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

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nat Immunol ; 24(6): 925-940, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37188941

RESUMEN

Aging accounts for increased risk and dismal outcome of ischemic stroke. Here, we investigated the impact of age-related changes in the immune system on stroke. Upon experimental stroke, compared with young mice, aged mice had increased neutrophil clogging of the ischemic brain microcirculation, leading to worse no-reflow and outcomes. Aged mice showed an enhanced granulopoietic response to stroke that led to the accumulation of CD101+CD62Llo mature and CD177hiCD101loCD62Llo and CD177loCD101loCD62Lhi immature atypical neutrophils in the blood, endowed with increased oxidative stress, phagocytosis and procoagulant features. Production of CXCL3 by CD62Llo neutrophils of the aged had a key role in the development and pathogenicity of aging-associated neutrophils. Hematopoietic stem cell rejuvenation reverted aging-associated neutropoiesis and improved stroke outcome. In elderly patients with ischemic stroke, single-cell proteome profile of blood leukocytes identified CD62Llo neutrophil subsets associated with worse reperfusion and outcome. Our results unveil how stroke in aging leads to a dysregulated emergency granulopoiesis impacting neurological outcome.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratones , Animales , Neutrófilos , Leucocitos , Accidente Cerebrovascular/patología , Envejecimiento , Accidente Cerebrovascular Isquémico/patología
2.
Neurol Sci ; 45(3): 849-859, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38169013

RESUMEN

INTRODUCTION: Guillain-Barré syndrome associated with Coronavirus-2-related severe acute respiratory syndrome (COV-GBS) occurs as para- or post-infectious forms, depending on the timing of disease onset. In these two forms, we aimed to compare the cerebrospinal fluid (CSF) and serum proinflammatory cytokine profiles to evaluate differences that could possibly have co-pathogenic relevance. MATERIALS AND METHODS: We studied a retrospective cohort of 26 patients with either post-COV-GBS (n = 15), with disease onset occurring > 7 days after SARS-CoV-2 infection, or para-COV-GBS (n = 11), with disease onset 7 days or less. TNF-α, IL-6, and IL-8 were measured in the serum with SimplePlex™ Ella™ immunoassay. In addition to the para-/post-COV-GBS patients, serum levels of these cytokines were determined in those with non-COVID-associated-GBS (NC-GBS; n = 43), paucisymptomatic SARS-CoV-2 infection without GBS (COVID, n = 20), and in healthy volunteers (HV; n = 12). CSF cytokine levels were measured in patients with para-/post-COV-GBS, in those with NC-GBS (n = 29), or with Alzheimer's disease (AD; n = 24). RESULTS: Serum/CSF cytokine levels did not differ in para- vs post-COV-GBS. We found that SARS-CoV-2 infection raises the serum levels of TNF-α, IL-6, and IL-8, as well as an increase of IL-6 (in serum and CSF) and IL-8 (in CSF) in either NC-GBS or COV-GBS than controls. CSF and serum cytokine levels resulted independent one with another. CONCLUSIONS: The change of cytokines linked to SARS-CoV-2 in COV-GBS appears to be driven by viral infection, although it has unique characteristics in GBS as such and does not account for cases with para- or post-infectious onset.


Asunto(s)
COVID-19 , Síndrome de Guillain-Barré , Humanos , COVID-19/complicaciones , SARS-CoV-2 , Síndrome de Guillain-Barré/complicaciones , Citocinas , Interleucina-6/líquido cefalorraquídeo , Factor de Necrosis Tumoral alfa , Estudios Retrospectivos , Interleucina-8
3.
Int J Mol Sci ; 23(8)2022 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-35457169

RESUMEN

Myeloid cells, including parenchymal microglia, perivascular and meningeal macrophages, and dendritic cells (DCs), are present in the central nervous system (CNS) and establish an intricate relationship with other cells, playing a crucial role both in health and in neurological diseases. In this context, DCs are critical to orchestrating the immune response linking the innate and adaptive immune systems. Under steady-state conditions, DCs patrol the CNS, sampling their local environment and acting as sentinels. During neuroinflammation, the resulting activation of DCs is a critical step that drives the inflammatory response or the resolution of inflammation with the participation of different cell types of the immune system (macrophages, mast cells, T and B lymphocytes), resident cells of the CNS and soluble factors. Although the importance of DCs is clearly recognized, their exact function in CNS disease is still debated. In this review, we will discuss modern concepts of DC biology in steady-state and during autoimmune neuroinflammation. Here, we will also address some key aspects involving DCs in CNS patrolling, highlighting the neuroprotective nature of DCs and emphasizing their therapeutic potential for the treatment of neurological conditions. Recently, inhibition of the NAD+-dependent deac(et)ylase sirtuin 6 was demonstrated to delay the onset of experimental autoimmune encephalomyelitis, by dampening DC trafficking towards inflamed LNs. Thus, a special focus will be dedicated to sirtuins' role in DCs functions.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Sirtuinas , Animales , Sistema Nervioso Central/metabolismo , Células Dendríticas , Encefalomielitis Autoinmune Experimental/metabolismo , Esclerosis Múltiple/metabolismo , Sirtuinas/metabolismo
4.
Glia ; 69(5): 1204-1215, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33381863

RESUMEN

Transplanted mesenchymal stromal/stem cells (MSC) ameliorate the clinical course of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), reducing inflammation and demyelination. These effects are mediated by instructive cross-talk between MSC and immune and neural cells. Astroglial reaction to injury is a prominent feature of both EAE and MS. Astrocytes constitute a relevant target to control disease onset and progression and, based on their potential to acquire stem cell properties in situ, to foster recovery in the post-acute phase of pathology. We have assessed how MSC impact astrocytes in vitro and ex vivo in EAE. Expression of astroglial factors implicated in EAE pathogenesis was quantified by real-time PCR in astrocytes co-cultured with MSC or isolated from EAE cerebral cortex; astrocyte morphology and expression of activation markers were analyzed by confocal microscopy. The acquisition of neural stem cell properties by astrocytes was evaluated by neurosphere assay. Our study shows that MSC prevented astrogliosis, reduced mRNA expression of inflammatory cytokines that sustain immune cell infiltration in EAE, as well as protein expression of endothelin-1, an astrocyte-derived factor that inhibits remyelination and contributes to neurodegeneration and disease progression in MS. Moreover, our data reveal that MSC promoted the acquisition of progenitor traits by astrocytes. These data indicate that MSC attenuate detrimental features of reactive astroglia and, based on the reacquisition of stem cell properties, also suggest that astrocytes may be empowered in their protective and reparative actions by MSC.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Células Madre Mesenquimatosas , Esclerosis Múltiple , Animales , Astrocitos , Ratones , Ratones Endogámicos C57BL , Fenotipo
5.
J Allergy Clin Immunol ; 139(5): 1667-1676, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27670240

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) display a therapeutic plasticity because of their ability to modulate immunity, foster tissue repair, and differentiate into mesodermal cells. IFN-γ has been described to differently affect human mesenchymal stem cell (hMSC) and mouse mesenchymal stem cell (mMSC) immunomodulation and differentiation, depending on the inflammatory milieu. OBJECTIVE: We aimed at dissecting the relevant intracellular pathways through which IFN-γ affects MSC plasticity and the consequence of their manipulation on MSC functions. METHODS: Modification of relevant IFN-γ-dependent pathways in mMSCs was carried out in vitro through gene silencing or chemical inhibition of key components. Functional outcomes were assessed by means of Western blotting, real-time PCR, differentiation, and proliferation assays on MSCs. The effect on T cells was addressed by T-cell proliferation assays; the effect of mammalian target of rapamycin (mTOR) manipulation in MSCs was studied in vivo in a mouse model of delayed-type hypersensitivity assay. To address whether similar mechanisms are involved also in hMSCs on IFN-γ stimulation, the effect of chemical inhibition on the same intracellular pathways was assessed by means of Western blotting, and the final outcome on immunomodulatory properties was evaluated based on real-time PCR and T-cell proliferation. RESULTS: We revealed that in mMSCs IFN-γ-induced immunoregulation is mediated by early phosphorylation of signal transducer and activator of transcription (STAT) 1 and STAT3, which is significantly enhanced by an extracellular signal-regulated kinase 1/2-dependent mTOR inhibition, thereby promoting pSTAT1 nuclear translocation. Accordingly, after intracellular mTOR inhibition, MSCs augmented their ability to inhibit T-cell proliferation and control delayed-type hypersensitivity in vivo. Similarly, on mTOR blockade, hMSCs also enhanced their immunoregulatory features. A sustained exposure to IFN-γ led to inhibition of STAT3 activity, which in mMSCs resulted in an impaired proliferation and differentiation. CONCLUSION: These results provide new insights about MSC intracellular pathways affected by IFN-γ, demonstrating that pharmacologic or genetic manipulation of MSCs can enhance their immunomodulatory functions, which could be translated into novel therapeutic approaches.


Asunto(s)
Interferón gamma/farmacología , Células Madre Mesenquimatosas/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/inmunología , Serina-Treonina Quinasas TOR/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Hipersensibilidad Tardía/inmunología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Transducción de Señal/efectos de los fármacos
6.
Front Immunol ; 15: 1393842, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39136008

RESUMEN

Chondroitin sulfate proteoglycans (CSPGs) are fundamental components of the extracellular matrix in the central nervous system (CNS). Among these, the Nerve-Glial antigen 2 (NG2) stands out as a transmembrane CSPG exclusively expressed in a different population of cells collectively termed NG2-expressing cells. These enigmatic cells, found throughout the developing and adult CNS, have been indicated with various names, including NG2 progenitor cells, polydendrocytes, synantocytes, NG2 cells, and NG2-Glia, but are more commonly referred to as oligodendrocyte progenitor cells. Characterized by high proliferation rates and unique morphology, NG2-expressing cells stand apart from neurons, astrocytes, and oligodendrocytes. Intriguingly, some NG2-expressing cells form functional glutamatergic synapses with neurons, challenging the long-held belief that only neurons possess the intricate machinery required for neurotransmission. In the CNS, the complexity surrounding NG2-expressing cells extends to their classification. Additionally, NG2 expression has been documented in pericytes and immune cells, suggesting a role in regulating brain innate immunity and neuro-immune crosstalk in homeostasis. Ongoing debates revolve around their heterogeneity, potential as progenitors for various cell types, responses to neuroinflammation, and the role of NG2. Therefore, this review aims to shed light on the enigma of NG2-expressing cells by delving into their structure, functions, and signaling pathways. We will critically evaluate the literature on NG2 expression across the CNS, and address the contentious issues surrounding their classification and roles in neuroinflammation and neurodegeneration. By unraveling the intricacies of NG2-expressing cells, we hope to pave the way for a more comprehensive understanding of their contributions to CNS health and during neurological disorders.


Asunto(s)
Antígenos , Sistema Nervioso Central , Humanos , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Antígenos/inmunología , Antígenos/metabolismo , Neuroglía/metabolismo , Neuroglía/inmunología , Neuroglía/fisiología , Neuronas/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteoglicanos
7.
J Neurol ; 271(4): 1999-2009, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38157030

RESUMEN

BACKGROUND: Neuronal pentraxin-2 (NPTX2), crucial for synaptic functioning, declines in cerebrospinal fluid (CSF) as cognition deteriorates. The variations of CSF NPTX2 across mild cognitive impairment (MCI) due to Alzheimer's disease (AD) and its association with brain metabolism remain elusive, albeit relevant for patient stratification and pathophysiological insights. METHODS: We retrospectively analyzed 49 MCI-AD patients grouped by time until dementia (EMCI, n = 34 progressing within 2 years; LMCI, n = 15 progressing later/stable at follow-up). We analyzed demographic variables, cognitive status (MMSE score), and CSF NPTX2 levels using a commercial ELISA assay in EMCI, LMCI, and a control group of age-/sex-matched individuals with other non-dementing disorders (OND). Using [18F]FDG PET scans for voxel-based analysis, we explored correlations between regional brain metabolism metrics and CSF NPTX2 levels in MCI-AD patients, accounting for age. RESULTS: Baseline and follow-up MMSE scores were lower in LMCI than EMCI (p value = 0.006 and p < 0.001). EMCI exhibited significantly higher CSF NPTX2 values than both LMCI (p = 0.028) and OND (p = 0.006). We found a significant positive correlation between NPTX2 values and metabolism of bilateral precuneus in MCI-AD patients (p < 0.005 at voxel level, p < 0.05 with family-wise error correction at the cluster level). CONCLUSIONS: Higher CSF NPTX2 in EMCI compared to controls and LMCI suggests compensatory synaptic responses to initial AD pathology. Disease progression sees these mechanisms overwhelmed, lowering CSF NPTX2 approaching dementia. Positive CSF NPTX2 correlation with precuneus glucose metabolism links to AD-related metabolic changes across MCI course. These findings posit CSF NPTX2 as a promising biomarker for both AD staging and progression risk stratification.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Estudios Retrospectivos , Biomarcadores/líquido cefalorraquídeo , Encéfalo/patología , Péptidos beta-Amiloides/metabolismo , Proteínas tau/líquido cefalorraquídeo , Progresión de la Enfermedad
8.
Neurobiol Aging ; 117: 212-221, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35780561

RESUMEN

We explored the brain metabolism correlates of emergent cerebrospinal fluid (CSF) biomarkers in a group of 26 patients with prodromal Alzheimer's disease (AD). Distinct volumes of interest (VOIs) expressed the sites of correlation between CSF biomarkers and brain metabolism as determined on [18F]FDG-PET images, as well as of significant hypometabolism in patients compared to healthy controls. Neurogranin- and α-synuclein-VOIs included left precuneus and/or posterior cingulate cortex (PC and/or PCC) and partially overlapped hypometabolism at those sites. ß-synuclein- and neurofilament light chain (NfL)-VOIs regarded either left or right lateral temporal areas, respectively, with partial overlap with hypometabolism only for the ß-synuclein-VOI, whereas the NfL-VOI did not include hypometabolic regions. We speculate that CSF neurogranin and α-synuclein express an already established hippocampal damage leading to PC and/or PCC deafferentation and hypometabolism. ß-synuclein may represent the progression of synaptopathy in the temporal lobe, while NfL the axonal injury in right temporal regions where neuronal loss is not yet evident.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Biomarcadores/líquido cefalorraquídeo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Humanos , Neurogranina , Tomografía de Emisión de Positrones/métodos , Datos Preliminares , alfa-Sinucleína/metabolismo , Sinucleína beta/metabolismo
9.
Life (Basel) ; 12(11)2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36362858

RESUMEN

Coronavirus disease 2019 (COVID-19) in hemodialysis patients (HD) is characterized by heterogeneity of clinical presentation and outcomes. To stratify patients, we collected clinical and laboratory data in two cohorts of HD patients at COVID-19 diagnosis and during the following 4 weeks. Baseline and longitudinal values were used to build a linear mixed effect model (LME) and define different clusters. The development of the LME model in the derivation cohort of 17 HD patients (66.7 ± 12.3 years, eight males) allowed the characterization of two clusters (cl1 and cl2). Patients in cl1 presented a prevalence of females, higher lymphocyte count, and lower levels of lactate dehydrogenase, C-reactive protein, and CD8 + T memory stem cells as a possible result of a milder inflammation. Then, this model was tested in an independent validation cohort of 30 HD patients (73.3 ± 16.3 years, 16 males) assigned to cl1 or cl2 (16 and 14 patients, respectively). The cluster comparison confirmed that cl1 presented a milder form of COVID-19 associated with reduced disease activity, hospitalization, mortality rate, and oxygen requirement. Clustering analysis on longitudinal data allowed patient stratification and identification of the patients at high risk of complications. This strategy could be suitable in different clinical settings.

10.
CNS Drugs ; 36(1): 83-96, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894339

RESUMEN

BACKGROUND: Defining immune mechanisms leading to multiple sclerosis (MS) is difficult, due to the great inter-individual difference in immune system responses. The anti-CD52 antibody alemtuzumab transiently abolishes differences in immune parameters among individuals, allowing analysis of subsequent immune cell repopulation patterns, and their possible role in MS. OBJECTIVE: To evaluate the correlation between innate and adaptive immune cell subsets and disease activity in MS in the context of treatment with alemtuzumab. METHODS: A two-center observational cohort of patients treated with alemtuzumab underwent immune profiling of T, B, and natural killer (NK) cells, biomarker, clinical and radiological follow-up. RESULTS: After treatment, the percentage of NK and B cells increased; NK, T- and B-cell populations underwent a profound rearrangement. Within the effector T-cell compartment, treatment led to a transient decrease, followed by an increase, of T-helper 1 cells, and to a transient decrease of T-helper 17 cells. Within the T-regulatory compartment, naïve T-regulatory cells increased. Within the B-cell compartment, memory B cells and mature B cells decreased, whereas transitional B cells increased. Within the NK cell compartment, CD56bright NK cells increased. Subjects without disease activity had a greater decrease in serum NfL and greater NK cell/CD3+ T cell ratio. NK cell numbers at baseline and after treatment influenced reconstitution of T and B cells, being inversely correlated with the reconstitution of proinflammatory CD3+ T cells and mature B cells, and directly correlated to the increase in transitional B cells. CONCLUSIONS: The results of this study provide novel evidence that NK cells influence reconstitution of adaptive immune cells upon alemtuzumab and that patients with a successful response to alemtuzumab have an early immune reconstitution dominated by NK cells.


Asunto(s)
Alemtuzumab/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Células Asesinas Naturales/inmunología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Adulto , Biomarcadores/sangre , Femenino , Humanos , Italia , Estudios Longitudinales , Masculino , Esclerosis Múltiple Recurrente-Remitente/inmunología
11.
Hum Mol Genet ; 17(13): 1877-89, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18337304

RESUMEN

Mutations in the gene MPZ, encoding myelin protein zero (MPZ), cause inherited neuropathies collectively called Charcot-Marie-Tooth type 1B (CMT1B). Based on the age of onset, clinical and pathological features, most MPZ mutations are separable into two groups: one causing a severe, early-onset, demyelinating neuropathy and a second, causing a late-onset neuropathy with prominent axonal loss. To investigate potential pathomechanisms underlying the two phenotypes, we transiently transfected HeLa cells with two late-onset (T95M, H10P) and two early-onset (H52R, S22_W28 deletion) mutations and analyzed their effects on intracellular protein trafficking, glycosylation, cell viability and intercellular adhesion. We found that the two late-onset mutations were both transported to the cell membrane and moderately reduced MPZ-mediated intercellular adhesion. The two early-onset mutations caused two distinct abnormalities. H52R was correctly glycosylated and trafficked to the plasma membrane, but strongly affected intercellular adhesion. When co-expressed with wild-type MPZ (wtMPZ), a functional dominant negative effect was observed. Alternatively, S22_W28 deletion was retained within the cytoplasm and reduced both adhesion caused by wtMPZ and cellular viability. Since the same trafficking patterns were observed in transfected murine Schwann cells, they are not an artifact of heterologous cell expression. Our results suggest that at least some late-onset mutations cause a partial loss of function in the transfected cells, whereas multiple abnormal gain of function pathways can result in early-onset neuropathy. Further characterization of these pathways will lead to a better understanding of the pathogenesis of CMT1B and a rational basis for treating these debilitating inherited neuropathies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/epidemiología , Enfermedad de Charcot-Marie-Tooth/genética , Mutación Missense , Proteína P0 de la Mielina/genética , Proteína P0 de la Mielina/metabolismo , Edad de Inicio , Animales , Apoptosis , Agregación Celular , Supervivencia Celular , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Genes Reporteros , Glicosilación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ratones , Proteína P0 de la Mielina/análisis , Pliegue de Proteína , Transporte de Proteínas
12.
J Neuropathol Exp Neurol ; 68(5): 441-55, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19525893

RESUMEN

We investigated the contribution of Schwann cell-derived ciliary neurotrophic factor (CNTF) to the pathogenesis of Charcot-Marie-Tooth disease type 1A (CMT1A) and addressed the question as to whether it plays a role in the development of axonal damage observed in the disease, with aging. Ciliary neurotrophic factor was underexpressed in experimental CMT1A but not in other models of hereditary neuropathies. Sciatic nerve crush experiments and dosage of CNTF at different time points showed that expression of this trophic factor remained significantly lower in CMT1A rats than in normal controls; moreover, in uninjured CMT1A sciatic nerves CNTF levels further decreased with ageing, thus paralleling the molecular signs of axonal impairment, that is increased expression of non-phosphorylated neurofilaments and amyloid precursor protein. Administration of CNTF to dorsal root ganglia cultures reduced dephosphorylation of neurofilaments in CMT1A cultures, without improving demyelination. Taken together, these results provide further evidence that the production of CNTF by Schwann cells is markedly reduced in CMT1A. Moreover, the observations suggest that trophic support to the axon is impaired in CMT1A and that further studies on the therapeutic use of trophic factors or their derivatives in experimental and human CMT1A are warranted.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de la Mielina/genética , Factores de Edad , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Axones/patología , Biopsia , Células Cultivadas , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/patología , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/farmacología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Ensayo de Inmunoadsorción Enzimática/métodos , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas de Neurofilamentos , Ratas , Ratas Transgénicas , Factor de Transcripción STAT3/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Nervio Ciático/citología , Neuropatía Ciática/metabolismo , Neuropatía Ciática/patología , Factores de Tiempo
13.
Cell Death Dis ; 10(2): 85, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30692524

RESUMEN

Administration of mesenchymal stem cells (MSC) ameliorate experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), at both clinical and neuropathological levels. The therapeutic properties of MSC in EAE are mainly mediated by the modulation of pathogenic immune response, but other neurotropic effects, including decreased demyelination and axonal loss as well as promotion of tissue repair, play also a role. Properly controlled phase II clinical trials to explore the potential of MSC transplantation as a treatment for MS are underway. Interferon beta (IFNß) is an approved treatment for relapsing-remitting and secondary progressive MS. Here, we explored the possibility that IFNß might influence the therapeutic potential of MSC, in view of possible synergistic effects as add-on therapy. IFNß enhanced the immunomodulatory functions of MSC and induced the expression of secretory leukocyte protease inhibitor (Slpi) and hepatocyte growth factor (Hgf), two soluble mediators involved in immune and regenerative functions of MSC. At molecular level, IFNß induced a rapid and transient phosphorylation of STAT1 and STAT3, the transcription factors responsible for Slpi and Hgf induction. Concomitantly, IFNß dynamically affected the activity of mTOR, a key checkpoint in the control of metabolic pathways. Indeed, the impairment of mTOR activity observed early upon exposure to IFNß, was followed by a long-lasting induction of mTOR signaling, that was associated with an increased glycolytic capacity in MSC. When induced to switch their energetic metabolism towards glycolysis, MSC showed an improved ability to control T-cell proliferation. These results suggest that modifications of MSC energetic metabolism induced by IFNß may contribute to promote MSC immunomodulatory function and support a role for metabolic pathways in the therapeutic function of MSC. Altogether, these findings support the idea of a combined treatment for MS, in which the immunomodulatory and possibly regenerative activity of MSC could be enhanced by the administration of IFNß.


Asunto(s)
Glucosa/metabolismo , Interferón beta/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/inmunología , Serina-Treonina Quinasas TOR/inmunología , Animales , Proliferación Celular/efectos de los fármacos , Glucosa/inmunología , Inmunomodulación/efectos de los fármacos , Activación de Linfocitos , Células Madre Mesenquimatosas/metabolismo , Ratones , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Transfección
14.
Arch Neurol ; 64(10): 1531-3, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17923639

RESUMEN

OBJECTIVE: To describe the unique case of a patient with multiple sclerosis (MS) and anti-myelin-associated glycoprotein (MAG) polyneuropathy who developed MS relapses after treatment with rituximab. DESIGN: Case report. SETTING: Department of Neurosciences, Ophthalmology, and Genetics, University of Genova, Genova, Italy. Patient A 59-year-old man with an 18-year history of MS presented with an unusually rapid progression of paraparesis with hypopallesthesia and areflexia in 4 limbs. Neurophysiological and serological studies led to the diagnosis of anti-MAG polyneuropathy. Cerebrospinal fluid analysis disclosed the loss of oligoclonal IgG bands that were previously detected at MS onset. Intervention Rituximab was administered at a dosage of 375 mg/m(2)/wk for 4 weeks. Result The patient developed 2 corticosteroid-responsive MS relapses with improvement of the polyneuropathy. CONCLUSION: Rituximab can be effective in anti-MAG polyneuropathy but can possibly lead to unexpected consequences in individuals with MS.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades Autoinmunes del Sistema Nervioso/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Glicoproteína Asociada a Mielina/inmunología , Polineuropatías/tratamiento farmacológico , Anticuerpos Monoclonales de Origen Murino , Autoanticuerpos/líquido cefalorraquídeo , Enfermedades Autoinmunes del Sistema Nervioso/complicaciones , Electrofisiología , Humanos , Inmunoglobulina G/líquido cefalorraquídeo , Inmunoglobulina M/líquido cefalorraquídeo , Focalización Isoeléctrica , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/complicaciones , Examen Neurológico , Polineuropatías/complicaciones , Recurrencia , Rituximab
15.
Stem Cell Res Ther ; 3(1): 3, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22277374

RESUMEN

Stem cells are currently seen as a treatment for tissue regeneration in neurological diseases such as multiple sclerosis, anticipating that they integrate and differentiate into neural cells. Mesenchymal stem cells (MSCs), a subset of adult progenitor cells, differentiate into cells of the mesodermal lineage but also, under certain experimental circumstances, into cells of the neuronal and glial lineage. Their clinical development, however, has been significantly boosted by the demonstration that MSCs display significant therapeutic plasticity mainly occurring through bystander mechanisms. These features have been exploited in the effective treatment of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis where the inhibition of the autoimmune response resulted in a significant amelioration of disease and decrease of demyelination, immune infiltrates and axonal loss. Surprisingly, these effects do not require MSCs to engraft in the central nervous system but depend on the cells' ability to inhibit pathogenic immune responses both in the periphery and inside the central nervous system and to release neuroprotective and pro-oligodendrogenic molecules favoring tissue repair. These results paved the road for the utilization of MSCs for the treatment of multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Linfocitos T CD4-Positivos/metabolismo , Sistema Nervioso Central/inmunología , Vías de Administración de Medicamentos , Factores de Transcripción Forkhead/metabolismo
16.
J Peripher Nerv Syst ; 12(2): 102-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17565535

RESUMEN

We evaluated the efficacy and safety of rituximab in an open-label, uncontrolled study of 13 patients with polyneuropathy associated with antibodies to myelin-associated glycoprotein (MAG) and correlated the response to therapy with clinical and laboratory features. One year after rituximab therapy, anti-MAG immunoglobulin M (IgM) titers were significantly reduced. At that time, eight patients (62%) had improved in both the inflammatory neuropathy cause and treatment (INCAT) sensory sumscore and the Medical Research Council sumscore for muscle strength and seven of them also in the INCAT disability score. The improvement in the mean INCAT sensory sumscore was significant at 12 months and correlated with lower anti-MAG antibody at entry and at follow-up. This study suggests that rituximab may be efficacious in patients with anti-MAG associated neuropathy and particularly on sensory impairment and in those with moderately elevated antibody titers. These findings suggest that antibody reduction below a critical level may be necessary to achieve clinical improvement.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Autoanticuerpos/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Glicoproteína Asociada a Mielina/inmunología , Polineuropatías/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Monoclonales de Origen Murino , Antígenos CD19/efectos de los fármacos , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Subgrupos de Linfocitos B/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Western Blotting , Electromiografía , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina M/sangre , Inmunoglobulina M/efectos de los fármacos , Inmunoglobulina M/inmunología , Masculino , Persona de Mediana Edad , Fuerza Muscular/efectos de los fármacos , Conducción Nerviosa/efectos de los fármacos , Polineuropatías/inmunología , Rituximab
17.
Eur J Neurosci ; 23(6): 1445-52, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16553608

RESUMEN

Clinical progression in hereditary and acquired demyelinating disorders of both the central and peripheral nervous system is mainly due to a time-dependent axonal impairment. We established 90-day dorsal root ganglia (DRG) cultures from a rat model of Charcot-Marie-Tooth type 1A (CMT1A) neuropathy to evaluate the structure of myelin and axons, and the expression of myelin-related proteins and cytoskeletal components, by morphological and molecular techniques. Both wild-type and CMT1A cultures were rich in myelinated fibres. Affected cultures showed dysmyelinated internodes and focal myelin swellings. Furthermore, uncompacted myelin and smaller axons with increased neurofilament (NF) density were found by electron microscopy, and Western blots showed higher levels of nonphosphorylated NF. Confocal microscopy demonstrated an abnormal distribution of the myelin-associated glycoprotein which, instead of being expressed at the noncompact myelin level, showed focal accumulation along the internodes while other myelin proteins were normally distributed. These findings suggest that CMT1A DRG cultures, similarly to the animal model and human disease, undergo axonal atrophy over a period of time. This model may be utilized to study the molecular changes underlying demyelination and secondary axonal impairment. As axonal damage may occur after just 3 months and tissue cultures represent a strictly controlled environment, this model may be ideal for testing neuroprotective therapies.


Asunto(s)
Axones/patología , Enfermedad de Charcot-Marie-Tooth/patología , Enfermedades Desmielinizantes/patología , Ganglios Espinales/patología , Animales , Axones/ultraestructura , Western Blotting , Células Cultivadas , Genotipo , Microscopía Confocal , Microscopía Electrónica , Proteínas de la Mielina/genética , Proteínas de Neurofilamentos/metabolismo , Organismos Modificados Genéticamente , Fenotipo , Fosforilación , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Mol Cell Neurosci ; 28(4): 703-14, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15797717

RESUMEN

To reveal the spectrum of genes that are modulated in Charcot-Marie-Tooth neuropathy type 1A (CMT1A), which is due to overexpression of the gene coding for the peripheral myelin protein 22 (pmp22), we performed a cDNA microarray experiment with cDNA from sciatic nerves of a rat model of the disease. In homozygous pmp22 overexpressing animals, we found a significant down-regulation of 86 genes, while only 23 known genes were up-regulated, suggesting that the increased dosage of pmp22 induces a general down-regulation of gene expression in peripheral nerve tissue. Classification of the modulated genes into functional categories leads to the identification of some pathways altered by overexpression of pmp22. In particular, a selective down-regulation of the ciliary neurotrophic factor transcript and of genes coding for proteins involved in cell cycle regulation, for cytoskeletal components and for proteins of the extracellular matrix, was observed. Cntf expression was further studied by real-time PCR and ELISA technique in pmp22 transgenic sciatic nerves, human CMT1A sural nerve biopsies, and primary cultures of transgenic Schwann cells. According to the results of cDNA microarray analysis, a down-regulation of cntf, both at the mRNA and protein level, was found in all the conditions tested. These results are relevant to reveal the molecular function of PMP22 and the pathogenic mechanism of CMT1A. In particular, finding a specific reduction of cntf expression in CMT1A Schwann cells suggests that overexpression of pmp22 significantly affects the ability of Schwann cells to offer a trophic support to the axon, which could be a factor, among other, responsible for the development of axonal atrophy in human and experimental CMT1A.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , ADN Complementario/genética , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Animales Modificados Genéticamente , Células Cultivadas , Enfermedad de Charcot-Marie-Tooth/metabolismo , ADN Complementario/biosíntesis , Perfilación de la Expresión Génica/métodos , Humanos , Proteínas de la Mielina/biosíntesis , Proteínas de la Mielina/genética , Ratas
19.
Neurobiol Dis ; 16(1): 263-73, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15207283

RESUMEN

Charcot-Marie-Tooth type 1A (CMT1A) is a hereditary demyelinating neuropathy due to an increased genetic dosage of the peripheral myelin protein 22 (PMP22). The mechanisms leading from PMP22 overexpression to impairment of myelination are still unclear. We evaluated expression and processing of PMP22, viability, proliferation, migration, motility and shaping properties, and ability of forming myelin of PMP22 transgenic (PMP22(tg)) Schwann cells in culture. In basal conditions, PMP22(tg) Schwann cells, although expressing higher PMP22 levels than control ones, show normal motility, migration and shaping properties. Addition of forskolin to the media induces an additional stimulation of PMP22 expression and results in an impairment of cells migration and motility, and a reduction of cell area and perimeter. Similarly, co-culturing transgenic Schwann cells with neurons causes an altered cells differentiation and an impairment of myelin formation. In conclusion, exposure of PMP22(tg) Schwann to the axon or to axonal-mimicking stimuli significantly affects the transition of transgenic Schwann cells to the myelinating phenotype.


Asunto(s)
Diferenciación Celular/genética , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/metabolismo , Proteínas de la Mielina/biosíntesis , Células de Schwann/citología , Células de Schwann/metabolismo , Animales , Animales Modificados Genéticamente , Células Cultivadas , Enfermedad de Charcot-Marie-Tooth/patología , Proteínas de la Mielina/genética , Ratas
20.
Exp Neurol ; 190(1): 213-23, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15473994

RESUMEN

We investigated early peripheral nervous system impairment in PMP22-transgenic rats ("CMT rat"), an established animal model for Charcot-Marie-Tooth disease 1A, at postnatal day 30 (P30), when the clinical phenotype is not yet apparent. Hemizygous CMT1A rats and wildtype littermates were studied by means of behavioral examination, electrophysiology, molecular biology, and light microscopy analysis. Behavioral studies only showed, a mild, but significant, decrease in toe spread 1-5, suggesting a weakness of distal foot muscles in CMT1A rats compared with normal littermates. Nerve conduction studies disclosed a severe slowing in motor conduction velocity, a temporal dispersion and a dramatic decrease of amplitude of motor waves in P30 transgenic animals. Coherently with a demyelinating process, affected nerves showed a significant thinning of myelin. Interestingly, axonal diameter and area were unchanged, but expression of non-phosphorylated neurofilaments was increased in CMT1A rats compared with normal controls. Our results confirm the fidelity of this animal model to human disease. Similarly, in young CMT1A patients, the MCV is significantly reduced and the muscle weakness is confined to distal segments, whereas morphological and morphometrical signs of axonal atrophy are absent. However, the presence of a molecular and functional damage of the axons suggests that this may be the correct moment to start neuroprotective therapies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/fisiopatología , Neuropatía Ciática/fisiopatología , Factores de Edad , Animales , Animales Modificados Genéticamente , Axones/patología , Conducta Animal , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electrofisiología , Predisposición Genética a la Enfermedad , Ratones , Actividad Motora/genética , Debilidad Muscular/genética , Debilidad Muscular/fisiopatología , Proteínas de la Mielina/genética , Vaina de Mielina/patología , Conducción Nerviosa/genética , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Neuropatía Ciática/genética , Neuropatía Ciática/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA