Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Glia ; 71(12): 2815-2831, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37610097

RESUMEN

Remyelination failure is considered a major obstacle in treating chronic-progressive multiple sclerosis (MS). Studies have shown blockage in the differentiation of resident oligodendrocyte progenitor cells (OPC) into myelin-forming cells, suggesting that pushing OPC into a differentiation program might be sufficient to overcome remyelination failure. Others stressed the need for a permissive environment to allow proper activation, migration, and differentiation of OPC. PD0325901, a MAPK/ERK inhibitor, was previously shown to induce OPC differentiation, non-specific immunosuppression, and a significant therapeutic effect in acute demyelinating MS models. We examined PD0325901 effects in the chronically inflamed central nervous system. Treatment with PD0325901 induced OPC differentiation into mature oligodendrocytes with high morphological complexity. However, treatment of Biozzi mice with chronic-progressive experimental autoimmune encephalomyelitis with PD0325901 showed no clinical improvement in comparison to the control group, no reduction in demyelination, nor induction of OPC migration into foci of demyelination. PD0325901 induced a direct general immunosuppressive effect on various cell populations, leading to a diminished phagocytic capability of microglia and less activation of lymph-node cells. It also significantly impaired the immune-modulatory functions of OPC. Our findings suggest OPC regenerative function depends on a permissive environment, which may include pro-regenerative inflammatory elements. Furthermore, they indicate that maintaining a delicate balance between the pro-myelinating and immune functions of OPC is of importance. Thus, the highly complex mission of creating a pro-regenerative environment depends upon an appropriate immune response controlled in time, place, and intensity. We suggest the need to employ a multi-systematic therapeutic approach, which cannot be achieved through a single molecule-based therapy.

2.
Glia ; 70(6): 1191-1209, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35266197

RESUMEN

Oligodendrocyte progenitor cells (OPCs) are responsible for remyelination in the central nervous system (CNS) in health and disease. For patients with multiple sclerosis (MS), remyelination is not always successful, and the mechanisms differentiating successful from failed remyelination are not well-known. Growing evidence suggests an immune role for OPCs, in addition to their regenerative role; however, it is not clear if this helps or hinders the regenerative process. We studied the effect of cerebrospinal fluid (CSF) from relapsing MS (rMS) and progressive MS (pMS) patients on primary OPC differentiation and immune gene expression and function. We observed that CSF from either rMS or pMS patients has a differential effect on the ability of mice OPCs to differentiate into mature oligodendrocytes and to express immune functions. CSF of pMS patients impaired differentiation into mature oligodendrocytes. In addition, it led to decreased major histocompatibility complex class (MHC)-II expression, tumor necrosis factor (TNF)-α secretion, nuclear factor kappa-B (NFκB) activation, and less activation and proliferation of T cells. Our findings suggest that OPCs are not only responsible for remyelination, but they may also play an active role as innate immune cells in the CNS.


Asunto(s)
Esclerosis Múltiple , Células Precursoras de Oligodendrocitos , Remielinización , Animales , Diferenciación Celular/fisiología , Humanos , Inmunidad , Ratones , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Remielinización/fisiología
3.
J Neuroinflammation ; 19(1): 5, 2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991645

RESUMEN

BACKGROUND: Neurodegeneration is considered the consequence of misfolded proteins' deposition. Little is known about external environmental effects on the neurodegenerative process. Infectious agent-derived pathogen-associated molecular patterns (PAMPs) activate microglia, key players in neurodegenerative diseases. We hypothesized that systemic microbial pathogens may accelerate neurodegeneration in Alzheimer's disease (AD) and that microglia play a central role in this process. METHODS: We examined the effect of an infectious environment and of microbial Toll-like receptor (TLR) agonists on cortical neuronal loss and on microglial phenotype in wild type versus 5xFAD transgenic mice, carrying mutated genes associated with familial AD. RESULTS: We examined the effect of a naturally bred environment on the neurodegenerative process. Earlier and accelerated cortical neuron loss occurred in 5xFAD mice housed in a natural ("dirty") environment than in a specific-pathogen-free (SPF) environment, without increasing the burden of Amyloid deposits and microgliosis. Neuronal loss occurred in a microglia-rich cortical region but not in microglia-poor CA regions of the hippocampus. Environmental exposure had no effect on cortical neuron density in wild-type mice. To model the neurodegenerative process caused by the natural infectious environment, we injected systemically the bacterial endotoxin lipopolysaccharide (LPS), a TLR4 agonist PAMP. LPS caused cortical neuronal death in 5xFAD, but not wt mice. We used the selective retinoic acid receptor α agonist Am580 to regulate microglial activation. In primary microglia isolated from 5xFAD mice, Am580 markedly attenuated TLR agonists-induced iNOS expression, without canceling their basic immune response. Intracerebroventricular delivery of Am580 in 5xFAD mice reduced significantly the fraction of (neurotoxic) iNOS + microglia and increased the fraction of (neuroprotective) TREM2 + microglia. Furthermore, intracerebroventricular delivery of Am580 prevented neurodegeneration induced by microbial TLR agonists. CONCLUSIONS: Exposure to systemic infections causes neurodegeneration in brain regions displaying amyloid pathology and high local microglia density. AD brains exhibit increased susceptibility to microbial PAMPs' neurotoxicity, which accelerates neuronal death. Microglial modulation protects the brain from microbial TLR agonist PAMP-induced neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Microglía/patología , Degeneración Nerviosa/patología , Neuronas/patología , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Lipopolisacáridos/farmacología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fagocitosis/efectos de los fármacos
4.
Brain Behav Immun ; 96: 40-53, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33989746

RESUMEN

Brain stimulation by electroconvulsive therapy is effective in neuropsychiatric disorders by unknown mechanisms. Microglial toxicity plays key role in neuropsychiatric, neuroinflammatory and degenerative diseases. We examined the mechanism by which electroconvulsive seizures (ECS) regulates microglial phenotype and response to stimuli. Microglial responses were examined by morphological analysis, Iba1 and cytokine expression. ECS did not affect resting microglial phenotype or morphology but regulated their activation by Lipopolysaccharide stimulation. Microglia were isolated after ECS or sham sessions in naïve mice for transcriptome analysis. RNA sequencing identified 141 differentially expressed genes. ECS modulated multiple immune-associated gene families and attenuated neurotoxicity-associated gene expression. Blood brain barrier was examined by injecting Biocytin-TMR tracer. There was no breakdown of the BBB, nor increase in gene-signature of peripheral monocytes, suggesting that ECS effect is mainly on resident microglia. Unbiased analysis of regulatory sequences identified the induction of microglial retinoic acid receptor α (RARα) gene expression and a putative common RARα-binding motif in multiple ECS-upregulated genes. The effects of AM580, a selective RARα agonist on microglial response to LPS was examined in vitro. AM580 prevented LPS-induced cytokine expression and reactive oxygen species production. Chronic murine experimental autoimmune encephalomyelitis (EAE) was utilized to confirm the role RARα signaling as mediator of ECS-induced transcriptional pathway in regulating microglial toxicity. Continuous intracerebroventricular delivery of AM580 attenuated effectively EAE severity. In conclusion, ECS regulates CNS innate immune system responses by activating microglial retinoic acid receptor α pathway, signifying a novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric and neurodegenerative diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Microglía , Receptor alfa de Ácido Retinoico , Animales , Terapia Electroconvulsiva , Lipopolisacáridos , Ratones , Transducción de Señal
5.
J Neuroinflammation ; 17(1): 55, 2020 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-32059733

RESUMEN

BACKGROUND: Accumulating data suggest a central role for brain microglia in mediating cortical neuronal death in Alzheimer's disease (AD), and for Toll-like receptor 2 (TLR2) in their toxic activation. Amyloid deposition in preclinical AD is associated with microglial activation but not directly with neurodegeneration. We examined in transgenic 5xFAD mice the hypothesis that systemic TLR2 agonists, derived from common infectious agents, may accelerate neurodegeneration in AD. METHODS: Microbial wall-derived TLR2 agonists zymosan and lipoteichoic acid were administered intraperitoneally or intracerebroventricularly to 7-month-old wild-type or 5xFAD mice. Immunofluorescent stainings were used to quantify cortical neurons and evaluate tissue reaction. Microglial activation was assessed using functional assays, RNA expression, and FACS analysis. RESULTS: Repeated low-dose systemic administration of zymosan or lipoteichoic acid killed cortical neurons in 5xFAD mice but not in wild-type mice. Direct CNS delivery of a selective TLR2 antagonist blocked the neurotoxicity of systemically administered zymosan, indicating that CNS TLR2 mediates this effect. Systemically administered zymosan crossed the disrupted blood-brain barrier in 5xFAD mice and entered brain parenchyma. By intracerebroventricular delivery, we found a dose- and exposure time-dependent acute neurotoxic effect of the microbial TLR2 agonist, killing cortical neurons. 5xFAD mice exhibited significantly increased vulnerability to TLR2 agonist-induced neuronal loss as compared to wild-type mice. Microbial TLR2-induced neurodegeneration was abolished by inhibiting microglia. The vulnerability of 5xFAD mice brains was mediated by an increase in number and neurotoxic phenotype of TLR2-expressing microglia. CONCLUSIONS: We suggest that repeated exposure to microbial TLR2 agonists may facilitate neurodegeneration in AD by their microglial-mediated toxicity to the hyper-vulnerable environment of the AD brain.


Asunto(s)
Enfermedad de Alzheimer/patología , Corteza Cerebral/efectos de los fármacos , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Receptor Toll-Like 2/agonistas , Animales , Corteza Cerebral/patología , Lipopolisacáridos/farmacología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Neuronas/patología , Ácidos Teicoicos/farmacología , Zimosan/farmacología
6.
Nanomedicine ; 10(6): 1353-63, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24704590

RESUMEN

Neurodegenerative diseases generate the accumulation of specific misfolded proteins, such as PrP(Sc) prions or A-beta in Alzheimer's diseases, and share common pathological features, like neuronal death and oxidative damage. To test whether reduced oxidation alters disease manifestation, we treated TgMHu2ME199K mice, modeling for genetic prion disease, with Nano-PSO, a nanodroplet formulation of pomegranate seed oil (PSO). PSO comprises large concentrations of a unique polyunsaturated fatty acid, Punicic acid, among the strongest natural antioxidants. Nano-PSO significantly delayed disease presentation when administered to asymptomatic TgMHu2ME199K mice and postponed disease aggravation in already sick mice. Analysis of brain samples revealed that Nano-PSO treatment did not decrease PrP(Sc) accumulation, but rather reduced lipid oxidation and neuronal loss, indicating a strong neuroprotective effect. We propose that Nano-PSO and alike formulations may be both beneficial and safe enough to be administered for long years to subjects at risk or to those already affected by neurodegenerative conditions. FROM THE CLINICAL EDITOR: This team of authors report that a nanoformulation of pomegranade seed oil, containing high levels of a strong antioxidant, can delay disease onset in a mouse model of genetic prion diseases, and the formulation also indicates a direct neuroprotective effect.


Asunto(s)
Síndrome de Creutzfeldt-Jakob/tratamiento farmacológico , Emulsiones/uso terapéutico , Lythraceae/química , Fármacos Neuroprotectores/uso terapéutico , Aceites de Plantas/uso terapéutico , Animales , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/metabolismo , Síndrome de Creutzfeldt-Jakob/patología , Emulsiones/química , Peroxidación de Lípido/efectos de los fármacos , Ratones , Fármacos Neuroprotectores/química , Oxidación-Reducción , Aceites de Plantas/química , Priones/metabolismo , Semillas/química
7.
Glia ; 61(2): 140-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23001547

RESUMEN

Fetal neural stem/precursor cells (NPCs) possess powerful immunomodulatory properties which enable them to protect the brain from immune-mediated injury. A major issue in developing neural stem/precursor cell (NPC) therapy for chronic neuroinflammatory disorders such as multiple sclerosis is whether cells maintain their immune-regulatory properties for prolonged periods of time. Therefore, we studied time-associated changes in NPC immunomodulatory properties. We examined whether intracerebrally-transplanted NPCs are able to inhibit early versus delayed induction of autoimmune brain inflammation and whether allogeneic NPC grafts continuously inhibit host rejection responses. In two experimental designs, intraventricular fetal NPC grafts attenuated clinically and pathologically brain inflammation during early EAE relapse but failed to inhibit the disease relapse if induced at a delayed time point. In correlation, long-term cultured neural precursors lost their capacity to inhibit immune cell proliferation in vitro. Loss of NPC immune functions was associated with transition into a quiescent undifferentiated state. Also, allogeneic fetal NPC grafts elicited a strong immune reaction of T cell and microglial infiltration and were rejected from the host brain. We conclude that long-term functional changes in transplanted neural precursor cells lead to loss of their therapeutic immune-regulatory properties, and render allogeneic grafts vulnerable to immunologic rejection. Thus, the immunomodulatory effects of neural precursor cell transplantation are limited in time.


Asunto(s)
Encefalitis/prevención & control , Encefalomielitis Autoinmune Experimental/cirugía , Células-Madre Neurales/inmunología , Síndromes de Neurotoxicidad/cirugía , Trasplante de Células Madre/métodos , Adrenérgicos/toxicidad , Animales , Proliferación Celular , Cuerpo Estriado/fisiopatología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Encefalitis/etiología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/patología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Proteínas Fluorescentes Verdes/genética , Interferón gamma/metabolismo , Linfocitos/fisiología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Síndromes de Neurotoxicidad/complicaciones , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/patología , Oxidopamina/toxicidad , Fragmentos de Péptidos/toxicidad , Embarazo , Factores de Tiempo
8.
Neurobiol Dis ; 49: 41-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22910454

RESUMEN

Fetal neural stem/precursor cells (NPCs) possess powerful neurotrophic properties by which they can facilitate self repair processes in the adult central nervous system. The therapeutic value of NPC therapy in neurodegenerative diseases is critically dependent on their long term survival and enduring functional properties. An important aspect of NPC neurotrophic properties is their ability to support their own survival independent of any exogenous growth factor. Here, we examined whether NPCs survive and maintain their properties for extended periods of time, or become dependent on environmental support. Two months following transplantation to naïve brains, large grafts were detected in the ventricles and hippocampus, but only little survival was evident in the striatum. To point at possible regional characteristics which underlie the differential survival of NPC grafts we performed several manipulations of the brain environment. Acute neurotoxic injury with 6-hydroxydopamine induced a 3-fold increase in striatal graft survival, associated with induction of nestin, CD31, ß1-integrin, GFAP and cycling cells. Disruption of the extracellular matrix structure of this reactive niche by continuous blockage of host striatum ß1-integrin caused 73% reduction in graft survival. In the hippocampus, NPC graft survival did not correspond to ß1-integrin and CD31 expression. This suggests that hippocampal environmental support to graft survival rely on different mechanisms than in the reactive striatum. In correlation with in vivo findings, long term cultured neural precursors exhibited an increase in apoptotic cells and dramatic decline in neurotrophic effects, as indicated by two in vitro functional assays. We conclude that long-term changes in transplanted NPC properties render them dependent on region specific environmental support. The major extracellular matrix protein ß1-integrin is essential for providing tissue support for graft survival in the striatum. The neurotrophic properties of transplanted neural stem cells are limited in time, representing a shortcoming which should be taken into consideration when developing clinical transplantation protocols for chronic neurological disorders.


Asunto(s)
Trasplante de Tejido Encefálico , Encéfalo/fisiología , Factores de Crecimiento Nervioso/metabolismo , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Animales , Encéfalo/cirugía , Células Cultivadas , Femenino , Supervivencia de Injerto/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrina beta1/metabolismo , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Oligodendroglía/fisiología , Factores de Tiempo
9.
Curr Neurol Neurosci Rep ; 13(11): 397, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24078453

RESUMEN

The strong rationale for cell-based therapy in multiple sclerosis is based on the ability of stem and precursor cells of neural and mesenchymal origin to attenuate neuroinflammation, to facilitate endogenous repair processes, and to participate directly in remyelination, if directed towards a myelin-forming fate. However, there are still major gaps in knowledge regarding induction of repair in chronic multiple sclerosis lesions, and whether transplanted cells can overcome the multiple environmental inhibitory factors which underlie the failure of endogenous repair. Major challenges in clinical translation include the determination of the optimal cellular platform, the route of cell delivery, and candidate patients for treatment.


Asunto(s)
Esclerosis Múltiple/cirugía , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Humanos , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/inmunología , Células-Madre Neurales/fisiología , Plasticidad Neuronal/fisiología , Trasplante de Células Madre/tendencias
10.
Sci Rep ; 13(1): 16513, 2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37783693

RESUMEN

The impact of high-intensity interval training (HIIT) on the central nervous system (CNS) in autoimmune neuroinflammation is not known. The aim of this study was to determine the direct effects of HIIT on the CNS and development of experimental autoimmune encephalomyelitis (EAE). Healthy mice were subjected to HIIT by treadmill running and the proteolipid protein (PLP) transfer EAE model was utilized. To examine neuroprotection, PLP-reactive lymph-node cells (LNCs) were transferred to HIIT and sedentary (SED) mice. To examine immunomodulation, PLP-reactive LNCs from HIIT and SED donor mice were transferred to naïve recipients and analyzed in vitro. HIIT in recipient mice did not affect the development of EAE following exposure to PLP-reactive LNCs. HIIT mice exhibited enhanced migration of systemic autoimmune cells into the CNS and increased demyelination. In contrast, EAE severity in recipient mice injected with PLP-reactive LNCs from HIIT donor mice was significantly diminished. The latter positive effect was associated with decreased migration of autoimmune cells into the CNS and inhibition of very late antigen (VLA)-4 expression in LNCs. Thus, the beneficial effect of HIIT on EAE development is attributed solely to systemic immunomodulatory effects, likely because of systemic inhibition of autoreactive cell migration and reduced VLA-4 integrin expression.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Encefalomielitis , Entrenamiento de Intervalos de Alta Intensidad , Ratones , Animales , Sistema Nervioso Central/metabolismo , Inmunomodulación , Proteína Proteolipídica de la Mielina
11.
Mol Neurodegener ; 17(1): 53, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35986296

RESUMEN

BACKGROUND: The Amyloid theory of Alzheimer's disease (AD) suggests that the deposition of Amyloid ß (Aß) in the brain triggers a chain of events, involving the deposition of phosphorylated Tau and other misfolded proteins, leading to neurodegeneration via neuroinflammation, oxidative stress, and neurovascular factors. The infectious theory linked various infectious agents with the development of AD, raising the possibility that they serve as etiological causes of the disease. Are these theories mutually exclusive, or do they coincide? MAIN BODY: In this review, we will discuss how the two theories converge. We present a model by which (1) the systemic infectious burden accelerates the development of AD brain pathology via bacterial Amyloids and other pathogen-associated molecular patterns (PAMPs), and (2) the developing AD brain pathology increases its susceptibility to the neurotoxicity of infectious agents -derived PAMPs, which drive neurodegeneration via activated microglia. CONCLUSIONS: The reciprocal effects of amyloid deposition and systemic infectious burden may lead to a vicious cycle fueling Alzheimer's disease pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Humanos , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo
12.
Cells ; 11(5)2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35269501

RESUMEN

The failure of brain microglia to clear excess amyloid ß (Aß) is considered a leading cause of the progression of Alzheimer's disease pathology. Resident brain neural precursor cells (NPCs) possess immune-modulatory and neuro-protective properties, which are thought to maintain brain homeostasis. We have recently showed that resident mouse brain NPCs exhibit an acquired decline in their trophic properties in the Alzheimer's disease brain environment. Therefore, we hypothesized that functional NPCs may support microglial phagocytic activity, and that NPCs derived from the adult AD mouse brain may fail to support the clearance of Aß by microglia. We first identified in the AD brain, in vivo and ex vivo, a subpopulation of microglia that express high Aß phagocytic activity. Time-lapse microscopy showed that co-culturing newborn NPCs with microglia induced a significant increase in the fraction of microglia with high Aß phagocytic activity. Freshly isolated NPCs from adult wild type, but not AD, mouse brain, induced an increase in the fraction of microglia with high Aß phagocytic activity. Finally, we showed that NPCs also possess the ability to promote Aß degradation within the microglia with high Aß phagocytic activity. Thus, resident brain NPCs support microglial function to clear Aß, but NPCs derived from the AD environment fail to do so. We suggest that the failure of AD brain NPCs to support Aß clearance from the brain by microglia may accelerate disease pathology.


Asunto(s)
Enfermedad de Alzheimer , Células-Madre Neurales , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Ratones , Ratones Transgénicos , Microglía/metabolismo , Células-Madre Neurales/metabolismo
13.
Ann Clin Transl Neurol ; 9(11): 1792-1806, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36217574

RESUMEN

BACKGROUND: The mechanisms by which exercise training (ET) elicits beneficial effects on the systemic immune system and the central nervous system (CNS) in autoimmune neuroinflammation are not fully understood. OBJECTIVES: To investigate (1) the systemic effects of high-intensity continuous training (HICT) on the migratory potential of autoimmune cells; (2) the direct effects of HICT on blood-brain-barrier (BBB) properties. METHODS: Healthy mice were subjected to high-intensity continuous training (HICT) by treadmill running. The proteolipid protein (PLP) transfer EAE model was utilized to examine the immunomodulatory effects of training, where PLP-reactive lymph-node cells (LNCs) from HICT and sedentary donor mice were analyzed in vitro and transferred to naïve recipients that developed EAE. To examine neuroprotection, encephalitogenic LNCs from donor mice were transferred into HICT or sedentary recipient mice and the BBB was analyzed. RESULTS: Transfer of PLP-reactive LNCs obtained from HICT donor mice attenuated EAE severity and inflammation in recipient mice. HICT markedly inhibited very late antigen (VLA)-4 and lymphocyte function-associated antigen (LFA)-1 expression in LNCs. Transfer of encephalitogenic LNCs into HICT recipients resulted in milder EAE and attenuated CNS inflammation. HICT reduced BBB permeability and the expression of intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 in CNS blood vessels. INTERPRETATION: HICT attenuates EAE development by both immunomodulatory and neuroprotective effects. The reduction in destructive CNS inflammation in EAE is attributed to systemic inhibition of autoreactive cell migratory potential, as well as reduction in BBB permeability, which are associated with reduced VLA-4/VCAM-1 and LFA-1/ICAM-1 interactions.


Asunto(s)
Encefalitis , Encefalomielitis Autoinmune Experimental , Encefalomielitis , Animales , Ratones , Encefalomielitis Autoinmune Experimental/terapia , Encéfalo/metabolismo , Barrera Hematoencefálica , Encefalitis/metabolismo , Inflamación/metabolismo
14.
Stem Cell Res ; 56: 102559, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34634761

RESUMEN

Neural stem/precursor cells (NPC) exhibit powerful immune-modulatory properties. Attenuation of neuroinflammation by intra-cerebroventricular transplantation of NPC, protects from immune-mediated demyelination and axonal injury. The immune modulatory properties of NPC are mediated by a non-species-specific, multiple bystander effect, mediated by both direct cell-cell contact, and by soluble factor(s). CD200 is a cell-surface molecule, with important roles in regulating diverse immune responses, and shown also to limit neuroinflammatory processes. We hypothesized that CD200 may play a role in mediating immune-modulatory effects of NPC. We used wild type and CD200-deficient NPC to examine the role of CD200 in mediating two vital aspects of NPC -immune modulatory properties: (1) Attenuation of autoimmune neuroinflammation; and (2) Suppression of immune rejection response towards transplanted allogeneic NPC from the host CNS. We found that CD200 is dispensable for attenuating acute experimental autoimmune neuroinflammation, but is required for protecting transplanted allogeneic NPC from immune rejection by the host tissue. CD200 deficient NPC showed similar growth, differentiation and survival properties as wild type NPC. CD200-deficient NPC attenuated efficiently T cell activation and proliferation, but exhibited reduced ability to inhibit macrophages. We conclude that CD200 plays a partial role in mediating the immune-modulatory properties of NPC. The differential effect on T cells versus macrophages may underlie the observed discrepancy in their function in vivo.


Asunto(s)
Células-Madre Neurales , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Diferenciación Celular , Inmunidad , Macrófagos , Ratones
15.
J Neuroimmunol ; 356: 577582, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33910137

RESUMEN

We explored whether experimental autoimmune encephalomyelitis (EAE) in Biozzi mice recapitulates temporal dynamics of tissue injury, immune-pathogenesis and CNS compartmentalization occurring in progressive multiple sclerosis (MS). Chronic EAE exhibited relapsing and progressing disease, partial closure of BBB, reduced tissue inflammatory activity, and development of meningeal ectopic lymphoid tissue, directly opposing (potentially driving) spinal subpial demyelinated plaques. A T cell predominant disease during relapses transformed into a B cell predominant disease in late chronic EAE, with high serum anti-MOG reactivity. Thus, late chronic Biozzi EAE recapitulates essential features of progressive MS, and is suitable for developing disease modifying and regenerative therapies.


Asunto(s)
Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple Crónica Progresiva/inmunología , Médula Espinal/inmunología , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Adyuvante de Freund/toxicidad , Ratones , Ratones Biozzi , Esclerosis Múltiple Crónica Progresiva/inducido químicamente , Esclerosis Múltiple Crónica Progresiva/patología , Médula Espinal/patología
16.
Front Cell Neurosci ; 15: 640724, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708074

RESUMEN

Background: Exercise training induces beneficial effects on neurodegenerative diseases, and specifically on multiple sclerosis (MS) and it's model experimental autoimmune encephalomyelitis (EAE). However, it is unclear whether exercise training exerts direct protective effects on the central nervous system (CNS), nor are the mechanisms of neuroprotection fully understood. In this study, we investigated the direct neuroprotective effects of high-intensity continuous training (HICT) against the development of autoimmune neuroinflammation and the role of resident microglia. Methods: We used the transfer EAE model to examine the direct effects of training on the CNS. Healthy mice performed HICT by treadmill running, followed by injection of encephalitogenic proteolipid (PLP)-reactive T-cells to induce EAE. EAE severity was assessed clinically and pathologically. Brain microglia from sedentary (SED) and HICT healthy mice, as well as 5-days post EAE induction (before the onset of disease), were analyzed ex vivo for reactive oxygen species (ROS) and nitric oxide (NO) formation, mRNA expression of M1/M2 markers and neurotrophic factors, and secretion of cytokines and chemokines. Results: Transfer of encephalitogenic T-cells into HICT mice resulted in milder EAE, compared to sedentary mice, as indicated by reduced clinical severity, attenuated T-cell, and neurotoxic macrophage/microglial infiltration, and reduced loss of myelin and axons. In healthy mice, HICT reduced the number of resident microglia without affecting their profile. Isolated microglia from HICT mice after transfer of encephalitogenic T-cells exhibited reduced ROS formation and released less IL-6 and monocyte chemoattractant protein (MCP) in response to PLP-stimulation. Conclusions: These findings point to the critical role of training intensity in neuroprotection. HICT protects the CNS against autoimmune neuroinflammation by reducing microglial-derived ROS formation, neurotoxicity, and pro-inflammatory responses involved in the propagation of autoimmune neuroinflammation.

17.
Ann Clin Transl Neurol ; 8(1): 190-200, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33285042

RESUMEN

BACKGROUND: Studies have reported beneficial effects of exercise training on autoimmunity, and specifically on multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). However, it is unknown whether different training paradigms affect disease course via shared or separate mechanisms. OBJECTIVE: To compare the effects and mechanism of immune modulation of high intensity continuous training (HICT) versus high intensity interval training (HIIT) on systemic autoimmunity in EAE. METHODS: We used the proteolipid protein (PLP)-induced transfer EAE model to examine training effects on the systemic autoimmune response. Healthy mice performed HICT or HIIT by running on a treadmill. Lymph-node (LN)-T cells from PLP-immunized trained- versus sedentary donor mice were transferred to naïve recipients and EAE clinical and pathological severity were assessed. LN cells derived from donor trained and sedentary PLP-immunized mice were analyzed in vitro for T-cell activation and proliferation, immune cell profiling, and cytokine mRNA levels and cytokine secretion measurements. RESULTS: Both HICT and HIIT attenuated the encephalitogenicity of PLP-reactive T cells, as indicated by reduced EAE clinical severity and inflammation and tissue pathology in the central nervous system, following their transfer into recipient mice. HICT caused a marked inhibition of PLP-induced T-cell proliferation without affecting the T-cell profile. In contrast, HIIT did not alter T-cell proliferation, but rather inhibited polarization of T cells into T-helper 1 and T-helper 17 autoreactive populations. INTERPRETATION: HICT and HIIT attenuate systemic autoimmunity and T cell encephalitogenicity by distinct immunomodulatory mechanisms.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Condicionamiento Físico Animal/métodos , Condicionamiento Físico Animal/fisiología , Animales , Femenino , Ganglios Linfáticos/inmunología , Ratones , Médula Espinal/inmunología , Médula Espinal/patología
18.
J Neurosci Res ; 88(5): 936-44, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19885865

RESUMEN

Recent progress in cell therapy research for brain diseases has raised the need for non-invasive monitoring of transplanted cells. For therapeutic application in multiple sclerosis, transplanted cells need to be tracked both spatially and temporally, in order to assess their migration and survival in the host tissue. Magnetic resonance imaging (MRI) of superparamagnetic iron oxide-(SPIO)-labeled cells has been widely used for high resolution monitoring of the biodistribution of cells after transplantation into the central nervous system (CNS). Here we labeled mouse glial-committed neural precursor cells (NPCs) with the clinically approved SPIO contrast agent ferumoxides and examined their survival and differentiation in vitro, as well as their functional response to environmental signals present within the inflamed brain of experimental autoimmune encephalomyelitis (EAE) mice in vivo. We show that ferumoxides labeling does not affect NPC survival and pluripotency in vitro. Following intracerebroventricular (ICV) transplantation in EAE mice, ferumoxides-labeled NPCs responded to inflammatory cues in a similar fashion as unlabeled cells. Ferumoxides-labeled NPCs migrated over comparable distances in white matter tracts and differentiated equally into the glial lineages. Furthermore, ferumoxides-labeled NPCs inhibited lymph node cell proliferation in vitro, similarly to non-labeled cells, suggesting a preserved immunomodulatory function. These results demonstrate that ferumoxides-based MRI cell tracking is well suited for non-invasive monitoring of NPC transplantation.


Asunto(s)
Óxido Ferrosoférrico/metabolismo , Supervivencia de Injerto/fisiología , Imagen por Resonancia Magnética/métodos , Coloración y Etiquetado/métodos , Trasplante de Células Madre/métodos , Células Madre/fisiología , Animales , Animales Recién Nacidos , Trasplante de Tejido Encefálico/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Medios de Contraste/metabolismo , Medios de Contraste/farmacología , Dextranos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/terapia , Óxido Ferrosoférrico/farmacología , Inmunomodulación/fisiología , Inyecciones Intraventriculares , Activación de Linfocitos/fisiología , Nanopartículas de Magnetita , Ratones , Ratones Endogámicos C57BL , Neuroglía/citología , Neuroglía/fisiología , Esferoides Celulares , Células Madre/citología
19.
JCI Insight ; 5(17)2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32780728

RESUMEN

Electroconvulsive therapy is highly effective in resistant depression by unknown mechanisms. Microglial toxicity was suggested to mediate depression and plays key roles in neuroinflammatory and degenerative diseases, where there is critical shortage in therapies. We examined the effects of electroconvulsive seizures (ECS) on chronic neuroinflammation and microglial neurotoxicity. Electric brain stimulation inducing full tonic-clonic seizures during chronic relapsing-progressive experimental autoimmune encephalomyelitis (EAE) reduced spinal immune cell infiltration, reduced myelin and axonal loss, and prevented clinical deterioration. Using the transfer EAE model, we examined the effect of ECS on systemic immune response in donor mice versus ECS effect on CNS innate immune activity in recipient mice. ECS did not affect encephalitogenicity of systemic T cells, but it targeted the CNS directly to inhibit T cell-induced neuroinflammation. In vivo and ex vivo assays indicated that ECS suppressed microglial neurotoxicity by reducing inducible NOS expression, nitric oxide, and reactive oxygen species (ROS) production, and by reducing CNS oxidative stress. Microglia from ECS-treated EAE mice expressed less T cell stimulatory and chemoattractant factors. Our findings indicate that electroconvulsive therapy targets the CNS innate immune system to reduce neuroinflammation by attenuating microglial neurotoxicity. These findings signify a potentially novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric, and neurodegenerative diseases.


Asunto(s)
Terapia Electroconvulsiva/métodos , Encefalomielitis Autoinmune Experimental/terapia , Animales , Axones/metabolismo , Movimiento Celular , Células Cultivadas , Citocinas/metabolismo , Femenino , Ratones , Microglía/metabolismo , Vaina de Mielina/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Linfocitos T/inmunología , Linfocitos T/fisiología
20.
Mol Neurobiol ; 57(2): 1021-1034, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31656989

RESUMEN

Neuroglial precursor cells (NPC) possess immune-modulatory properties by which they prevent immune-mediated injury in experimental autoimmune encephalomyelitis (EAE). It is unclear whether cell transplantation in a clinical-relevant setup induces ongoing therapeutic effects in a chronic-active model of progressive multiple sclerosis (MS). We examined whether human embryonic stem cell (hESC)-derived NPCs inhibit progressive EAE in Biozzi AB/H mice, manifesting with chronic-active neuroinflammation and demyelinated plaques. hESC-derived NPCs were propagated for 6-8 weeks as spheres enriched for Olig2+ cells to switch from neuronal to glial commitment and to enrich for oligodendrocyte progenitor cells. NPC were transplanted intracerebroventricularly at 30 days post-EAE induction, after the acute relapse. We evaluated effects of cell transplantation on clinical parameters, neuroinflammation, myelination, and axonal loss. Transplanted animals exhibited a significantly milder disease, reduced neuroinflammation, reduced demyelination, and reduced axonal loss as compared to control EAE mice. Toluidine-blue semi-thin staining showed a bystander neuroprotective effect of human precursor cells preventing the loss of myelinated fibers in superficial layer of the cervical dorsal funiculus. Human Olig2+ cells were detected along spinal cord meninges after 65 days of follow-up. In co-cultures in vitro, Olig2+ human precursors inhibited Concanavalin A-induced murine T cell activation and proliferation. To conclude, glial-committed human NPC induce ongoing immune-regulatory and neuroprotective effects, following transplantation into mice with a clinical-relevant model of chronic-active MS and during established disease, entering the chronic phase. These properties highlight the therapeutic potential of human NPC transplantation in chronic MS and their delivery via the cerebrospinal fluid.


Asunto(s)
Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Células-Madre Neurales/trasplante , Células Precursoras de Oligodendrocitos/citología , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ratones , Vaina de Mielina/inmunología , Neuronas/citología , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Trasplante de Células Madre/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA