at the chronic phase of the disease. Intriguingly, it is not a generalized action over leukocytes since 2-AG increased the presence and suppressive potency of myeloid derived suppressor cells (MDSCs) in the brain resulting in higher apoptotic CD4+ T cells at the injection site. Together, these data suggest a robust modulatory effect in the peripheral and central immunity by 2-AG and highlight the interest of modulating endogenous cannabinoids to regulate CNS inflammatory conditions.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Infecciones por Cardiovirus/inmunología , Endocannabinoides/metabolismo , Glicéridos/metabolismo , Inflamación/inmunología , Microglía/inmunología , Theilovirus , Animales , Ácidos Araquidónicos/administración & dosificación , Encéfalo/inmunología , Encéfalo/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Infecciones por Cardiovirus/patología , Modelos Animales de Enfermedad , Endocannabinoides/administración & dosificación , Femenino , Glicéridos/administración & dosificación , Inmunidad Innata/inmunología , Inflamación/patología , Ratones , Microglía/patología , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/metabolismo , Receptor Cannabinoide CB2/antagonistas & inhibidores , Receptor Cannabinoide CB2/metabolismo
11.
J Neurosci ; 36(36): 9313-25, 2016 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-27605608

RESUMEN

UNLABELLED: Hyperglycemia is common in patients with acute stroke, even in those without preexisting diabetes, and denotes a bad outcome. However, the mechanisms underlying the detrimental effects of hyperglycemia are largely unclear. In a mouse model of ischemic stroke, we found that hyperglycemia increased the infarct volume and decreased the number of protective noninflammatory monocytes/macrophages in the ischemic brain. Ablation of peripheral monocytes blocked the detrimental effect of hyperglycemia, suggesting that monocytes are required. In hyperglycemic mice, α-dicarbonyl glucose metabolites, the precursors for advanced glycation end products, were significantly elevated in plasma and ischemic brain tissue. The receptor of advanced glycation end products, AGER (previously known as RAGE), interfered with polarization of macrophages to a noninflammatory phenotype. When Ager was deleted, hyperglycemia did not aggravate ischemic brain damage any longer. Independently of AGER, methylglyoxal reduced the release of endothelial CSF-1 (M-CSF), which stimulates polarization of macrophages to a noninflammatory phenotype in the microenvironment of the ischemic brain. In summary, our study identified α-dicarbonyls and AGER as mediators by which hyperglycemia lowers the number of protective noninflammatory macrophages and consequently increases ischemic brain damage. Modulating the metabolism of α-dicarbonyls or blocking AGER may improve the treatment of stroke patients with hyperglycemia. SIGNIFICANCE STATEMENT: Although glucose is the main energy substrate of the brain, hyperglycemia aggravates ischemic brain damage in acute stroke. So far, clinical trials have indicated that insulin treatment provides no solution to this common clinical problem. This study shows, in an experimental stroke model, that hyperglycemia interferes with the polarization of monocytes/macrophages to a protective cell type. Key players are α-dicarbonyls and the receptor for advanced glycation end products (AGER). Deletion of AGER normalized monocyte/macrophage polarization and reversed the detrimental effects of hyperglycemia, suggesting new avenues to treat stroke patients.


Asunto(s)
Polaridad Celular/fisiología , Hiperglucemia/etiología , Hiperglucemia/patología , Macrófagos/patología , Monocitos/patología , Accidente Cerebrovascular/complicaciones , Animales , Trasplante de Médula Ósea , Encéfalo/citología , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Receptor 1 de Quimiocinas CX3C , Polaridad Celular/genética , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Regulación de la Expresión Génica/genética , Hiperglucemia/cirugía , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Accidente Cerebrovascular/cirugía
12.
Biochim Biophys Acta ; 1862(3): 368-80, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26527182

RESUMEN

The knowledge of the immune system elements and their relationship with other tissues, organs and systems are key approximations for the resolution of many immune-related disorders. The control of the immune response and/or its modulation from the pro-inflammatory to the anti-inflammatory response is being deeply studied in the field. In the last years, the study of myeloid-derived suppressor cells (MDSCs), a group of immature myeloid cells with a high suppressive activity on T cells has been extensively addressed in cancer. In contrast, their role in neuroimmune diseases is far from being totally understood. In this review, we will summarize data about MDSCs coming from the study of neuroinflammatory diseases in general and their potential role in multiple sclerosis, in order to introduce the putative use of this extraordinary promising cell type for future cell-based therapies. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Inflamación/patología , Esclerosis Múltiple/patología , Células Supresoras de Origen Mieloide/patología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Humanos , Tolerancia Inmunológica , Inflamación/inmunología , Inflamación/terapia , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/trasplante , Neuroinmunomodulación , Linfocitos T/inmunología , Linfocitos T/patología
13.
Brain ; 139(Pt 9): 2406-19, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27435092

RESUMEN

Multiple sclerosis is among the most common causes of neurological disability in young adults. Here we provide the preclinical proof of concept of the benefit of a novel strategy of treatment for multiple sclerosis targeting neuroendothelial N-methyl-D-aspartate glutamate receptors. We designed a monoclonal antibody against N-methyl-D-aspartate receptors, which targets a regulatory site of the GluN1 subunit of N-methyl-D-aspartate receptor sensitive to the protease tissue plasminogen activator. This antibody reverted the effect of tissue plasminogen activator on N-methyl-D-aspartate receptor function without affecting basal N-methyl-D-aspartate receptor activity (n = 21, P < 0.01). This antibody bound N-methyl-D-aspartate receptors on the luminal surface of neurovascular endothelium in human tissues and in mouse, at the vicinity of tight junctions of the blood-spinal cord barrier. Noteworthy, it reduced human leucocyte transmigration in an in vitro model of the blood-brain barrier (n = 12, P < 0.05). When injected during the effector phase of MOG-induced experimental autoimmune encephalomyelitis (n = 24), it blocked the progression of neurological impairments, reducing cumulative clinical score (P < 0.001) and mean peak score (P < 0.001). This effect was observed in wild-type animals but not in tissue plasminogen activator knock-out animals (n = 10). This therapeutic effect was associated to a preservation of the blood-spinal cord barrier (n = 6, P < 0.001), leading to reduced leucocyte infiltration (n = 6, P < 0.001). Overall, this study unveils a critical function of endothelial N-methyl-D-aspartate receptor in multiple sclerosis, and highlights the therapeutic potential of strategies targeting the protease-regulated site of N-methyl-D-aspartate receptor.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/farmacología , Proteínas del Tejido Nervioso/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Activador de Tejido Plasminógeno/metabolismo , Animales , Células Endoteliales , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Neurobiol Dis ; 67: 149-64, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24709559

RESUMEN

Relapsing-remitting multiple sclerosis (RR-MS) is an inflammatory and demyelinating disease of the central nervous system (CNS). It is characterized by relapsing phases with ongoing neurological affectation that are followed by a remitting period in which inflammatory events are controlled and the patients partially recover. Experimental Autoimmune Encephalomyelitis (EAE) is the animal model most often used to study the inflammatory component of MS. Several cell types are involved in controlling the immune response in EAE and immature myeloid-derived suppressor cells (MDSCs) have emerged as important actors in the immunomodulation that occurs in EAE due to their ability to suppress inflammatory responses by inducing T cell apoptosis. In this study, we assessed whether MDSC differentiation may have consequences on the clinical course of EAE by treating mice around the peak of the clinical course EAE with the MDSC-differentiating agent Am80, an analogue of retinoid acid. Am80 administration abrogates the immunomodulation that occurs in EAE mice through different MDSC-related mechanisms: i) induction of MDSC apoptosis; ii) polarization of MDSCs to mature subsets of myeloid cells (dendritic cells/macrophages/neutrophils); and iii) altering their immunosuppressor phenotype. Consequently, T cell density increases and their viability is promoted, delaying the animal's recovery. Therefore, our data point to MDSC behaviour as a crucial factor in facilitating the transition from the relapsing to the remission phase in EAE, which should be considered for future immune-related therapies for MS.


Asunto(s)
Benzoatos/farmacología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Células Progenitoras Mieloides/inmunología , Receptores de Ácido Retinoico/agonistas , Tetrahidronaftalenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Neutrófilos/inmunología , Receptor alfa de Ácido Retinoico , Médula Espinal/metabolismo , Médula Espinal/patología , Linfocitos T/inmunología
15.
Nanoscale ; 16(15): 7515-7531, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38498071

RESUMEN

Despite the pharmacological arsenal approved for Multiple Sclerosis (MS), there are treatment-reluctant patients for whom cell therapy appears as the only therapeutic alternative. Myeloid-derived suppressor cells (MDSCs) are immature cells of the innate immunity able to control the immune response and to promote oligodendroglial differentiation in the MS animal model experimental autoimmune encephalomyelitis (EAE). However, when isolated and cultured for cell therapy purposes, MDSCs lose their beneficial immunomodulatory properties. To prevent this important drawback, culture devices need to be designed so that MDSCs maintain a state of immaturity and immunosuppressive function similar to that exerted in the donor organism. With this aim, we select graphene oxide (GO) as a promising candidate as it has been described as a biocompatible nanomaterial with the capacity to biologically modulate different cell types, yet its immunoactive potential has been poorly explored to date. In this work, we have fabricated GO films with two distintive redox and roughness properties and explore their impact in MDSC culture right after isolation. Our results show that MDSCs isolated from immune organs of EAE mice maintain an immature phenotype and highly immunosuppressive activity on T lymphocytes after being cultured on highly-reduced GO films (rGO200) compared to those grown on conventional glass coverslips. This immunomodulation effect is depleted when MDSCs are exposed to slightly rougher and more oxidized GO substrates (rGO90), in which cells experience a significant reduction in cell size associated with the activation of apoptosis. Taken together, the exposure of MDSCs to GO substrates with different redox state and roughness is presented as a good strategy to control MDSC activity in vitro. The versatility of GO nanomaterials in regards to the impact of their physico-chemical properties in immunomodulation opens the door to their selective therapeutic potential for pathologies where MDSCs need to be enhanced (MS) or inhibited (cancer).


Asunto(s)
Encefalomielitis Autoinmune Experimental , Grafito , Esclerosis Múltiple , Células Supresoras de Origen Mieloide , Humanos , Ratones , Animales , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Grafito/farmacología , Inmunosupresores , Ratones Endogámicos C57BL
16.
PLoS One ; 18(5): e0283469, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37205683

RESUMEN

COVID-19 pandemic has put the protocols and the capacity of our Hospitals to the test. The management of severe patients admitted to the Intensive Care Units has been a challenge for all health systems. To assist in this challenge, various models have been proposed to predict mortality and severity, however, there is no clear consensus for their use. In this work, we took advantage of data obtained from routine blood tests performed on all individuals on the first day of hospitalization. These data has been obtained by standardized cost-effective technique available in all the hospitals. We have analyzed the results of 1082 patients with COVID19 and using artificial intelligence we have generated a predictive model based on data from the first days of admission that predicts the risk of developing severe disease with an AUC = 0.78 and an F1-score = 0.69. Our results show the importance of immature granulocytes and their ratio with Lymphocytes in the disease and present an algorithm based on 5 parameters to identify a severe course. This work highlights the importance of studying routine analytical variables in the early stages of hospital admission and the benefits of applying AI to identify patients who may develop severe disease.


Asunto(s)
COVID-19 , Humanos , Inteligencia Artificial , Pandemias , Curva ROC , Hospitalización , Estudios Retrospectivos
17.
J Neurosci ; 31(42): 14899-909, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22016523

RESUMEN

Multiple sclerosis is a demyelinating disease that affects ≈ 2,000,000 people worldwide. In the advanced stages of the disease, endogenous oligodendrocyte precursors cannot colonize the lesions or differentiate into myelinating oligodendrocytes. During development, both FGF-2 and Anosmin-1 participate in oligodendrocyte precursor cell migration, acting via the FGF receptor 1 (FGFR1). Hence, we performed a histopathological and molecular analysis of these developmental modulators in postmortem tissue blocks from multiple sclerosis patients. Accordingly, we demonstrate that the distribution of FGF-2 and Anosmin-1 varies between the different types of multiple sclerosis lesions: FGF-2 is expressed only within active lesions and in the periplaque of chronic lesions, whereas Anosmin-1 is upregulated within chronic lesions and is totally absent in active lesions. We show that the endogenous oligodendrocyte precursor cells recruited toward chronic-active lesions express FGFR1, possibly in response to the FGF-2 produced by microglial cells in the periplaque. Also in human tissue, FGF-2 is upregulated in perivascular astrocytes in regions of the normal-appearing gray matter, where the integrity of the blood-brain barrier is compromised. In culture, FGF-2 and Anosmin-1 influence adult mouse oligodendrocyte precursor cell migration in the same manner as at embryonic stages, providing an explanation for the histopathological observations: FGF-2 attracts/enhances its migration, which is hindered by Anosmin-1. We propose that FGF-2 and Anosmin-1 are markers for the histopathological type and the level of inflammation of multiple sclerosis lesions, and that they may serve as novel pharmacogenetic targets to design future therapies that favor effective remyelination and protect the blood-brain barrier.


Asunto(s)
Corteza Cerebral/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Esclerosis Múltiple/clasificación , Esclerosis Múltiple/patología , Proteínas del Tejido Nervioso/metabolismo , Regulación hacia Arriba/fisiología , Adulto , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Antígenos/metabolismo , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Corteza Cerebral/patología , Cricetinae , Cricetulus , Inhibidores Enzimáticos/farmacología , Femenino , Gangliósidos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Fosfoproteínas/metabolismo , Cambios Post Mortem , Proteoglicanos/metabolismo , Pirroles/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transfección/métodos , Regulación hacia Arriba/efectos de los fármacos , Proteína de la Zonula Occludens-1
18.
Glia ; 60(6): 851-66, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22354480

RESUMEN

Oligodendrocyte precursor cells (OPCs) of the optic nerve are generated in the preoptic area, from where they migrate to colonize it entirely. Sonic hedgehog (Shh) induces the proliferation of these cells as well as influencing their migration, acting through its canonical receptor (Ptc-1). However, the multiligand receptor megalin (or LRP-2) is also involved in Shh-induced OPC proliferation and migration, and thus, we have evaluated the relevance of this interaction. During the stages at which Shh influences OPC development, we found megalin to be selectively expressed by optic nerve astrocytes, whereas Ptc-1 and Gli1 were found in OPCs. Indeed, this pattern of expression paralleled the rostral-caudal expression of the three Shh-related molecules during the time course of plp-dm20(+) -OPC colonization. The blockage of megalin partially abolished OPC chemoattraction and fully impaired Shh-induced proliferation. Using in vitro co-cultures of dissociated optic nerve cells, we demonstrated that Shh was internalized by astrocytes via megalin, and sufficient Shh was subsequently released to produce the biological effects on OPCs observed in the nerve. Together, these data indicate that at least part of the influence of Shh on OPCs is mediated by megalin during optic nerve development, and that astrocytes expressing megalin transiently capture Shh to present it to OPCs and/or to control the gradient of this molecule during development.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Hedgehog/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Oligodendroglía/fisiología , Animales , Anticuerpos/farmacología , Astrocitos/fisiología , Bromodesoxiuridina/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis/fisiología , Técnicas de Cocultivo/métodos , Cricetinae , Cricetulus , Citarabina/farmacología , Embrión de Mamíferos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/fisiología , Exocitosis/fisiología , Ojo/embriología , Ojo/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Gangliósidos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Hedgehog/genética , Inmunosupresores/farmacología , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/inmunología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Proteína Proteolipídica de la Mielina/metabolismo , Oligodendroglía/efectos de los fármacos , Nervio Óptico/citología , Nervio Óptico/embriología , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/metabolismo , Transfección , Vimentina/metabolismo , Proteína con Dedos de Zinc GLI1
19.
Biomedicines ; 10(2)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35203544

RESUMEN

Multiple sclerosis (MS) is a chronic, autoimmune, and neurodegenerative disease of the central nervous system (CNS) that affects myelin. The etiology of MS is unclear, although a variety of environmental and genetic factors are thought to increase the risk of developing the disease. Historically, T cells were considered to be the orchestrators of MS pathogenesis, but evidence has since accumulated implicating B lymphocytes and innate immune cells in the inflammation, demyelination, and axonal damage associated with MS disease progression. However, more recently the importance of the protective role of immunoregulatory cells in MS has become increasingly evident, such as that of myeloid-derived suppressor cells (MDSCs), regulatory T (Treg) and B (Breg) cells, or CD56bright natural killer cells. In this review, we will focus on how peripheral regulatory cells implicated in innate and adaptive immune responses are involved in the physiopathology of MS. Moreover, we will discuss how these cells are thought to act and contribute to MS histopathology, also addressing their promising role as promoters of successful remyelination within the CNS. Finally, we will analyze how understanding these protective mechanisms may be crucial in the search for potential therapies for MS.

20.
Front Cell Neurosci ; 15: 773696, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34916910

RESUMEN

The progression of neurodegenerative diseases is reciprocally associated with impairments in peripheral immune responses. We investigated different contexts of selective neurodegeneration to identify specific alterations of peripheral immune cells and, at the same time, discover potential biomarkers associated to this pathological condition. Consequently, a model of human cerebellar degeneration and ataxia -the Purkinje Cell Degeneration (PCD) mouse- has been employed, as it allows the study of different processes of selective neuronal death in the same animal, i.e., Purkinje cells in the cerebellum and mitral cells in the olfactory bulb. Infiltrated leukocytes were studied in both brain areas and compared with those from other standardized neuroinflammatory models obtained by administering either gamma radiation or lipopolysaccharide. Moreover, both myeloid and lymphoid splenic populations were analyzed by flow cytometry, focusing on markers of functional maturity and antigen presentation. The severity and type of neural damage and inflammation affected immune cell infiltration. Leukocytes were more numerous in the cerebellum of PCD mice, being located predominantly within those cerebellar layers mostly affected by neurodegeneration, in a completely different manner than the typical models of induced neuroinflammation. Furthermore, the milder degeneration of the olfactory bulb did not foster leukocyte attraction. Concerning the splenic analysis, in PCD mice we found: (1) a decreased percentage of several myeloid cell subsets, and (2) a reduced mean fluorescence intensity in those myeloid markers related to both antigen presentation and functional maturity. In conclusion, the selective degeneration of Purkinje cells triggers a specific effect on peripheral immune cells, fostering both attraction and functional changes. This fact endorses the employment of peripheral immune cell populations as concrete biomarkers for monitoring different neuronal death processes.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA