Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(21): eadj8769, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38787942

RESUMEN

Circular RNAs (circRNAs) are a large class of noncoding RNAs. Despite the identification of thousands of circular transcripts, the biological significance of most of them remains unexplored, partly because of the lack of effective methods for generating loss-of-function animal models. In this study, we focused on circTulp4, an abundant circRNA derived from the Tulp4 gene that is enriched in the brain and synaptic compartments. By creating a circTulp4-deficient mouse model, in which we mutated the splice acceptor site responsible for generating circTulp4 without affecting the linear mRNA or protein levels, we were able to conduct a comprehensive phenotypic analysis. Our results demonstrate that circTulp4 is critical in regulating neuronal and brain physiology, modulating the strength of excitatory neurotransmission and sensitivity to aversive stimuli. This study provides evidence that circRNAs can regulate biologically relevant functions in neurons, with modulatory effects at multiple levels of the phenotype, establishing a proof of principle for the regulatory role of circRNAs in neural processes.


Asunto(s)
Encéfalo , ARN Circular , Transmisión Sináptica , ARN Circular/genética , Animales , Ratones , Encéfalo/metabolismo , Encéfalo/fisiología , Ratones Noqueados , Neuronas/metabolismo , Neuronas/fisiología
2.
Clin Sci (Lond) ; 133(2): 351-365, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30626728

RESUMEN

Obesity, metabolic syndrome, and type 2 diabetes, three strongly interrelated diseases, are associated to increased morbidity and mortality worldwide. The pathogenesis of obesity-associated disorders is still under study. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycoprotein expressed in many cell types including adipocytes, parenchymal, and non-parenchymal hepatic cells and pancreatic cells. Studies have demonstrated that SPARC inhibits adipogenesis and promotes insulin resistance; in addition, circulating SPARC levels were positively correlated with body mass index in obese individuals. Therefore, SPARC is being proposed as a key factor in the pathogenesis of obesity-associated disorders. The aim of this study is to elucidate the role of SPARC in glucose homeostasis. We show here that SPARC null (SPARC-/-) mice displayed an abnormal insulin-regulated glucose metabolism. SPARC-/- mice presented an increased adipose tissue deposition and an impaired glucose homeostasis as animals aged. In addition, the absence of SPARC worsens high-fat diet-induced diabetes in mice. Interestingly, although SPARC-/- mice on high-fat diet were sensitive to insulin they showed an impaired insulin secretion capacity. Of note, the expression of glucose transporter 2 in islets of SPARC-/- mice was dramatically reduced. The present study provides the first evidence that deleted SPARC expression causes diabetes in mice. Thus, SPARC deficient mice constitute a valuable model for studies concerning obesity and its related metabolic complications, including diabetes.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Insulina/sangre , Islotes Pancreáticos/metabolismo , Osteonectina/metabolismo , Envejecimiento/sangre , Animales , Biomarcadores/sangre , Diabetes Mellitus Experimental/genética , Dieta Alta en Grasa , Sacarosa en la Dieta , Transportador de Glucosa de Tipo 2/metabolismo , Homeostasis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Osteonectina/deficiencia , Osteonectina/genética , Vías Secretoras
3.
Stem Cell Res Ther ; 8(1): 227, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-29041955

RESUMEN

Multilineage differentiating stress enduring (Muse) cells, discovered in the spring of 2010 at Tohoku University in Sendai, Japan, were quickly recognized by scientists as a possible source of pluripotent cells naturally present within mesenchymal tissues. Muse cells normally exist in a quiescent state, singularly activated by severe cellular stress in vitro and in vivo. Muse cells have the capacity for self-renewal while maintaining pluripotent cell characteristics indicated by the expression of pluripotent stem cell markers. Muse cells differentiate into cells representative of all three germ cell layers both spontaneously and under media-specific induction. In contrast to embryonic stem and induced pluripotent stem cells, Muse cells exhibit low telomerase activity, a normal karyotype, and do not undergo tumorigenesis once implanted in SCID mice. Muse cells efficiently home into damaged tissues and differentiate into specific cells leading to tissue regeneration and functional recovery as described in different animal disease models (i.e., fulminant hepatitis, muscle degeneration, skin ulcers, liver cirrhosis, cerebral stroke, vitiligo, and focal segmental glomerulosclerosis). Circulating Muse cells have been detected in peripheral blood, with higher levels present in stroke patients during the acute phase. Furthermore, Muse cells have inherent immunomodulatory properties, which could contribute to tissue generation and functional repair in vivo. Genetic studies in Muse cells indicate a highly conserved cellular mechanism as seen in more primitive organisms (yeast, Saccharomyces cerevisiae, Caenorhabditis elegans, chlamydomonas, Torpedo californica, drosophila, etc.) in response to cellular stress and acute injury. This review details the molecular and cellular properties of Muse cells as well as their capacity for tissue repair and functional recovery, highlighting their potential for clinical application in regenerative medicine.


Asunto(s)
Células Madre Adultas/citología , Células Madre Pluripotentes/citología , Estrés Fisiológico , Células Madre Adultas/metabolismo , Células Madre Adultas/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Humanos , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/fisiología
4.
Stem Cells Transl Med ; 6(1): 161-173, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28170177

RESUMEN

Adult mesenchymal stromal cell-based interventions have shown promising results in a broad range of diseases. However, their use has faced limited effectiveness owing to the low survival rates and susceptibility to environmental stress on transplantation. We describe the cellular and molecular characteristics of multilineage-differentiating stress-enduring (Muse) cells derived from adipose tissue (AT), a subpopulation of pluripotent stem cells isolated from human lipoaspirates. Muse-AT cells were efficiently obtained using a simple, fast, and affordable procedure, avoiding cell sorting and genetic manipulation methods. Muse-AT cells isolated under severe cellular stress, expressed pluripotency stem cell markers and spontaneously differentiated into the three germ lineages. Muse-AT cells grown as spheroids have a limited proliferation rate, a diameter of ∼15 µm, and ultrastructural organization similar to that of embryonic stem cells. Muse-AT cells evidenced high stage-specific embryonic antigen-3 (SSEA-3) expression (∼60% of cells) after 7-10 days growing in suspension and did not form teratomas when injected into immunodeficient mice. SSEA-3+ -Muse-AT cells expressed CD105, CD29, CD73, human leukocyte antigen (HLA) class I, CD44, and CD90 and low levels of HLA class II, CD45, and CD34. Using lipopolysaccharide-stimulated macrophages and antigen-challenged T-cell assays, we have shown that Muse-AT cells have anti-inflammatory activities downregulating the secretion of proinflammatory cytokines, such as interferon-γ and tumor necrosis factor-α. Muse-AT cells spontaneously gained transforming growth factor-ß1 expression that, in a phosphorylated SMAD2-dependent manner, might prove pivotal in their observed immunoregulatory activity through decreased expression of T-box transcription factor in T cells. Collectively, the present study has demonstrated the feasibility and efficiency of obtaining Muse-AT cells that can potentially be harnessed as immunoregulators to treat immune-related disorders. Stem Cells Translational Medicine 2017;6:161-173.


Asunto(s)
Tejido Adiposo/patología , Carcinogénesis/patología , Inmunomodulación , Células Madre Pluripotentes/citología , Factor de Crecimiento Transformador beta1/farmacología , Animales , Biomarcadores/metabolismo , Carcinogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Estratos Germinativos/citología , Humanos , Inmunomodulación/efectos de los fármacos , Cariotipo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Células Madre Pluripotentes/trasplante , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Bazo/citología , Estrés Fisiológico , Teratoma/patología
5.
Sci Rep ; 6: 36646, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27857212

RESUMEN

Dendritic cells (DC) initiate the adaptive immune response. Glucocorticoids (GCs) down-modulate the function of DC. Compound A (CpdA, (2-(4-acetoxyphenyl)-2-chloro-N-methyl-ethylammonium chloride) is a plant-derived GR-ligand with marked dissociative properties. We investigated the effects of CpdA on in vitro generated GM-CSF-conditioned bone marrow-derived DC (BMDC). CpdA-exposed BMDC exhibited low expression of cell-surface molecules and diminution of the release of proinflammatory cytokines upon LPS stimulation; processes associated with BMDC maturation and activation. CpdA-treated BMDC were inefficient at Ag capture via mannose receptor-mediated endocytosis and displayed reduced T-cell priming. CpdA prevented the LPS-induced rise in pErk1/2 and pP38, kinases involved in TLR4 signaling. CpdA fully inhibited LPS-induced pAktSer473, a marker associated with the generation of tolerogenic DC. We used pharmacological blockade and selective genetic loss-of-function tools and demonstrated GR-independent inhibitory effects of CpdA in BMDC. Mechanistically, CpdA-mediated inactivation of the NF-κB intracellular signaling pathway was associated with a short-circuiting of pErk1/2 and pP38 upstream signaling. Assessment of the in vivo function of CpdA-treated BMDC pulsed with the hapten trinitrobenzenesulfonic acid showed impaired cell-mediated contact hypersensitivity. Collectively, we provide evidence that CpdA is an effective BMDC modulator that might have a benefit for immune disorders, even when GR is not directly targeted.


Asunto(s)
Acetatos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Regulación hacia Abajo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Tiramina/análogos & derivados , Animales , Antígeno B7-1/metabolismo , Células de la Médula Ósea/citología , Células Dendríticas/citología , Endocitosis/efectos de los fármacos , Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Ratones , Receptor Toll-Like 4/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Tiramina/farmacología , Regulación hacia Arriba/efectos de los fármacos
6.
PLoS One ; 9(6): e100369, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24963634

RESUMEN

The conversion of differentiated cells into insulin-producing cells is a promising approach for the autologous replacement of pancreatic cells in patients with type 1 diabetes (T1D). At present, cellular reprogramming strategies encompass ethical problems, epigenetic failure or teratoma formation, which has prompted the development of new approaches. Here, we report a novel technique for the conversion of skin fibroblasts from T1D patients into insulin-expressing clusters using only drug-based induction. Our results demonstrate that skin fibroblasts from diabetic patients have pancreatic differentiation capacities and avoid the necessity of using transgenic strategies, stem cell sources or global demethylation steps. These findings open new possibilities for studying diabetes mechanisms, drug screenings and ultimately autologous transgenic-free regenerative medicine therapies in patients with T1D.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/patología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Células Secretoras de Insulina/citología , Piel/citología , Adolescente , Animales , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Niño , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Hormonas/metabolismo , Humanos , Hiperglucemia/patología , Hiperglucemia/prevención & control , Masculino , Ratones , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Trasplantes
7.
Expert Opin Biol Ther ; 14(7): 917-29, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24745973

RESUMEN

INTRODUCTION: Embryonic stem cells and induced pluripotent stem cells have emerged as the gold standard of pluripotent stem cells and the class of stem cell with the highest potential for contribution to regenerative and therapeutic application; however, their translational use is often impeded by teratoma formation, commonly associated with pluripotency. We discuss a population of nontumorigenic pluripotent stem cells, termed Multilineage Differentiating Stress Enduring (Muse) cells, which offer an innovative and exciting avenue of exploration for the potential treatment of various human diseases. AREAS COVERED: This review discusses the origin of Muse cells, describes in detail their various unique characteristics, and considers future avenues of their application and investigation with respect to what is currently known of adult pluripotent stem cells in scientific literature. We begin by defining cell potency, then discuss both mesenchymal and various reported populations of pluripotent stem cells, and finally delve into Muse cells and the characteristics that set them apart from their contemporaries. EXPERT OPINION: Muse cells derived from adipose tissue (Muse-AT) are efficiently, routinely and painlessly isolated from human lipoaspirate material, exhibit tripoblastic differentiation both spontaneously and under media-specific induction, and do not form teratomas. We describe qualities specific to Muse-AT cells and their potential impact on the field of regenerative medicine and cell therapy.


Asunto(s)
Tejido Adiposo/citología , Carcinogénesis , Diferenciación Celular , Proliferación Celular , Células Madre Pluripotentes/citología , Adulto , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes/trasplante , Medicina Regenerativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...