Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(8): 112816, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37505981

RESUMEN

Glioblastoma (GBM) is known as an intractable, highly heterogeneous tumor encompassing multiple subclones, each supported by a distinct glioblastoma stem cell (GSC). The contribution of GSC genetic and transcriptional heterogeneity to tumor subclonal properties is debated. In this study, we describe the systematic derivation, propagation, and characterization of multiple distinct GSCs from single, treatment-naive GBMs (GSC families). The tumorigenic potential of each GSC better correlates with its transcriptional profile than its genetic make-up, with classical GSCs being inherently more aggressive and mesenchymal more dependent on exogenous growth factors across multiple GBMs. These GSCs can segregate and recapitulate different histopathological aspects of the same GBM, as shown in a paradigmatic tumor with two histopathologically distinct components, including a conventional GBM and a more aggressive primitive neuronal component. This study provides a resource for investigating how GSCs with distinct genetic and/or phenotypic features contribute to individual GBM heterogeneity and malignant escalation.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/patología , Neoplasias Encefálicas/metabolismo , Amplificación de Genes , Células Madre Neoplásicas/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral
2.
Cancer Res ; 83(2): 195-218, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36409826

RESUMEN

Glioblastoma (GBM) is a common and deadly form of brain tumor in adults. Dysregulated metabolism in GBM offers an opportunity to deploy metabolic interventions as precise therapeutic strategies. To identify the molecular drivers and the modalities by which different molecular subgroups of GBM exploit metabolic rewiring to sustain tumor progression, we interrogated the transcriptome, the metabolome, and the glycoproteome of human subgroup-specific GBM sphere-forming cells (GSC). L-fucose abundance and core fucosylation activation were elevated in mesenchymal (MES) compared with proneural GSCs; this pattern was retained in subgroup-specific xenografts and in subgroup-affiliated human patient samples. Genetic and pharmacological inhibition of core fucosylation significantly reduced tumor growth in MES GBM preclinical models. Liquid chromatography-mass spectrometry (LC-MS)-based glycoproteomic screening indicated that most MES-restricted core-fucosylated proteins are involved in therapeutically relevant GBM pathological processes, such as extracellular matrix interaction, cell adhesion, and integrin-mediated signaling. Selective L-fucose accumulation in MES GBMs was observed using preclinical minimally invasive PET, implicating this metabolite as a potential subgroup-restricted biomarker.Overall, these findings indicate that L-fucose pathway activation in MES GBM is a subgroup-specific dependency that could provide diagnostic markers and actionable therapeutic targets. SIGNIFICANCE: Metabolic characterization of subgroup-specific glioblastoma (GBM) sphere-forming cells identifies the L-fucose pathway as a vulnerability restricted to mesenchymal GBM, disclosing a potential precision medicine strategy for targeting cancer metabolism.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/patología , Fucosa/metabolismo , Transducción de Señal , Neoplasias Encefálicas/patología , Células Madre Neoplásicas/patología , Línea Celular Tumoral
3.
Cell ; 185(22): 4153-4169.e19, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36306735

RESUMEN

Genetic studies have highlighted microglia as pivotal in orchestrating Alzheimer's disease (AD). Microglia that adhere to Aß plaques acquire a transcriptional signature, "disease-associated microglia" (DAM), which largely emanates from the TREM2-DAP12 receptor complex that transmits intracellular signals through the protein tyrosine kinase SYK. The human TREM2R47H variant associated with high AD risk fails to activate microglia via SYK. We found that SYK-deficient microglia cannot encase Aß plaques, accelerating brain pathology and behavioral deficits. SYK deficiency impaired the PI3K-AKT-GSK-3ß-mTOR pathway, incapacitating anabolic support required for attaining the DAM profile. However, SYK-deficient microglia proliferated and advanced to an Apoe-expressing prodromal stage of DAM; this pathway relied on the adapter DAP10, which also binds TREM2. Thus, microglial responses to Aß involve non-redundant SYK- and DAP10-pathways. Systemic administration of an antibody against CLEC7A, a receptor that directly activates SYK, rescued microglia activation in mice expressing the TREM2R47H allele, unveiling new options for AD immunotherapy.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Animales , Ratones , Humanos , Microglía/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/patología , Placa Amiloide/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Quinasa Syk/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo
4.
Int J Mol Sci ; 23(16)2022 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-36012705

RESUMEN

Globoid cell leukodystrophy (GLD), or Krabbe disease, is a neurodegenerative sphingolipidosis caused by genetic deficiency of lysosomal ß-galactosylceramidase (GALC), characterized by neuroinflammation and demyelination of the central (CNS) and peripheral nervous system. The acute phase protein long pentraxin-3 (PTX3) is a soluble pattern recognition receptor and a regulator of innate immunity. Growing evidence points to the involvement of PTX3 in neurodegeneration. However, the expression and role of PTX3 in the neurodegenerative/neuroinflammatory processes that characterize GLD remain unexplored. Here, immunohistochemical analysis of brain samples from Krabbe patients showed that macrophages and globoid cells are intensely immunoreactive for PTX3. Accordingly, Ptx3 expression increases throughout the course of the disease in the cerebrum, cerebellum, and spinal cord of GALC-deficient twitcher (Galctwi/twi) mice, an authentic animal model of GLD. This was paralleled by the upregulation of proinflammatory genes and M1-polarized macrophage/microglia markers and of the levels of PTX3 protein in CNS and plasma of twitcher animals. Crossing of Galctwi/twi mice with transgenic PTX3 overexpressing animals (hPTX3 mice) demonstrated that constitutive PTX3 overexpression reduced the severity of clinical signs and the upregulation of proinflammatory genes in the spinal cord of P35 hPTX3/Galctwi/twi mice when compared to Galctwi/twi littermates, leading to a limited increase of their life span. However, this occurred in the absence of a significant impact on the histopathological findings and on the accumulation of the neurotoxic metabolite psychosine when evaluated at this late time point of the disease. In conclusion, our results provide the first evidence that PTX3 is produced in the CNS of GALC-deficient Krabbe patients and twitcher mice. PTX3 may exert a protective role by reducing the neuroinflammatory response that occurs in the spinal cord of GALC-deficient animals.


Asunto(s)
Proteína C-Reactiva , Galactosilceramidasa , Leucodistrofia de Células Globoides , Proteínas del Tejido Nervioso , Animales , Proteína C-Reactiva/genética , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Galactosilceramidasa/deficiencia , Galactosilceramidasa/genética , Humanos , Leucodistrofia de Células Globoides/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Psicosina , Regulación hacia Arriba
5.
JCI Insight ; 6(23)2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34673573

RESUMEN

Medulloblastoma (MB), one of the most malignant brain tumors of childhood, comprises distinct molecular subgroups, with p53 mutant sonic hedgehog-activated (SHH-activated) MB patients having a very severe outcome that is associated with unfavorable histological large cell/anaplastic (LC/A) features. To identify the molecular underpinnings of this phenotype, we analyzed a large cohort of MB developing in p53-deficient Ptch+/- SHH mice that, unexpectedly, showed LC/A traits that correlated with mTORC1 hyperactivation. Mechanistically, mTORC1 hyperactivation was mediated by a decrease in the p53-dependent expression of mTORC1 negative regulator Tsc2. Ectopic mTORC1 activation in mouse MB cancer stem cells (CSCs) promoted the in vivo acquisition of LC/A features and increased malignancy; accordingly, mTORC1 inhibition in p53-mutant Ptch+/- SHH MB and CSC-derived MB resulted in reduced tumor burden and aggressiveness. Most remarkably, mTORC1 hyperactivation was detected only in p53-mutant SHH MB patient samples, and treatment with rapamycin of a human preclinical model phenocopying this subgroup decreased tumor growth and malignancy. Thus, mTORC1 may act as a specific druggable target for this subset of SHH MB, resulting in the implementation of a stringent risk stratification and in the potentially rapid translation of this precision medicine approach into the clinical setting.


Asunto(s)
Proteínas Hedgehog/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Meduloblastoma/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Humanos , Meduloblastoma/patología , Ratones
7.
Nat Med ; 26(1): 131-142, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31932797

RESUMEN

Glia have been implicated in Alzheimer's disease (AD) pathogenesis. Variants of the microglia receptor triggering receptor expressed on myeloid cells 2 (TREM2) increase AD risk, and activation of disease-associated microglia (DAM) is dependent on TREM2 in mouse models of AD. We surveyed gene-expression changes associated with AD pathology and TREM2 in 5XFAD mice and in human AD by single-nucleus RNA sequencing. We confirmed the presence of Trem2-dependent DAM and identified a previously undiscovered Serpina3n+C4b+ reactive oligodendrocyte population in mice. Interestingly, remarkably different glial phenotypes were evident in human AD. Microglia signature was reminiscent of IRF8-driven reactive microglia in peripheral-nerve injury. Oligodendrocyte signatures suggested impaired axonal myelination and metabolic adaptation to neuronal degeneration. Astrocyte profiles indicated weakened metabolic coordination with neurons. Notably, the reactive phenotype of microglia was less evident in TREM2-R47H and TREM2-R62H carriers than in non-carriers, demonstrating a TREM2 requirement in both mouse and human AD, despite the marked species-specific differences.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Núcleo Celular/metabolismo , Núcleo Celular/patología , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Transcriptoma/genética , Anciano , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Axones/patología , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Degeneración Nerviosa/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Transcripción Genética
8.
Proc Natl Acad Sci U S A ; 116(18): 8985-8994, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30988194

RESUMEN

The extrusion of DNA traps contributes to a key mechanism in which innate immune cells clear pathogens or induce sterile inflammation. Here we provide evidence that CD4+ T cells, a critical regulator of adaptive immunity, release extracellular threads of DNA on activation. These DNA extrusions convey autocrine costimulatory signals to T lymphocytes and can be detected in lymph nodes isolated during the priming phase of experimental autoimmune encephalomyelitis (EAE), a CD4+ T cell-driven mouse model of multiple sclerosis. Pharmacologic inhibition of mitochondrial reactive oxygen species (mtROS) abolishes the extrusion of DNA by CD4+ T cells, reducing cytokine production in vitro and T cell priming against myelin in vivo. Moreover, mtROS blockade during established EAE markedly ameliorates disease severity, dampening autoimmune inflammation of the central nervous system. Taken together, these experimental results elucidate a mechanism of intrinsic immune costimulation mediated by DNA threads released by activated T helper cells, and identify a potential therapeutic target for such disorders as multiple sclerosis, neuromyelitis optica, and CD4+ T cell-mediated disorders.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Ácidos Nucleicos Libres de Células/genética , ADN/genética , Animales , Comunicación Autocrina/genética , Linfocitos T CD8-positivos/inmunología , Ácidos Nucleicos Libres de Células/metabolismo , Sistema Nervioso Central/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Vaina de Mielina , Glicoproteína Mielina-Oligodendrócito
9.
Cell Death Differ ; 26(9): 1813-1831, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30538287

RESUMEN

Achaete-scute homolog 1 gene (ASCL1) is a gene classifier for the proneural (PN) transcriptional subgroup of glioblastoma (GBM) that has a relevant role in the neuronal-like differentiation of GBM cancer stem cells (CSCs) through the activation of a PN gene signature. Besides prototypical ASCL1 PN target genes, the molecular effectors mediating ASCL1 function in regulating GBM differentiation and, most relevantly, subgroup specification are currently unknown. Here we report that ASCL1 not only promotes the acquisition of a PN phenotype in CSCs by inducing a glial-to-neuronal lineage switch but also concomitantly represses mesenchymal (MES) features by directly downregulating the expression of N-Myc downstream-regulated gene 1 (NDRG1), which we propose as a novel gene classifier of MES GBMs. Increasing the expression of ASCL1 in PN CSCs results in suppression of self-renewal, promotion of differentiation and, most significantly, decrease in tumorigenesis, which is also reproduced by NDRG1 silencing. Conversely, both abrogation of ASCL1 expression in PN CSCs and enforcement of NDRG1 expression in either PN or MES CSCs induce proneural-to-mesenchymal transition (PMT) and enhanced mesenchymal features. Surprisingly, ASCL1 overexpression in MES CSCs increases malignant features and gives rise to a neuroendocrine-like secretory phenotype. Altogether, our results propose that the fine interplay between ASCL1 and its target NDRG1 might serve as potential subgroup-specific targetable vulnerability in GBM; enhancing ASCL1 expression in PN GBMs might reduce tumorigenesis, whereas repressing NDRG1 expression might be actionable to hamper the malignancy of GBM belonging to the MES subgroup.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Glioblastoma/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Autorrenovación de las Células/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neuronas/metabolismo , Neuronas/patología , Transducción de Señal
10.
J Clin Invest ; 128(4): 1688-1706, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29389670

RESUMEN

Tuberous sclerosis complex (TSC) is a dominantly inherited disease caused by hyperactivation of the mTORC1 pathway and characterized by the development of hamartomas and benign tumors, including in the brain. Among the neurological manifestations associated with TSC, the tumor progression of static subependymal nodules (SENs) into subependymal giant cell astrocytomas (SEGAs) is one of the major causes of morbidity and shortened life expectancy. To date, mouse modeling has failed in reproducing these 2 lesions. Here we report that simultaneous hyperactivation of mTORC1 and Akt pathways by codeletion of Tsc1 and Pten, selectively in postnatal neural stem cells (pNSCs), is required for the formation of bona fide SENs and SEGAs. Notably, both lesions closely recapitulate the pathognomonic morphological and molecular features of the corresponding human abnormalities. The establishment of long-term expanding pNSC lines from mouse SENs and SEGAs made possible the identification of mTORC2 as one of the mediators conferring tumorigenic potential to SEGA pNSCs. Notably, in spite of concurrent Akt hyperactivation in mouse brain lesions, single mTOR inhibition by rapamycin was sufficient to strongly impair mouse SEGA growth. This study provides evidence that, concomitant with mTORC1 hyperactivation, sustained activation of Akt and mTORC2 in pNSCs is a mandatory step for the induction of SENs and SEGAs, and, at the same time, makes available an unprecedented NSC-based in vivo/in vitro model to be exploited for identifying actionable targets in TSC.


Asunto(s)
Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Astrocitoma/genética , Astrocitoma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Transgénicos , Células-Madre Neurales/patología , Proteínas Proto-Oncogénicas c-akt/genética , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/patología
11.
Front Immunol ; 8: 490, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28512459

RESUMEN

Wiskott-Aldrich syndrome (WAS) is a primary immunodeficiency caused by mutations in the gene encoding the hematopoietic-specific WAS protein (WASp). WAS is frequently associated with autoimmunity, indicating a critical role of WASp in maintenance of tolerance. The role of B cells in the induction of autoreactive immune responses in WAS has been investigated in several settings, but the mechanisms leading to the development of autoimmune manifestations have been difficult to evaluate in the mouse models of the disease that do not spontaneously develop autoimmunity. We performed an extensive characterization of Was-/- mice that provided evidence of the potential alteration in B cell selection, because of the presence of autoantibodies against double-stranded DNA, platelets, and tissue antigens. To uncover the mechanisms leading to the activation of the potentially autoreactive B cells in Was-/- mice, we performed in vivo chronic stimulations with toll-like receptors agonists (LPS and CpG) and apoptotic cells or infection with lymphocytic choriomeningitis virus. All treatments led to increased production of autoantibodies, increased proteinuria, and kidney tissue damage in Was-/- mice. These findings demonstrate that a lower clearance of pathogens and/or self-antigens and the resulting chronic inflammatory state could cause B cell tolerance breakdown leading to autoimmunity in WAS.

12.
Sci Rep ; 7: 40136, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28074903

RESUMEN

Adenosine Deaminase (ADA) deficiency is an autosomal recessive variant of severe combined immunodeficiency (SCID) caused by systemic accumulation of ADA substrates. Neurological and behavioral abnormalities observed in ADA-SCID patients surviving after stem cell transplantation or gene therapy represent an unresolved enigma in the field. We found significant neurological and cognitive alterations in untreated ADA-SCID patients as well as in two groups of patients after short- and long-term enzyme replacement therapy with PEG-ADA. These included motor dysfunction, EEG alterations, sensorineural hypoacusia, white matter and ventricular alterations in MRI as well as a low mental development index or IQ. Ada-deficient mice were significantly less active and showed anxiety-like behavior. Molecular and metabolic analyses showed that this phenotype coincides with metabolic alterations and aberrant adenosine receptor signaling. PEG-ADA treatment corrected metabolic adenosine-based alterations, but not cellular and signaling defects, indicating an intrinsic nature of the neurological and behavioral phenotype in ADA deficiency.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina/metabolismo , Encéfalo/metabolismo , Enfermedades del Sistema Nervioso/fisiopatología , Animales , Conducta , Conducta Animal , Humanos , Ratones , Enfermedades del Sistema Nervioso/patología
13.
EMBO Mol Med ; 8(5): 550-68, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27138567

RESUMEN

Glioblastoma (GBM) contains stem-like cells (GSCs) known to be resistant to ionizing radiation and thus responsible for therapeutic failure and rapidly lethal tumor recurrence. It is known that GSC radioresistance relies on efficient activation of the DNA damage response, but the mechanisms linking this response with the stem status are still unclear. Here, we show that the MET receptor kinase, a functional marker of GSCs, is specifically expressed in a subset of radioresistant GSCs and overexpressed in human GBM recurring after radiotherapy. We elucidate that MET promotes GSC radioresistance through a novel mechanism, relying on AKT activity and leading to (i) sustained activation of Aurora kinase A, ATM kinase, and the downstream effectors of DNA repair, and (ii) phosphorylation and cytoplasmic retention of p21, which is associated with anti-apoptotic functions. We show that MET pharmacological inhibition causes DNA damage accumulation in irradiated GSCs and their depletion in vitro and in GBMs generated by GSC xenotransplantation. Preclinical evidence is thus provided that MET inhibitors can radiosensitize tumors and convert GSC-positive selection, induced by radiotherapy, into GSC eradication.


Asunto(s)
Glioblastoma/radioterapia , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Células Madre/fisiología , Células Madre/efectos de la radiación , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Aurora Quinasa A/metabolismo , Supervivencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Reparación del ADN , Xenoinjertos , Humanos , Ratones , Proteína Oncogénica v-akt/metabolismo
14.
Sci Rep ; 5: 15454, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26482194

RESUMEN

Multiple primary malignant neoplasms are rare entities in the clinical setting, but represent an important issue in the clinical management of patients since they could be expression of a genetic predisposition to malignancy. A high resolution genome wide array CGH led us to identify the first case of a de novo constitutional deletion confined to the FBXW7 gene, a well known tumor suppressor, in a patient with a syndromic phenotype characterized by focal segmental glomerulosclerosis and multiple primary early/atypical onset tumors, including Hodgkin's lymphoma, Wilms tumor and breast cancer. Other genetic defects may be associated with patient's phenotype. In this light, constitutional mutations at BRCA1, BRCA2, TP53, PALB2 and WT1 genes were excluded by performing sequencing and MLPA analysis; similarly, we ruled out constitutional abnormalities at the imprinted 11p15 region by methylation specific -MLPA assay. Our observations sustain the role of FBXW7 as cancer predisposition gene and expand the spectrum of its possible associated diseases.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Eliminación de Gen , Glomeruloesclerosis Focal y Segmentaria/genética , Neoplasias Primarias Múltiples/genética , Ubiquitina-Proteína Ligasas/genética , Adulto , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Hibridación Genómica Comparativa , Análisis Mutacional de ADN , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Humanos , Hibridación Fluorescente in Situ , Cariotipo , Masculino , Persona de Mediana Edad , Neoplasias Primarias Múltiples/diagnóstico , Neoplasias Primarias Múltiples/metabolismo
15.
Lab Invest ; 95(6): 585-602, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25822667

RESUMEN

Rhabdomyosarcoma (RMS) is a childhood soft tissue tumor with broad expression of markers that are typically found in skeletal muscle. Cavin-1 is a recently discovered protein actively cooperating with Caveolin-1 (Cav-1) in the morphogenesis of caveolae and whose role in cancer is drawing increasing attention. Using a combined in silico and in vitro analysis here we show that Cavin-1 is expressed in myogenic RMS tumors as well as in human and primary mouse RMS cultures, exhibiting a broad subcellular localization, ranging from nuclei and cytosol to plasma membrane. In particular, the coexpression and plasma membrane interaction between Cavin-1 and Cav-1 characterized the proliferation of human and mouse RMS cell cultures, while a downregulation of their expression levels was observed during the myogenic differentiation. Knockdown of Cavin-1 or Cav-1 in the human RD and RH30 cells led to impairment of cell proliferation and migration. Moreover, loss of Cavin-1 in RD cells impaired the anchorage-independent cell growth in soft agar. While the loss of Cavin-1 did not affect the Cav-1 protein levels in RMS cells, Cav-1 overexpression and knockdown triggered a rise or depletion of Cavin-1 protein levels in RD cells, respectively, in turn reflecting on increased or decreased cell proliferation, migration and anchorage-independent cell growth. Collectively, these data indicate that the interaction between Cavin-1 and Cav-1 underlies the cell growth and migration in myogenic tumors.


Asunto(s)
Caveolina 1/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Rabdomiosarcoma/metabolismo , Animales , Caveolina 1/genética , Diferenciación Celular , Línea Celular Tumoral , Células Cultivadas , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Ratones , Proteínas de Unión al ARN/genética , Células Satélite del Músculo Esquelético/metabolismo
16.
Stem Cells ; 33(5): 1377-89, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25639612

RESUMEN

microRNAs (miRNAs) are short noncoding RNAs, which regulate gene expression post-transcriptionally and play crucial roles in relevant biological and pathological processes. Here, we investigated the putative role of miRNAs in modulating the tumor-initiating potential of mouse medulloblastoma (MB)-derived cancer stem cells (CSCs). We first subjected bona fide highly tumorigenic (HT) CSCs as well as lowly tumorigenic MB CSCs and normal neural stem cells to miRNA profiling, which identified a HT CSC-specific miRNA signature. Next, by cross-checking CSC mRNA/miRNA profiles, we pinpointed miR-135a as a potential tumor suppressor gene, which was strongly downregulated in HT CSCs as well as in the highly malignant experimental tumors derived from them. Remarkably, enforced expression of miR-135a in HT CSCs strongly inhibited tumorigenesis by repressing the miR-135a direct target gene Arhgef6. Considering the upregulation of Arhgef6 in human MBs and its involvement in mediating experimental medulloblastomagenesis, its efficient suppression by miR-135a might make available an effective therapeutic strategy to selectively impair the tumorigenic potential of MB CSCs. Stem Cells 2015;33:1377-1389.


Asunto(s)
Carcinogénesis/patología , Meduloblastoma/patología , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Animales , Carcinogénesis/genética , Agregación Celular , Transformación Celular Neoplásica/genética , Regulación hacia Abajo , Perfilación de la Expresión Génica , Meduloblastoma/genética , Ratones Endogámicos C57BL , MicroARNs/genética , Células Madre Neoplásicas/patología , Células-Madre Neurales/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
18.
PLoS One ; 9(1): e86942, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466296

RESUMEN

Wiskott-Aldrich Syndrome protein (WASP) is a key regulator of the actin cytoskeleton in hematopoietic cells. Defective expression of WASP leads to multiple abnormalities in different hematopoietic cells. Despite severe impairment of T cell function, WAS patients exhibit a high prevalence of autoimmune disorders. We attempted to induce EAE, an animal model of organ-specific autoimmunity affecting the CNS that mimics human MS, in Was(-/-) mice. We describe here that Was(-/-) mice are markedly resistant against EAE, showing lower incidence and milder score, reduced CNS inflammation and demyelination as compared to WT mice. Microglia was only poorly activated in Was(-/-) mice. Antigen-induced T-cell proliferation, Th-1 and -17 cytokine production and integrin-dependent adhesion were increased in Was(-/-) mice. However, adoptive transfer of MOG-activated T cells from Was(-/-) mice in WT mice failed to induce EAE. Was(-/-) mice were resistant against EAE also when induced by adoptive transfer of MOG-activated T cells from WT mice. Was(+/-) heterozygous mice developed an intermediate clinical phenotype between WT and Was(-/-) mice, and they displayed a mixed population of WASP-positive and -negative T cells in the periphery but not in their CNS parenchyma, where the large majority of inflammatory cells expressed WASP. In conclusion, in absence of WASP, T-cell responses against a CNS autoantigen are increased, but the ability of autoreactive T cells to induce CNS autoimmunity is impaired, most probably because of an inefficient T-cell transmigration into the CNS and defective CNS resident microglial function.


Asunto(s)
Autoinmunidad/inmunología , Movimiento Celular , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Activación de Linfocitos/inmunología , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Animales , Western Blotting , Adhesión Celular , Proliferación Celular , Células Cultivadas , Sistema Nervioso Central/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Humanos , Técnicas para Inmunoenzimas , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía , Vaina de Mielina
19.
J Allergy Clin Immunol ; 133(4): 1116-23, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24332219

RESUMEN

BACKGROUND: Recombination-activating gene 1 (RAG1) deficiency results in severe combined immunodeficiency (SCID) caused by a complete lack of T and B lymphocytes. If untreated, patients succumb to recurrent infections. OBJECTIVES: We sought to develop lentiviral gene therapy for RAG1-induced SCID and to test its safety. METHODS: Constructs containing the viral spleen-focus-forming virus (SF), ubiquitous promoters, or cell type-restricted promoters driving sequence-optimized RAG1 were compared for efficacy and safety in sublethally preconditioned Rag1(-/-) mice undergoing transplantation with transduced bone marrow progenitors. RESULTS: Peripheral blood CD3(+) T-cell reconstitution was achieved with SF, ubiquitous promoters, and cell type-restricted promoters but 3- to 18-fold lower than that seen in wild-type mice, and with a compromised CD4(+)/CD8(+) ratio. Mitogen-mediated T-cell responses and T cell-dependent and T cell-independent B-cell responses were not restored, and T-cell receptor patterns were skewed. Reconstitution of mature peripheral blood B cells was approximately 20-fold less for the SF vector than in wild-type mice and often not detectable with the other promoters, and plasma immunoglobulin levels were abnormal. Two months after transplantation, gene therapy-treated mice had rashes with cellular tissue infiltrates, activated peripheral blood CD44(+)CD69(+) T cells, high plasma IgE levels, antibodies against double-stranded DNA, and increased B cell-activating factor levels. Only rather high SF vector copy numbers could boost T- and B-cell reconstitution, but mRNA expression levels during T- and B-cell progenitor stages consistently remained less than wild-type levels. CONCLUSIONS: These results underline that further development is required for improved expression to successfully treat patients with RAG1-induced SCID while maintaining low vector copy numbers and minimizing potential risks, including autoimmune reactions resembling Omenn syndrome.


Asunto(s)
Terapia Genética , Vectores Genéticos/genética , Proteínas de Homeodominio/genética , Lentivirus/genética , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia , Animales , Autoinmunidad/genética , Células de la Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Femenino , Dosificación de Gen , Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Masculino , Ratones , Ratones Noqueados , Fenotipo , Inmunodeficiencia Combinada Grave/inmunología , Bazo/inmunología , Linfocitos T/metabolismo , Timo/inmunología , Transducción Genética , Quimera por Trasplante
20.
Brain ; 136(Pt 9): 2859-75, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23983033

RESUMEN

Globoid cell leukodystrophy (Krabbe disease) is a neurological disorder of infants caused by genetic deficiency of the lysosomal enzyme ß-galactosylceramidase leading to accumulation of the neurotoxic metabolite 1-ß-d-galactosylsphingosine (psychosine) in the central nervous system. Angiogenesis plays a pivotal role in the physiology and pathology of the brain. Here, we demonstrate that psychosine has anti-angiogenic properties by causing the disassembling of endothelial cell actin structures at micromolar concentrations as found in the brain of patients with globoid cell leukodystrophy. Accordingly, significant alterations of microvascular endothelium were observed in the post-natal brain of twitcher mice, an authentic model of globoid cell leukodystrophy. Also, twitcher endothelium showed a progressively reduced capacity to respond to pro-angiogenic factors, defect that was corrected after transduction with a lentiviral vector harbouring the murine ß-galactosylceramidase complementary DNA. Finally, RNA interference-mediated ß-galactosylceramidase gene silencing causes psychosine accumulation in human endothelial cells and hampers their mitogenic and motogenic response to vascular endothelial growth factor. Accordingly, significant alterations were observed in human microvasculature from brain biopsy of a globoid cell leukodystrophy case. Together these data demonstrate that ß-galactosylceramidase deficiency induces significant alterations in endothelial neovascular responses that may contribute to central nervous system and systemic damages that occur in globoid cell leukodystrophy.


Asunto(s)
Leucodistrofia de Células Globoides/complicaciones , Neovascularización Patológica/etiología , Neovascularización Patológica/patología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Aorta/patología , Aorta/ultraestructura , Materiales Biocompatibles , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/ultraestructura , Bovinos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Colágeno/toxicidad , Modelos Animales de Enfermedad , Combinación de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Laminina/toxicidad , Leucodistrofia de Células Globoides/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Neovascularización Patológica/prevención & control , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteoglicanos/toxicidad , Psicosina/metabolismo , Psicosina/farmacología , ARN Interferente Pequeño/administración & dosificación , Factores de Tiempo , Transfección , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de la Zonula Occludens-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...