Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Neuroinflammation ; 19(1): 10, 2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991629

RESUMEN

BACKGROUND: Astrocytes are the most numerous glial cell type with important roles in maintaining homeostasis and responding to diseases in the brain. Astrocyte function is subject to modulation by microRNAs (miRs), which are short nucleotide strands that regulate protein expression in a post-transcriptional manner. Understanding the miR expression profile of astrocytes in disease settings provides insight into the cellular stresses present in the microenvironment and may uncover pathways of therapeutic interest. METHODS: Laser-capture microdissection was used to isolate human astrocytes surrounding stroke lesions and those from neurological control tissue. Astrocytic miR expression profiles were examined using quantitative reverse transcription polymerase chain reaction (RT-qPCR). Primary human fetal astrocytes were cultured under in vitro stress conditions and transfection of a miR mimic was used to better understand how altered levels of miR-210 affect astrocyte function. The astrocytic response to stress was studied using qPCR, enzyme-linked immunosorbent assays (ELISAs), measurement of released lactate, and Seahorse. RESULTS: Here, we measured miR expression levels in astrocytes around human ischemic stroke lesions and observed differential expression of miR-210 in chronic stroke astrocytes compared to astrocytes from neurological control tissue. We also identified increased expression of miR-210 in mouse white matter tissue around middle cerebral artery occlusion (MCAO) brain lesions. We aimed to understand the role of miR-210 in primary human fetal astrocytes by developing an in vitro assay of hypoxic, metabolic, and inflammatory stresses. A combination of hypoxic and inflammatory stresses was observed to upregulate miR-210 expression. Transfection with miR-210-mimic (210M) increased glycolysis, enhanced lactate export, and promoted an anti-inflammatory transcriptional and translational signature in astrocytes. Additionally, 210M transfection resulted in decreased expression of complement 3 (C3) and semaphorin 5b (Sema5b). CONCLUSIONS: We conclude that miR-210 expression in human astrocytes is modulated in response to ischemic stroke disease and under in vitro stress conditions, supporting a role for miR-210 in the astrocytic response to disease conditions. Further, the anti-inflammatory and pro-glycolytic impact of miR-210 on astrocytes makes it a potential candidate for further research as a neuroprotective agent.


Asunto(s)
Astrocitos/metabolismo , Inflamación/metabolismo , MicroARNs/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Células HeLa , Humanos , Inflamación/genética , Captura por Microdisección con Láser , Ratones , MicroARNs/genética , Accidente Cerebrovascular/genética
2.
Ann Neurol ; 81(6): 811-824, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28470695

RESUMEN

OBJECTIVE: Degeneration of oligodendroglial distal processes has been identified as an early event in multiple sclerosis (MS) lesion development. Our objective was to further define the development of the "dying-back" oligodendrocyte lesion in situ and to model the development and potential reversibility of such responses using dissociated cultures of adult human brain-derived oligodendrocytes. METHODS: In situ analyses were performed on glutaraldehyde-fixed thin sections of clinically acute and pathologically active cases of MS. In vitro studies were conducted using adult human brain-derived oligodendrocytes challenged by metabolic stress conditions (low nutrient/glucose). RESULTS: In situ analyses indicated a spectrum of myelin changes in the presence of morphologically intact oligodendrocytes; these included degeneration of the inner cytoplasmic tongue with increasing sizes of intramyelinic bleb formation that could result in radial fractures of the myelin sheath. Macrophages with ingested myelin fragments were identified only once the fragmentation was established. In vitro studies indicated that oligodendrocyte process retraction, which was linked to reduced glycolytic respiratory activity, is reversible until a critical time point. Subsequent cell death was not linked to caspase-3-dependent programs. Gene expression studies conducted at the latest reversible time point revealed reduced expression of pathways associated with cell process outgrowth and myelination, as well as with metabolic activity. INTERPRETATION: Our findings reveal the potential to protect and possibly restore myelin elaborated by existent oligodendrocytes in early and evolving MS lesions, and suggest the necessity of ongoing studies of the mechanisms underlying subsequent adult human oligodendrocyte cell death. Ann Neurol 2017;81:811-824.


Asunto(s)
Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Animales , Caspasa 3/metabolismo , Muerte Celular , Humanos , Ratas , Ratas Sprague-Dawley
3.
J Immunol ; 196(8): 3375-84, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26962228

RESUMEN

Multifocal inflammatory lesions featuring destruction of lipid-rich myelin are pathologic hallmarks of multiple sclerosis. Lesion activity is assessed by the extent and composition of myelin uptake by myeloid cells present in such lesions. In the inflamed CNS, myeloid cells are comprised of brain-resident microglia, an endogenous cell population, and monocyte-derived macrophages, which infiltrate from the systemic compartment. Using microglia isolated from the adult human brain, we demonstrate that myelin phagocytosis is dependent on the polarization state of the cells. Myelin ingestion is significantly enhanced in cells exposed to TGF-ß compared with resting basal conditions and markedly reduced in classically activated polarized cells. Transcriptional analysis indicated that TGF-ß-treated microglia closely resembled M0 cells. The tyrosine kinase phagocytic receptor MerTK was one of the most upregulated among a select number of differentially expressed genes in TGF-ß-treated microglia. In contrast, MerTK and its known ligands, growth arrest-specific 6 and Protein S, were downregulated in classically activated cells. MerTK expression and myelin phagocytosis were higher in CNS-derived microglia than observed in monocyte-derived macrophages, both basally and under all tested polarization conditions. Specific MerTK inhibitors reduced myelin phagocytosis and the resultant anti-inflammatory biased cytokine responses for both cell types. Defining and modulating the mechanisms that regulate myelin phagocytosis has the potential to impact lesion and disease evolution in multiple sclerosis. Relevant effects would include enhancing myelin clearance, increasing anti-inflammatory molecule production by myeloid cells, and thereby permitting subsequent tissue repair.


Asunto(s)
Esclerosis Múltiple/inmunología , Vaina de Mielina/inmunología , Células Mieloides/inmunología , Fagocitosis/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Adulto , Encéfalo/citología , Encéfalo/inmunología , Polaridad Celular/fisiología , Células Cultivadas , Regulación hacia Abajo , Humanos , Inflamación/inmunología , Inflamación/patología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Macrófagos/inmunología , Microglía/citología , Microglía/inmunología , Esclerosis Múltiple/patología , Proteína S/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba , Tirosina Quinasa c-Mer
4.
J Neurosci ; 36(17): 4698-707, 2016 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-27122029

RESUMEN

UNLABELLED: Multiple sclerosis (MS) lesions feature demyelination with limited remyelination. A distinct injury phenotype of MS lesions features dying back of oligodendrocyte (OL) terminal processes, a response that destabilizes myelin/axon interactions. This oligodendrogliopathy has been linked with local metabolic stress, similar to the penumbra of ischemic/hypoxic states. Here, we developed an in vitro oligodendrogliopathy model using human CNS-derived OLs and related this injury response to their distinct bioenergetic properties. We determined the energy utilization properties of adult human surgically derived OLs cultured under either optimal or metabolic stress conditions, deprivation of growth factors, and glucose and/or hypoxia using a Seahorse extracellular flux analyzer. Baseline studies were also performed on OL progenitor cells derived from the same tissue and postnatal rat-derived cells. Under basal conditions, adult human OLs were less metabolically active than their progenitors and both were less active than the rat cells. Human OLs and progenitors both used aerobic glycolysis for the majority of ATP production, a process that contributes to protein and lipid production necessary for myelin biosynthesis. Under stress conditions that induce significant process retraction with only marginal cell death, human OLs exhibited a significant reduction in overall energy utilization, particularly in glycolytic ATP production. The stress-induced reduction of glycolytic ATP production by the human OLs would exacerbate myelin process withdrawal while favoring cell survival, providing a potential basis for the oligodendrogliopathy observed in MS. The glycolytic pathway is a potential therapeutic target to promote myelin maintenance and enhance repair in MS. SIGNIFICANCE STATEMENT: The neurologic deficits that characterize multiple sclerosis (MS) reflect disruption of myelin (demyelination) within the CNS and failure of repair (remyelination). We define distinct energy utilization properties of human adult brain-derived oligodendrocytes and oligodendrocyte progenitor cells under conditions of metabolic stress that model the initial relapsing and subsequent progressive phases of MS. The observed changes in energy utilization affect both cell survival and myelination capacity. These processes may be amenable to therapeutic interventions to limit the extent of cumulative tissue injury and to promote repair in MS.


Asunto(s)
Enfermedades Desmielinizantes/patología , Glucólisis , Esclerosis Múltiple/patología , Oligodendroglía/metabolismo , Células Madre/metabolismo , Animales , Encéfalo/metabolismo , Muerte Celular , Supervivencia Celular , Células Cultivadas , Humanos , Vaina de Mielina/metabolismo , Oligodendroglía/patología , Ratas , Ratas Sprague-Dawley
6.
Mult Scler ; 22(9): 1114-24, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27207458

RESUMEN

Recent experimental and clinical studies on astrocytes are unraveling the capabilities of these multi-functional cells in normal homeostasis, and in central nervous system (CNS) disease. This review focuses on understanding their behavior in all aspects of the initiation, evolution, and resolution of the multiple sclerosis (MS) lesion. Astrocytes display remarkable flexibility and variability of their physical structure and biochemical output, each aspect finely tuned to the specific stage and location of the disease, participating in both pathogenic and beneficial changes seen in acute and progressive forms. As examples, chemo-attractive or repulsive molecules may facilitate the entry of destructive immune cells but may also aid in the recruitment of oligodendrocyte precursors, essential for repair. Pro-inflammatory cytokines may attack pathogenic cells and also destroy normal oligodendrocytes, myelin, and axons. Protective trophic factors may also open the blood-brain barrier and modulate the extracellular matrix to favor recruitment and persistence of CNS-specific immune cells. A chronic glial scar may confer structural support following tissue loss and inhibit ingress of further noxious insults and also inhibit migration of reparative cells and molecules into the damaged tissue. Continual study into these processes offers the therapeutic opportunities to enhance the beneficial capabilities of these cells while limiting their destructive effects.


Asunto(s)
Astrocitos/patología , Sistema Nervioso Central/patología , Esclerosis Múltiple/patología , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/fisiopatología , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/fisiopatología , Fenotipo , Transducción de Señal
7.
Ann Neurol ; 74(5): 709-20, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23818336

RESUMEN

OBJECTIVE: To define the functional significance of increased miR-155 expression in myeloid cells in multiple sclerosis (MS). METHODS: miR-155 expression levels were measured in CD14+ monocytes from untreated relapsing-remitting MS patients and compared to healthy controls. Similar microRNA (miRNA) analyses were performed in laser-captured CD68+ cells from perivascular (blood-derived macrophages) and parenchymal (microglia) brain regions in both active MS lesions and noninflammatory cases. Using human adult blood-derived macrophages and brain-derived microglia, in vitro experiments were performed to demonstrate how miR-155 influences the polarization state, phenotype, and functional properties of myeloid cells, in addition to their ability to subsequently impact adaptive T-cell responses. RESULTS: In MS, miR-155 expression was significantly increased in both peripheral circulating CD14+ monocytes and active lesions (CD68+ cells) compared to control donor monocytes and parenchymal microglia, respectively. In vitro, miR-155 was significantly increased in both M1-polarized primary human macrophages and microglia. Transfection of an miR-155 mimic increased proinflammatory cytokine secretion and costimulatory surface marker expression in both cell types; an miR-155 inhibitor decreased proinflammatory cytokine expression. Coculture experiments demonstrated that allogeneic T-cell responses were significantly enhanced in the presence of miR-155-transfected myeloid cells compared to controls. INTERPRETATION: Our results demonstrate that miR-155 regulates proinflammatory responses in both blood-derived and central nervous system (CNS)-resident myeloid cells, in addition to impacting subsequent adaptive immune responses. Differential miRNA expression may therefore provide insight into mechanisms responsible for distinct phenotypic and functional properties of myeloid cells, thus impacting their ability to influence CNS injury and repair.


Asunto(s)
Polaridad Celular/fisiología , MicroARNs/genética , Esclerosis Múltiple/genética , Células Mieloides/patología , Inmunidad Adaptativa , Adulto , Anciano , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Polaridad Celular/inmunología , Proliferación Celular , Femenino , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , MicroARNs/metabolismo , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
8.
FASEB J ; 25(5): 1509-18, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21248243

RESUMEN

Fingolimod (FTY720) is a sphingosine 1-phosphate (S1P) receptor modulator that regulates lymphocyte trafficking and exerts pleiotropic actions on oligodendrocytes (OLGs) and other neural cells. The purpose of this study was to investigate the role of S1P receptors in a non-T-cell model of demyelination, the cuprizone (cupr) model in C57BL/6 mice. Treatment with FTY720 (1 mg/kg) led to attenuated injury to OLGs, myelin, and axons in the corpus callosum (percentage of myelinated fibers was 44.7% in cupr-water and 63% in cupr-FTY720). Reactive astrogliosis and microgliosis were ameliorated when FTY720 was given from d 1, but astrogliosis was augmented when FTY720 was given from wk 4-9. FTY720 did not promote remyelination in this model. The protective effect of FTY720 was associated with decreased interleukin-1ß and CCL2 transcripts in the corpus callosum, as well as altered S1P1 expression. Targeted deletion of S1P1 in OLG lineage cells did not lead to obvious clinical phenotype, but resulted in subtle abnormalities in myelin and an increased susceptibility to cupr-induced demyelination. We conclude that S1P receptors expressed by neuroglia are involved in regulating the response to injury, and CNS effects of FTY720 could contribute to its favorable therapeutic response in multiple sclerosis.


Asunto(s)
Cuprizona/toxicidad , Receptores de Lisoesfingolípidos/metabolismo , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Western Blotting , Cuerpo Calloso/citología , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Clorhidrato de Fingolimod , Inmunohistoquímica , Inmunosupresores/farmacología , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Glicoles de Propileno/farmacología , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esfingosina/análogos & derivados , Esfingosina/farmacología , Esfingosina/uso terapéutico
9.
10.
Am J Pathol ; 176(6): 2682-94, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20413685

RESUMEN

Remyelination, which occurs subsequent to demyelination, contributes to functional recovery and is mediated by oligodendrocyte progenitor cells (OPCs) that have differentiated into myelinating cells. Therapeutics that impact remyelination in the CNS could be critical determinants of long-term functional outcome in multiple sclerosis (MS). Fingolimod is a S1P receptor modulator in MS clinical trials due to systemic anti-inflammatory properties, yet may impact cells within the CNS by crossing the blood-brain barrier. Previous studies using isolated dissociated cultures indicate that neural cells express S1P receptors and respond to receptor engagement. Our objective was to assess the effects of fingolimod on myelin-related processes within a multicellular environment that maintains physiological cell-cell interactions, using organotypic cerebellar slice cultures. Fingolimod treatment had no impact on myelin under basal conditions. Fingolimod treatment subsequent to lysolecithin-induced demyelination enhanced remyelination and process extension by OPCs and mature oligodendrocytes, while increasing microglia numbers and immunoreactivity for the astrocytic marker glial fibrillary acidic protein. The number of phagocytosing microglia was not increased by fingolimod. Using S1P receptor specific agonists and antagonists, we determined that fingolimod-induced effects on remyelination and astrogliosis were mediated primarily through S1P3 and S1P5, whereas enhanced microgliosis was mediated through S1P1 and S1P5. Taken together, these data demonstrate that fingolimod modulates multiple neuroglial cell responses, resulting in enhanced remyelination in organotypic slice cultures that maintain the complex cellular interactions of the mammalian brain.


Asunto(s)
Cerebelo/citología , Cerebelo/efectos de los fármacos , Enfermedades Desmielinizantes/tratamiento farmacológico , Inmunosupresores/farmacología , Vaina de Mielina/fisiología , Glicoles de Propileno , Esfingosina/análogos & derivados , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Cerebelo/patología , Cerebelo/fisiología , Enfermedades Desmielinizantes/inducido químicamente , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/uso terapéutico , Lisofosfatidilcolinas/toxicidad , Ratones , Microglía/citología , Microglía/efectos de los fármacos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/fisiopatología , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Glicoles de Propileno/farmacología , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/farmacología , Esfingosina/uso terapéutico , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Técnicas de Cultivo de Tejidos
11.
J Exp Med ; 200(2): 223-34, 2004 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-15263029

RESUMEN

Amino acid residues 111-129 represent an immunodominant epitope of myelin basic protein (MBP) in humans with human leukocyte antigen (HLA)-DRB1*0401 allele(s). The MBP 111-129-specific T cell clone MS2-3C8 was repeatedly isolated from a patient with multiple sclerosis (MS), suggesting an involvement of MS2-3C8 T cells in the pathogenesis. To address the pathogenic potential of the MS2-3C8 T cell clone, we generated transgenic (Tg) mice expressing its T cell receptor and restriction element, HLA-DRB1*0401, to examine the pathogenic characteristics of MS2-3C8 Tg T cells by adoptive transfer into HLA-DRB1*0401 Tg mice. In addition to the ascending paralysis typical of experimental autoimmune encephalomyelitis, mice displayed dysphagia due to restriction in jaw and tongue movements and abnormal gait. In accordance with the clinical phenotype, infiltrates of MS2-3C8 Tg T cells and inflammatory lesions were predominantly located in the brainstem and the cranial nerve roots in addition to the spinal cord and spinal nerve roots. Together, these data suggest a pathogenic role of MBP-specific T cells in inflammatory demyelination within the brainstem and cranial nerve roots during the progression of MS. This notion may help to explain the clinical and pathological heterogeneity of MS.


Asunto(s)
Antígenos HLA-DR/metabolismo , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Secuencia de Aminoácidos , Animales , Separación Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Cadenas HLA-DRB1 , Humanos , Inmunohistoquímica , Inflamación , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Esclerosis Múltiple/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/metabolismo , Factores de Tiempo
12.
Am J Pathol ; 174(5): 1880-90, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19349355

RESUMEN

Remyelination of lesions in the central nervous system contributes to neural repair following clinical relapses in multiple sclerosis. Remyelination is initiated by recruitment and differentiation of oligodendrocyte progenitor cells (OPCs) into myelinating oligodendrocytes. Simvastatin, a blood-brain barrier-permeable statin in multiple sclerosis clinical trials, has been shown to impact the in vitro processes that have been implicated in remyelination. Animals were fed a cuprizone-supplemented diet for 6 weeks to induce localized demyelination in the corpus callosum; subsequent return to normal diet for 3 weeks stimulated remyelination. Simvastatin was injected intraperitoneally during the period of coincident demyelination and OPC maturation (weeks 4 to 6), throughout the entire period of OPC responses (weeks 4 to 9), or during the remyelination-only phase (weeks 7 to 9). Simvastatin treatment (weeks 4 to 6) caused a decrease in myelin load and both Olig2(strong) and Nkx2.2(strong) OPC numbers. Simvastatin treatment (weeks 4 to 9 and 7 to 9) caused a decrease in myelin load, which was correlated with a reduction in Nkx2.2(strong) OPCs and an increase in Olig2(strong) cells, suggesting that OPCs were maintained in an immature state (Olig2(strong)/Nkx2.2(weak)). NogoA+ oligodendrocyte numbers were decreased during all simvastatin treatment regimens. Our findings suggest that simvastatin inhibits central nervous system remyelination by blocking progenitor differentiation, indicating the need to monitor effects of systemic immunotherapies that can access the central nervous system on brain tissue-repair processes.


Asunto(s)
Anticolesterolemiantes/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Simvastatina/farmacología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Western Blotting , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quelantes/administración & dosificación , Cuprizona/administración & dosificación , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/fisiología , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/patología , Proteínas del Tejido Nervioso/fisiología , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Factores de Transcripción/fisiología , Proteínas de Pez Cebra
13.
Lancet Oncol ; 10(5): 459-66, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19269895

RESUMEN

BACKGROUND: In 2004, a randomised phase III trial by the European Organisation for Research and Treatment of Cancer (EORTC) and National Cancer Institute of Canada Clinical Trials Group (NCIC) reported improved median and 2-year survival for patients with glioblastoma treated with concomitant and adjuvant temozolomide and radiotherapy. We report the final results with a median follow-up of more than 5 years. METHODS: Adult patients with newly diagnosed glioblastoma were randomly assigned to receive either standard radiotherapy or identical radiotherapy with concomitant temozolomide followed by up to six cycles of adjuvant temozolomide. The methylation status of the methyl-guanine methyl transferase gene, MGMT, was determined retrospectively from the tumour tissue of 206 patients. The primary endpoint was overall survival. Analyses were by intention to treat. This trial is registered with Clinicaltrials.gov, number NCT00006353. FINDINGS: Between Aug 17, 2000, and March 22, 2002, 573 patients were assigned to treatment. 278 (97%) of 286 patients in the radiotherapy alone group and 254 (89%) of 287 in the combined-treatment group died during 5 years of follow-up. Overall survival was 27.2% (95% CI 22.2-32.5) at 2 years, 16.0% (12.0-20.6) at 3 years, 12.1% (8.5-16.4) at 4 years, and 9.8% (6.4-14.0) at 5 years with temozolomide, versus 10.9% (7.6-14.8), 4.4% (2.4-7.2), 3.0% (1.4-5.7), and 1.9% (0.6-4.4) with radiotherapy alone (hazard ratio 0.6, 95% CI 0.5-0.7; p<0.0001). A benefit of combined therapy was recorded in all clinical prognostic subgroups, including patients aged 60-70 years. Methylation of the MGMT promoter was the strongest predictor for outcome and benefit from temozolomide chemotherapy. INTERPRETATION: Benefits of adjuvant temozolomide with radiotherapy lasted throughout 5 years of follow-up. A few patients in favourable prognostic categories survive longer than 5 years. MGMT methylation status identifies patients most likely to benefit from the addition of temozolomide. FUNDING: EORTC, NCIC, Nélia and Amadeo Barletta Foundation, Schering-Plough.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Terapia Combinada , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/uso terapéutico , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Regiones Promotoras Genéticas , Análisis de Supervivencia , Tasa de Supervivencia , Temozolomida , Proteínas Supresoras de Tumor/genética
14.
J Neuroimmunol ; 331: 28-35, 2019 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29566973

RESUMEN

Remyelination following myelin/oligodendrocyte injury in the central nervous system (CNS) is dependent on oligodendrocyte progenitor cells (OPCs) migrating into lesion sites, differentiating into myelinating oligodendrocytes (OLs), and ensheathing axons. Experimental models indicate that robust OPC-dependent remyelination can occur in the CNS; in contrast, histologic and imaging studies of lesions in the human disease multiple sclerosis (MS) indicate the variable extent of this response, which is particularly limited in more chronic MS lesions. Immune-mediated mechanisms can contribute either positively or negatively to the presence and functional responses of OPCs. This review addresses i) the molecular signature and functional properties of OPCs in the adult human brain; ii) the status (presence and function) of OPCs in MS lesions; iii) experimental models and in vitro data highlighting the contribution of adaptive and innate immune constituents to OPC injury and remyelination; and iv) effects of MS-directed immunotherapies on OPCs, either directly or indirectly via effects on specific immune constituents.


Asunto(s)
Encéfalo/citología , Células Precursoras de Oligodendrocitos/inmunología , Inmunidad Adaptativa , Adulto , Animales , Antígenos de Diferenciación/análisis , Diferenciación Celular , Células Cultivadas , Glucosa/farmacología , Humanos , Inmunidad Innata , Inmunoterapia , Ratones , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Proteínas del Tejido Nervioso/análisis , Neuroinmunomodulación , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Células Precursoras de Oligodendrocitos/fisiología , Oligodendroglía/citología , Ratas , Remielinización/fisiología
15.
J Neuropathol Exp Neurol ; 78(12): 1130-1146, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31665376

RESUMEN

Astrocytes are increasingly recognized as active contributors to the disease process in multiple sclerosis (MS), rather than being merely reactive. We investigated the expression of a selected microRNA (miRNA) panel that could contribute both to the injury and to the recovery phases of the disease. Individual astrocytes were laser microdissected from brain sections. We then compared the miRNAs' expressions in MS and control brain samples at different lesional stages in white versus grey matter regions. In active MS lesions, we found upregulation of ischemia-related miRNAs in white but not grey matter, often with reversion to the normal state in inactive lesions. In contrast to our previous findings on MS macrophages, expression of 2 classical inflammatory-related miRNAs, miRNA-155 and miRNA-146a, was reduced in astrocytes from active and chronic active MS lesions in white and grey matter, suggesting a lesser direct pathogenetic role for these miRNAs in astrocytes. miRNAs within the categories regulating aquaporin4 (-100, -145, -320) and glutamate transport/apoptosis/neuroprotection (-124a, -181a, and -29a) showed some contrasting responses. The regional and lesion-stage differences of expression of these miRNAs indicate the remarkable ability of astrocytes to show a wide range of selective responses in the face of differing insults and phases of resolution.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/patología , MicroARNs/metabolismo , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/metabolismo , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo , Encefalitis/complicaciones , Encefalitis/metabolismo , Femenino , Sustancia Gris/patología , Humanos , Masculino , Esclerosis Múltiple/etiología , Sustancia Blanca/patología
16.
N Engl J Med ; 352(10): 987-96, 2005 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-15758009

RESUMEN

BACKGROUND: Glioblastoma, the most common primary brain tumor in adults, is usually rapidly fatal. The current standard of care for newly diagnosed glioblastoma is surgical resection to the extent feasible, followed by adjuvant radiotherapy. In this trial we compared radiotherapy alone with radiotherapy plus temozolomide, given concomitantly with and after radiotherapy, in terms of efficacy and safety. METHODS: Patients with newly diagnosed, histologically confirmed glioblastoma were randomly assigned to receive radiotherapy alone (fractionated focal irradiation in daily fractions of 2 Gy given 5 days per week for 6 weeks, for a total of 60 Gy) or radiotherapy plus continuous daily temozolomide (75 mg per square meter of body-surface area per day, 7 days per week from the first to the last day of radiotherapy), followed by six cycles of adjuvant temozolomide (150 to 200 mg per square meter for 5 days during each 28-day cycle). The primary end point was overall survival. RESULTS: A total of 573 patients from 85 centers underwent randomization. The median age was 56 years, and 84 percent of patients had undergone debulking surgery. At a median follow-up of 28 months, the median survival was 14.6 months with radiotherapy plus temozolomide and 12.1 months with radiotherapy alone. The unadjusted hazard ratio for death in the radiotherapy-plus-temozolomide group was 0.63 (95 percent confidence interval, 0.52 to 0.75; P<0.001 by the log-rank test). The two-year survival rate was 26.5 percent with radiotherapy plus temozolomide and 10.4 percent with radiotherapy alone. Concomitant treatment with radiotherapy plus temozolomide resulted in grade 3 or 4 hematologic toxic effects in 7 percent of patients. CONCLUSIONS: The addition of temozolomide to radiotherapy for newly diagnosed glioblastoma resulted in a clinically meaningful and statistically significant survival benefit with minimal additional toxicity.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Corticoesteroides/uso terapéutico , Adulto , Anciano , Antineoplásicos Alquilantes/efectos adversos , Neoplasias Encefálicas/mortalidad , Quimioterapia Adyuvante , Dacarbazina/efectos adversos , Progresión de la Enfermedad , Femenino , Glioblastoma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Radioterapia Asistida por Computador/efectos adversos , Análisis de Supervivencia , Temozolomida
17.
J Neuropathol Exp Neurol ; 65(4): 305-18, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16691112

RESUMEN

Multiple sclerosis (MS) is a complex disease of unknown etiology. A careful study of the pathology of its component elements in relation to relevant experimental models has helped to understand some of the mechanisms that might be present in the disease. However, the autoimmune nature of the disease has recently come under question and there is a growing recognition of the importance of axonal, cortical, and white matter changes in the genesis and evolution of the lesions, their clinical diagnostic characteristics, and their response to treatment. This review highlights the emerging issues in MS from experimental, imaging and clinical perspectives.


Asunto(s)
Sistema Nervioso Central/patología , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Animales , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Humanos
18.
J Neuropathol Exp Neurol ; 75(2): 156-66, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26802178

RESUMEN

Anatomic distribution and age are variables linked to functions of astrocytes under physiologic and pathologic conditions. We measured the relative expression of a panel of microRNAs (miRNAs) in astrocytes captured by laser micro-dissection from normal human adult white and grey matter, human fetal white matter and germinal matrix samples. Although expression of most miRNAs was comparable between adult and fetal samples, regional differences were observed. In the adult cerebral cortex, expression of miRNAs in morphologically distinct inter-laminar astrocytes underlying the glial limitans differed from those in deeper cortical layers, suggesting functional specialization possibly related to structural stability and defense from potentially harmful factors in the cerebrospinal fluid. Differences between adult white and grey matter miRNA expression included higher expression of pro-inflammatory miRNAs in the former, potentially contributing to differences in inflammation between grey and white matter plaques in multiple sclerosis. Lower expression of miRNAs in fetal versus adult white matter astrocytes likely reflects the immaturity of these migrating cells. Highly expressed miRNAs in the fetal germinal matrix are probably relevant in development and also recapitulate some responses to injury. Future studies can address regional alterations of miRNA expression in pathological conditions.


Asunto(s)
Envejecimiento/metabolismo , Astrocitos/metabolismo , MicroARNs/genética , Adulto , Anciano , Femenino , Feto/metabolismo , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/biosíntesis , Proteína Ácida Fibrilar de la Glía/genética , Sustancia Gris/crecimiento & desarrollo , Sustancia Gris/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Persona de Mediana Edad , Embarazo , Sustancia Blanca/crecimiento & desarrollo , Sustancia Blanca/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA