Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Reprod Biomed Online ; 43(4): 587-597, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34474974

RESUMEN

Isolation and characterization of presumptive human adult ovarian stem cells (OSC) has broken the long standing dogma of the absence of postnatal neo-oogenesis. Human adult OSC have been immunosorted by antibodies reacting against the RNA helicase VASA and have been reported to engraft into appropriate stem cell niches to promote neo-oogenesis. Analysis of published research, however, questions some of the findings on isolation, characterization, in-vitro self-renewal and clinical safety of the presumptive human adult OSC. In the present study, human VASApos embryo-fetal primordial germ cells and presumptive adult OSC are shown to share several pluripotency and early germ cell markers not ascertained in the initial characterization of adult OSC. A new hypothesis is made that the restoration of fertility claimed to result from presumptive human adult OSC may be attributed instead to VASApos embryo-fetal primordial germ cell remnants in the adult ovary, or alternatively to earlier VASAneg germ cells generated by in-vitro de-differentiation of the presumptive OSC. The suggested hypotheses have extensive implications for the practice and safety of adult OSC in the development of new treatments aimed at rescuing the ovarian reserve.


Asunto(s)
Células Madre Adultas/enzimología , ARN Helicasas DEAD-box/metabolismo , Células Germinativas/enzimología , Infertilidad Femenina/terapia , Ovario/citología , Trasplante de Células Madre , Animales , Biomarcadores/metabolismo , Separación Celular , Técnicas de Cultivo , Embrión de Mamíferos/citología , Femenino , Humanos
2.
J Reprod Immunol ; 144: 103252, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33549903

RESUMEN

OBJECTIVES: Uncontrolled TH17 differentiation has been suggested to play a role in the pathogenesis of pregnancy loss. We recently showed that menstrual blood stromal/stem cells (MenSCs) alter functional features of natural killer cells. Here, we hypothesized that MenSCs could modulate differentiation of TH17 cells. METHOD: MenSCs were collected from 18 apparently healthy women and characterized. Bone marrow mesenchymal stem cells (BMSCs) served as a control. TH17 polarization and proliferation of purified T CD4+ cells were assessed by flow cytometry in a well-defined co-culture system containing T CD4+ cells and MenSCs or BMSCs. Indoleamine 2,3-Dioxygenase (IDO) activity was evaluated in MenSC and BMSC culture supernatants by a colorimetric assay. The impact of MenSCs on expression of transcription factors, RORC, T-bet, Gata3, NRP-1 and Helios were studied by qPCR. RESULTS: MenSCs significantly inhibited TH17 differentiation (p = 0.0383) and percentage of the cells co-expressing IL-17 and IFN-γ (p = 0.0023). PGE2 blockade significantly reduced percentage and proliferation of T CD4+IL-17+ (p = 0.003, p = 0.0018), T CD4+ IFN-γ+ (p = 0.002, p = 0.0022) and T CD4+IL-17+ IFN-γ+ (p = 0.004, p = 0.02) cells. MenSCs produced a considerable activity of IDO (p = 0.0002), induced a significant rise in the Treg frequency (p = 0.0091) and a sharp increase in TH17/Tregs ratio (p = 0.0022). MenSCs increased expression of NRP1 (p = 0.001), while downregulated expression of RORC in T cells (p = 0.001). CONCLUSION: Our results suggest a supportive role for MenSCs in establishing a pregnancy-friendly microenvironment in the uterus and put forth the idea that inherent abnormalities of MenSCs may be a basis for dysregulated endometrial immune network leading to pregnancy loss.


Asunto(s)
Células Madre Adultas/inmunología , Menstruación/sangre , Embarazo/inmunología , Células del Estroma/inmunología , Células Th17/inmunología , Adulto , Células Madre Adultas/enzimología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Endometrio/citología , Endometrio/inmunología , Femenino , Voluntarios Sanos , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/análisis , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Células del Estroma/enzimología
3.
Cells ; 10(2)2021 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-33498808

RESUMEN

In adult stem cells, Glycogen Synthase Kinase 3ß (GSK3ß) is at the crossroad of signaling pathways controlling survival, proliferation, adhesion and differentiation. The microenvironment plays a key role in the regulation of these cell functions and we have demonstrated that the GSK3ß activity is strongly dependent on the engagement of integrins and protease-activated receptors (PARs). Downstream of the integrin α5ß1 or PAR2 activation, a molecular complex is organized around the scaffolding proteins RACK1 and ß-arrestin-2 respectively, containing the phosphatase PP2A responsible for GSK3ß activation. As a consequence, a quiescent stem cell phenotype is established with high capacities to face apoptotic and metabolic stresses. A protective role of GSK3ß has been found for hematopoietic and intestinal stem cells. Latters survived to de-adhesion through PAR2 activation, whereas formers were protected from cytotoxicity through α5ß1 engagement. However, a prolonged activation of GSK3ß promoted a defect in epithelial regeneration and a resistance to chemotherapy of leukemic cells, paving the way to chronic inflammatory diseases and to cancer resurgence, respectively. In both cases, a sexual dimorphism was measured in GSK3ß-dependent cellular functions. GSK3ß activity is a key marker for inflammatory and cancer diseases allowing adjusted therapy to sex, age and metabolic status of patients.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/enzimología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Integrinas/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Medicina de Precisión , Nicho de Células Madre
4.
Cell Stem Cell ; 27(6): 890-904.e8, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33128895

RESUMEN

Coronavirus infection causes diffuse alveolar damage leading to acute respiratory distress syndrome. The absence of ex vivo models of human alveolar epithelium is hindering an understanding of coronavirus disease 2019 (COVID-19) pathogenesis. Here, we report a feeder-free, scalable, chemically defined, and modular alveolosphere culture system for the propagation and differentiation of human alveolar type 2 cells/pneumocytes derived from primary lung tissue. Cultured pneumocytes express the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor angiotensin-converting enzyme receptor type-2 (ACE2) and can be infected with virus. Transcriptome and histological analysis of infected alveolospheres mirror features of COVID-19 lungs, including emergence of interferon (IFN)-mediated inflammatory responses, loss of surfactant proteins, and apoptosis. Treatment of alveolospheres with IFNs recapitulates features of virus infection, including cell death. In contrast, alveolospheres pretreated with low-dose IFNs show a reduction in viral replication, suggesting the prophylactic effectiveness of IFNs against SARS-CoV-2. Human stem cell-based alveolospheres, thus, provide novel insights into COVID-19 pathogenesis and can serve as a model for understanding human respiratory diseases.


Asunto(s)
Células Madre Adultas/virología , Células Epiteliales Alveolares/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , Interferones/farmacología , SARS-CoV-2/inmunología , Adulto , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/enzimología , Anciano , Anciano de 80 o más Años , Células Epiteliales Alveolares/enzimología , Células Epiteliales Alveolares/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/fisiopatología , Técnicas de Cultivo de Célula , Diferenciación Celular , Femenino , Humanos , Inflamación , Masculino , Ratones , Receptores de Coronavirus/metabolismo , Transcriptoma , Replicación Viral
5.
Stem Cells Dev ; 28(24): 1632-1645, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31650883

RESUMEN

Age-related cellular changes and limited replicative capacity of adult mesenchymal stem cells (MSCs) are few of the challenges confronting stem cell research. MSCs from human fetal membranes (hFM-MSCs), including placental, umbilical cord, and amniotic membrane, are considered an alternative to adult MSCs. However, the effect of mothers' age on hFM-MSC cellular properties is still not clearly established. This study aimed to evaluate the effect of mothers' age on hFM-MSC telomere length, telomerase activity, and proliferation ability in three different age groups: GI (20-29 years), GII (30-39 years), and GIII (≥40 years). hFM samples were collected from pregnant women ≤37 weeks after obtaining consent. hFM-MSCs were isolated and cultured to characterize them by flow cytometry and assess proliferation by MTT assay and doubling time. Telomere length and expression levels of human telomerase reverse transcriptase were assessed by quantitative real-time polymerase chain reaction (qRT-RCR). hFM-MSCs in the three age groups were spindle-shaped, plastic-adherent, and exhibited high proliferation rates and strong expression of hMSC markers. GI showed the longest telomere length in hMSCs in various FM regions, whereas GIII showed the highest level of telomerase expression. There was no difference in telomere length between GII and GIII, and both groups showed the same hMSC characteristics. In conclusion, although the hFM-MSCs derived from different fetal membranes maintained the MSC characteristics in all study groups, the hFM-MSCs of older mothers had shorter telomeres and higher telomerase activity and proliferation rate than did those derived from younger mothers. Thus, the hFM-MSCs of older mothers could be unsuitable for expansion in vitro or stem cell therapy. Determination of telomere length and telomerase expression level of hFM might help characterizing and understanding the biological differences of hFM-MSCs in different age groups.


Asunto(s)
Células Madre Adultas/enzimología , Células Madre Mesenquimatosas/enzimología , Telomerasa/genética , Homeostasis del Telómero/genética , Adulto , Células Madre Adultas/metabolismo , Factores de Edad , Diferenciación Celular/genética , Proliferación Celular/genética , Membranas Extraembrionarias/enzimología , Membranas Extraembrionarias/crecimiento & desarrollo , Femenino , Citometría de Flujo , Humanos , Células Madre Mesenquimatosas/metabolismo , Madres , Placenta/citología , Embarazo , Telómero/genética , Cordón Umbilical/crecimiento & desarrollo , Cordón Umbilical/metabolismo
6.
Am J Respir Cell Mol Biol ; 61(4): 481-491, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30917006

RESUMEN

The discovery of mutant tyrosine kinases as oncogenic drivers of lung adenocarcinomas has changed the basic understanding of lung cancer development and therapy. Yet, expressed kinases (kinome) in lung cancer progenitor cells, as well as whether kinase expression and the overall kinome changes or is reprogrammed upon transformation, is incompletely understood. We hypothesized that the kinome differs between lung cancer progenitor cells, alveolar type II cells (ATII), and basal cells (BC) and that their respective kinomes undergo distinct lineage-specific reprogramming to adenocarcinomas and squamous cell carcinomas upon transformation. We performed RNA sequencing on freshly isolated human ATII, BC, and lung cancer cell lines to define the kinome in nontransformed cells and transformed cells. Our studies identified a unique kinome for ATII and BC and changes in their kinome upon transformation to their respective carcinomas.


Asunto(s)
Células Madre Adultas/enzimología , Células Epiteliales Alveolares/enzimología , Transformación Celular Neoplásica , Neoplasias Pulmonares/enzimología , Pulmón/enzimología , Proteínas de Neoplasias/análisis , Proteínas Tirosina Quinasas/análisis , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Animales , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Linaje de la Célula , Células Cultivadas , Inducción Enzimática , Humanos , Pulmón/citología , Neoplasias Pulmonares/genética , Ratones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/enzimología , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , ARN Mensajero/análisis , ARN Neoplásico/análisis , Transcriptoma
7.
Int J Exp Pathol ; 99(3): 113-120, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30073722

RESUMEN

Precartilaginous stem cells (PSCs) are adult stem cells which could self-renew or differentiate into chondrocytes to promote bone growth. In this study, we aimed to understand the role of transforming growth factor-ß1 (TGF-ß1) in precartilaginous stem cell (PSC) differentiation and to study the mechanisms that underlie this role. We purified PSCs from the neonatal murine perichondrial mesenchyme using immunomagnetic beads, and primary cultured them. Their phenotype was confirmed by the PSC marker fibroblast growth factor receptor-3 (FGFR-3) overexpression. TGF-ß1 was added to induce PSCs differentiation. TGF-ß1 increased mRNA expression of chondrogenesis-related genes (collagen type II, Sox 9 and aggrecan) in the cultured PSCs. This was abolished by TGF-ß receptor II (TGFRII) and Casein kinase 1 epsilon (CK1ε) lentiviral shRNA depletion. Meanwhile, we found that TGF-ß1 induced CK1ε activation, glycogen synthase kinase-3ß (GSK3ß) phosphorylation and ß-catenin nuclear translocation in the mouse PSCs, which was almost completely blocked by TGFRII and CK1ε shRNA knockdown. Based on these results, we suggest that TGF-ß1 induces CK1ε activation to promote ß-catenin nuclear accumulation, which then regulates chondrogenesis-related gene transcription to eventually promote mouse PSC differentiation.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Caseína Cinasa 1 épsilon/metabolismo , Diferenciación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Receptor Tipo II de Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta1/farmacología , beta Catenina/metabolismo , Células Madre Adultas/enzimología , Agrecanos/genética , Agrecanos/metabolismo , Animales , Animales Recién Nacidos , Caseína Cinasa 1 épsilon/genética , Diferenciación Celular/genética , Células Cultivadas , Condrocitos/enzimología , Condrogénesis/genética , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratones Endogámicos C57BL , Técnicas de Transferencia Nuclear , Fenotipo , Fosforilación , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Mol Med Rep ; 18(1): 1067-1073, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29845240

RESUMEN

As one type of adult stem cells (ASCs), human dental pulp stem cells (HDPSCs) have several properties, including high proliferation rate, self­renewal capability, and multi­lineage differentiation. However, the apoptotic mechanism underlying the development of dental pulp cells remains unclear. In the present study, a significant increase of apoptosis was observed in HDPSCs from the deciduous teeth compared with that from adult permanent teeth. In addition, the occurrence of cytochrome c expression and mitochondrial­mediated apoptosis pathway activity in HDPSCs were confirmed by quantitative polymerase chain reaction, and western blotting. Although caspase­8 and caspase­9 showed higher expression in deciduous teeth than in adult permanent teeth, only the knockdown of caspase­9 via RNA interference in HDPSC cells exhibited a significant reduction in apoptosis, and caspase­3 expression and activity. All these results revealed that caspase­9 and activated caspase­3 predominantly regulates cell apoptosis in HDPSCs from deciduous teeth.


Asunto(s)
Células Madre Adultas/enzimología , Apoptosis , Caspasa 9/biosíntesis , Pulpa Dental/enzimología , Regulación Enzimológica de la Expresión Génica , Diente Primario/enzimología , Adolescente , Adulto , Células Madre Adultas/citología , Caspasa 3/biosíntesis , Niño , Pulpa Dental/citología , Femenino , Humanos , Masculino , Diente Primario/citología
9.
J Biol Chem ; 293(8): 2711-2724, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29284678

RESUMEN

Chromatin in embryonic stem cells (ESCs) differs markedly from that in somatic cells, with ESCs exhibiting a more open chromatin configuration. Accordingly, ATP-dependent chromatin remodeling complexes are important regulators of ESC homeostasis. Depletion of the remodeler SMARCAD1, an ATPase of the SNF2 family, has been shown to affect stem cell state, but the mechanistic explanation for this effect is unknown. Here, we set out to gain further insights into the function of SMARCAD1 in mouse ESCs. We identified KRAB-associated protein 1 (KAP1) as the stoichiometric binding partner of SMARCAD1 in ESCs. We found that this interaction occurs on chromatin and that SMARCAD1 binds to different classes of KAP1 target genes, including zinc finger protein (ZFP) and imprinted genes. We also found that the RING B-box coiled-coil (RBCC) domain in KAP1 and the proximal coupling of ubiquitin conjugation to ER degradation (CUE) domain in SMARCAD1 mediate their direct interaction. Of note, retention of SMARCAD1 in the nucleus depended on KAP1 in both mouse ESCs and human somatic cells. Mutations in the CUE1 domain of SMARCAD1 perturbed the binding to KAP1 in vitro and in vivo Accordingly, an intact CUE1 domain was required for tethering this remodeler to the nucleus. Moreover, mutation of the CUE1 domain compromised SMARCAD1 binding to KAP1 target genes. Taken together, our results reveal a mechanism that localizes SMARCAD1 to genomic sites through the interaction of SMARCAD1's CUE1 motif with KAP1.


Asunto(s)
Células Madre Adultas/metabolismo , Núcleo Celular/metabolismo , ADN Helicasas/metabolismo , Regulación de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/enzimología , Sustitución de Aminoácidos , Animales , Línea Celular , Núcleo Celular/enzimología , Cromatina/química , Cromatina/enzimología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/química , ADN Helicasas/genética , Eliminación de Gen , Humanos , Cinética , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/enzimología , Mutación , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteína 28 que Contiene Motivos Tripartito/antagonistas & inhibidores , Proteína 28 que Contiene Motivos Tripartito/química , Proteína 28 que Contiene Motivos Tripartito/genética
10.
Cell Death Differ ; 24(12): 2101-2116, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28800128

RESUMEN

Multipotent adult resident cardiac stem cells (CSCs) were first identified by the expression of c-kit, the stem cell factor receptor. However, in the adult myocardium c-kit alone cannot distinguish CSCs from other c-kit-expressing (c-kitpos) cells. The adult heart indeed contains a heterogeneous mixture of c-kitpos cells, mainly composed of mast and endothelial/progenitor cells. This heterogeneity of cardiac c-kitpos cells has generated confusion and controversy about the existence and role of CSCs in the adult heart. Here, to unravel CSC identity within the heterogeneous c-kit-expressing cardiac cell population, c-kitpos cardiac cells were separated through CD45-positive or -negative sorting followed by c-kitpos sorting. The blood/endothelial lineage-committed (Lineagepos) CD45posc-kitpos cardiac cells were compared to CD45neg(Lineageneg/Linneg) c-kitpos cardiac cells for stemness and myogenic properties in vitro and in vivo. The majority (~90%) of the resident c-kitpos cardiac cells are blood/endothelial lineage-committed CD45posCD31posc-kitpos cells. In contrast, the LinnegCD45negc-kitpos cardiac cell cohort, which represents ⩽10% of the total c-kitpos cells, contain all the cardiac cells with the properties of adult multipotent CSCs. These characteristics are absent from the c-kitneg and the blood/endothelial lineage-committed c-kitpos cardiac cells. Single Linnegc-kitpos cell-derived clones, which represent only 1-2% of total c-kitpos myocardial cells, when stimulated with TGF-ß/Wnt molecules, acquire full transcriptome and protein expression, sarcomere organisation, spontaneous contraction and electrophysiological properties of differentiated cardiomyocytes (CMs). Genetically tagged cloned progeny of one Linnegc-kitpos cell when injected into the infarcted myocardium, results in significant regeneration of new CMs, arterioles and capillaries, derived from the injected cells. The CSC's myogenic regenerative capacity is dependent on commitment to the CM lineage through activation of the SMAD2 pathway. Such regeneration was not apparent when blood/endothelial lineage-committed c-kitpos cardiac cells were injected. Thus, among the cardiac c-kitpos cell cohort only a very small fraction has the phenotype and the differentiation/regenerative potential characteristics of true multipotent CSCs.


Asunto(s)
Células Madre Adultas/enzimología , Células Madre Multipotentes/enzimología , Miocardio/enzimología , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Células Madre Adultas/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Masculino , Ratones , Células Madre Multipotentes/citología , Miocardio/citología , Ratas , Ratas Wistar
11.
J Cell Biol ; 216(9): 2715-2729, 2017 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-28739678

RESUMEN

Pre-B-cell leukemia homeobox (PBX) and myeloid ecotropic viral integration site (MEIS) proteins control cell fate decisions in many physiological and pathophysiological contexts, but how these proteins function mechanistically remains poorly defined. Focusing on the first hours of neuronal differentiation of adult subventricular zone-derived stem/progenitor cells, we describe a sequence of events by which PBX-MEIS facilitates chromatin accessibility of transcriptionally inactive genes: In undifferentiated cells, PBX1 is bound to the H1-compacted promoter/proximal enhancer of the neuron-specific gene doublecortin (Dcx) Once differentiation is induced, MEIS associates with chromatin-bound PBX1, recruits PARP1/ARTD1, and initiates PARP1-mediated eviction of H1 from the chromatin fiber. These results for the first time link MEIS proteins to PARP-regulated chromatin dynamics and provide a mechanistic basis to explain the profound cellular changes elicited by these proteins.


Asunto(s)
Células Madre Adultas/enzimología , Linaje de la Célula , Cromatina/enzimología , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/enzimología , Neurogénesis , Neuropéptidos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Cromatina/genética , Ensamble y Desensamble de Cromatina , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/genética , Neuropéptidos/genética , Fenotipo , Poli(ADP-Ribosa) Polimerasa-1/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , Esferoides Celulares , Nicho de Células Madre , Factores de Tiempo , Factores de Transcripción/genética , Transcripción Genética , Transfección
12.
Nat Commun ; 8: 14328, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28094257

RESUMEN

Satellite cells (SCs) are myogenic stem cells required for regeneration of adult skeletal muscles. A proper balance among quiescence, activation and differentiation is essential for long-term maintenance of SCs and their regenerative function. Here we show a function of Pten (phosphatase and tensin homologue) in quiescent SCs. Deletion of Pten in quiescent SCs leads to their spontaneous activation and premature differentiation without proliferation, resulting in depletion of SC pool and regenerative failure. However, prior to depletion, Pten-null activated SCs can transiently proliferate upon injury and regenerate injured muscles, but continually decline during regeneration, suggesting an inability to return to quiescence. Mechanistically, Pten deletion increases Akt phosphorylation, which induces cytoplasmic translocation of FoxO1 and suppression of Notch signalling. Accordingly, constitutive activation of Notch1 prevents SC depletion despite Pten deletion. Our findings delineate a critical function of Pten in maintaining SC quiescence and reveal an interaction between Pten and Notch signalling.


Asunto(s)
Células Madre Adultas/enzimología , Senescencia Celular , Fosfohidrolasa PTEN/metabolismo , Células Satélite del Músculo Esquelético/enzimología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Masculino , Ratones , Ratones Noqueados , Desarrollo de Músculos , Fosfohidrolasa PTEN/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo
13.
Genes Dev ; 29(23): 2420-34, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26584619

RESUMEN

Telomerase inactivation causes loss of the male germline in worms, fish, and mice, indicating a conserved dependence on telomere maintenance in this cell lineage. Here, using telomerase reverse transcriptase (Tert) reporter mice, we found that very high telomerase expression is a hallmark of undifferentiated spermatogonia, the mitotic population where germline stem cells reside. We exploited these high telomerase levels as a basis for purifying undifferentiated spermatogonia using fluorescence-activated cell sorting. Telomerase levels in undifferentiated spermatogonia and embryonic stem cells are comparable and much greater than in somatic progenitor compartments. Within the germline, we uncovered an unanticipated gradient of telomerase activity that also enables isolation of more mature populations. Transcriptomic comparisons of Tert(High) undifferentiated spermatogonia and Tert(Low) differentiated spermatogonia by RNA sequencing reveals marked differences in cell cycle and key molecular features of each compartment. Transplantation studies show that germline stem cell activity is confined to the Tert(High) cKit(-) population. Telomere shortening in telomerase knockout strains causes depletion of undifferentiated spermatogonia and eventual loss of all germ cells after undifferentiated spermatogonia drop below a critical threshold. These data reveal that high telomerase expression is a fundamental characteristic of germline stem cells, thus explaining the broad dependence on telomerase for germline immortality in metazoans.


Asunto(s)
Células Madre Adultas/enzimología , Regulación Enzimológica de la Expresión Génica , Espermatogonias/enzimología , Telomerasa/genética , Telomerasa/metabolismo , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/enzimología , Citometría de Flujo , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética
14.
J Am Heart Assoc ; 4(7)2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26178404

RESUMEN

BACKGROUND: Mammalian heart regenerative activity is lost before adulthood but increases after cardiac injury. Cardiac repair mechanisms, which involve both endogenous cardiac stem cells (CSCs) and cardiomyocyte cell-cycle reentry, are inadequate to achieve full recovery after myocardial infarction (MI). Mice deficient in S-nitrosoglutathione reductase (GSNOR(-/-)), an enzyme regulating S-nitrosothiol turnover, have preserved cardiac function after MI. Here, we tested the hypothesis that GSNOR activity modulates cardiac cell proliferation in the post-MI adult heart. METHODS AND RESULTS: GSNOR(-/-) and C57Bl6/J (wild-type [WT]) mice were subjected to sham operation (n=3 GSNOR(-/-); n=3 WT) or MI (n=41 GSNOR(-/-); n=65 WT). Compared with WT, GSNOR(-/-) mice exhibited improved survival, cardiac performance, and architecture after MI, as demonstrated by higher ejection fraction (P<0.05), lower endocardial volumes (P<0.001), and diminished scar size (P<0.05). In addition, cardiomyocytes from post-MI GSNOR(-/-) hearts exhibited faster calcium decay and sarcomeric relaxation times (P<0.001). Immunophenotypic analysis illustrated that post-MI GSNOR(-/-) hearts demonstrated enhanced neovascularization (P<0.001), c-kit(+) CSC abundance (P=0.013), and a ≈3-fold increase in proliferation of adult cardiomyocytes and c-kit(+)/CD45(-) CSCs (P<0.0001 and P=0.023, respectively) as measured by using 5-bromodeoxyuridine. CONCLUSIONS: Loss of GSNOR confers enhanced post-MI cardiac regenerative activity, characterized by enhanced turnover of cardiomyocytes and CSCs. Endogenous denitrosylases exert an inhibitory effect over cardiac repair mechanisms and therefore represents a potential novel therapeutic target.


Asunto(s)
Células Madre Adultas/enzimología , Alcohol Deshidrogenasa/deficiencia , Proliferación Celular , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , Regeneración , Células Madre Adultas/patología , Alcohol Deshidrogenasa/genética , Animales , Biomarcadores/metabolismo , Señalización del Calcio , Células Cultivadas , Modelos Animales de Enfermedad , Homocigoto , Antígenos Comunes de Leucocito/deficiencia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Volumen Sistólico , Factores de Tiempo
15.
Neurosci Res ; 95: 1-11, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25659757

RESUMEN

Adult neurogenesis persists throughout life in the dentate gyrus (DG) of the hippocampus, and its importance has been highlighted in hippocampus-dependent learning and memory. Adult neurogenesis consists of multiple processes: maintenance and neuronal differentiation of neural stem/precursor cells (NS/PCs), followed by survival and maturation of newborn neurons and their integration into existing neuronal circuitry. However, the mechanisms that govern these processes remain largely unclear. Here we show that DNA methyltransferase 1 (DNMT1), an enzyme responsible for the maintenance of DNA methylation, is highly expressed in proliferative cells in the adult DG and plays an important role in the survival of newly generated neurons. Deletion of Dnmt1 in adult NS/PCs (aNS/PCs) did not affect the proliferation and differentiation of aNS/PCs per se. However, it resulted in a decrease of newly generated mature neurons, probably due to gradual cell death after aNS/PCs differentiated into neurons in the hippocampus. Interestingly, loss of DNMT1 in post-mitotic neurons did not influence their survival. Taken together, these findings suggest that the presence of DNMT1 in aNS/PCs is crucial for the survival of newly generated neurons, but is dispensable once they accomplish neuronal differentiation in the adult hippocampus.


Asunto(s)
Células Madre Adultas/enzimología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Giro Dentado/enzimología , Células-Madre Neurales/enzimología , Neuronas/enzimología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Giro Dentado/citología , Ratones , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/citología
16.
Proc Natl Acad Sci U S A ; 111(37): 13541-6, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25161285

RESUMEN

The maintenance of the resident adult neural stem/progenitor cell (NSPC) pool depends on the precise balance of proliferation, differentiation, and maintenance of the undifferentiated state. Identifying the mechanisms that regulate this balance in adult hippocampal NSPCs can provide insight into basic stem cell self-renewal principles important for tissue homeostasis and preventing tumor formation. Pharmacological inhibition of histone deacetylases (HDACs), a class of histone-modifying enzymes, have promising effects in cancer cells, yet the specific roles of individual HDACs in stem cell proliferation is unclear. Here using conditional KO (cKO) mice and in vitro cell culture, we show that histone deacetylase 3 (HDAC3) is required for the proliferation of adult NSPCs. Detailed cell cycle analysis of NSPCs from Hdac3 cKO mice reveals a defect in cell cycle progression through the gap 2/mitosis (G2/M) but not the S phase. Moreover, HDAC3 controls G2/M phase progression mainly through posttranslational stabilization of the G2/M cyclin-dependent kinase 1 (CDK1). These results demonstrate that HDAC3 plays a critical role in NSPC proliferation and suggest that strategies aimed at pharmacological modulation of HDAC3 may be beneficial for tissue regeneration and controlling tumor cell growth.


Asunto(s)
Células Madre Adultas/citología , Proteína Quinasa CDC2/metabolismo , Fase G2 , Histona Desacetilasas/metabolismo , Mitosis , Células-Madre Neurales/citología , Células Madre Adultas/enzimología , Envejecimiento/fisiología , Animales , Proliferación Celular , Giro Dentado/citología , Giro Dentado/enzimología , Regulación hacia Abajo , Eliminación de Gen , Ratones Noqueados , Células-Madre Neurales/enzimología , Neurogénesis , Neuronas/citología , Neuronas/enzimología , Proteolisis
17.
Mol Hum Reprod ; 20(9): 905-18, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24939730

RESUMEN

Retrograde flow of menstrual blood cells during menstruation is considered as the dominant theory for the development of endometriosis. Moreover, current evidence suggests that endometrial-derived stem cells are key players in the pathogenesis of endometriosis. In particular, endometrial stromal stem cells have been suggested to be involved in the pathogenesis of this disease. Here, we aimed to use menstrual blood, as a novel source of endometrial stem cells, to investigate whether stromal stem cells from endometriosis (E-MenSCs) and non-endometriosis (NE-MenSCs) women differed regarding their morphology, CD marker expression pattern, proliferation, invasion and adhesion capacities and their ability to express certain immunomodulatory molecules. E-MenSCs were morphologically different from NE-MenSCs and showed higher expression of CD9, CD10 and CD29. Furthermore, E-MenSCs had higher proliferation and invasion potentials compared with NE-MenSCs. The amount of indoleamine 2,3-dioxygenase-1 (IDO1) and cyclooxygenase-2 (COX-2) in E-MenSCs co-cultured with allogenic peripheral blood mononuclear cells (PBMCs) was shown to be higher both at the gene and protein levels, and higher IDO1 activity was detected in the endometriosis group. However, NE-MenSCs revealed increased concentrations of forkhead transcription factor-3 (FOXP3) when compared with E-MenSCs. Nonetheless, interferon (IFN)-γ, Interleukin (IL)-10 and monocyte chemoattractant protein-1 (MCP-1) levels were higher in the supernatant of E-MenSCs-PBMC co-cultures. Here, we showed that there are inherent differences between E-MenSCs and NE-MenSCs. These findings propose the key role MenSCs could play in the pathogenesis of endometriosis and further support the retrograde and stem cell theories of endometriosis. Hence, considering its renewable and easily available nature, menstrual blood could be viewed as a reliable and inexpensive material for studies addressing the cellular and molecular aspects of endometriosis.


Asunto(s)
Células Madre Adultas/patología , Endometriosis/patología , Endometrio/patología , Menstruación , Células del Estroma/patología , Adulto , Células Madre Adultas/enzimología , Células Madre Adultas/metabolismo , Biomarcadores/metabolismo , Comunicación Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Endometriosis/inmunología , Endometriosis/metabolismo , Endometriosis/fisiopatología , Endometrio/citología , Endometrio/metabolismo , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Irán , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Células del Estroma/enzimología , Células del Estroma/metabolismo , Regulación hacia Arriba , Adulto Joven
18.
Stem Cells ; 32(7): 1878-89, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24677703

RESUMEN

Jak2, a member of the Janus kinase family of nonreceptor protein tyrosine kinases, is activated in response to a variety of cytokines, and functions in survival and proliferation of cells. An activating JAK2V617F mutation has been found in most patients with myeloproliferative neoplasms, and patients treated with Jak2 inhibitors show significant hematopoietic toxicities. However, the role of Jak2 in adult hematopoietic stem cells (HSCs) has not been clearly elucidated. Using a conditional Jak2 knockout allele, we have found that Jak2 deletion results in rapid loss of HSCs/progenitors leading to bone marrow failure and early lethality in adult mice. Jak2 deficiency causes marked impairment in HSC function, and the mutant HSCs are severely defective in reconstituting hematopoiesis in recipient animals. Jak2 deficiency also causes significant apoptosis and loss of quiescence in HSC-enriched LSK (Lin(-)Sca-1(+)c-Kit(+)) cells. Jak2-deficient LSK cells exhibit elevated reactive oxygen species levels and enhanced p38 MAPK activation. Mutant LSK cells also show defective Stat5, Erk, and Akt activation in response to thrombopoietin and stem cell factor. Gene expression analysis reveals significant downregulation of genes related to HSC quiescence and self-renewal in Jak2-deficient LSK cells. These data suggest that Jak2 plays a critical role in the maintenance and function of adult HSCs.


Asunto(s)
Células Madre Adultas/enzimología , Células Madre Hematopoyéticas/enzimología , Janus Quinasa 2/fisiología , Células Madre Adultas/fisiología , Anemia Aplásica , Animales , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Proliferación Celular , Supervivencia Celular , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Hemoglobinuria Paroxística/enzimología , Ratones Endogámicos C57BL , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo , Factor de Células Madre/fisiología , Trombopoyetina/fisiología
19.
J Mater Sci Mater Med ; 25(4): 1137-48, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24378848

RESUMEN

Mineralized poly(ε-caprolactone)/gelatin core-shell nanofibers were prepared via co-axial electrospinning and subsequent incubation in biomimetic simulated body fluid containing ten times the calcium and phosphate ion concentrations found in human blood plasma. The deposition of calcium phosphate on the nanofiber surfaces was investigated through scanning electronic microscopy and X-ray diffraction. Energy dispersive spectroscopy results indicated that calcium-deficient hydroxyapatite had grown on the fibers. Fourier transform infrared spectroscopy analysis suggested the presence of hydroxyl-carbonate-apatite. The results of a viability assay (MTT) and alkaline phosphatase activity analysis suggested that these mineralized matrices promote osteogenic differentiation of human adipose-derived stem cells (hASCs) when cultured in an osteogenic medium and have the potential to be used as a scaffold in bone tissue engineering. hASCs cultured in the presence of nanofibers in endothelial differentiation medium showed lower rates of proliferation than cells cultured without the nanofibers. However, endothelial cell markers were detected in cells cultured in the presence of nanofibers in endothelial differentiation medium.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/citología , Materiales Biocompatibles/química , Nanofibras/química , Células Madre Adultas/enzimología , Fosfatasa Alcalina/metabolismo , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Endoteliales/citología , Gelatina/química , Humanos , Ensayo de Materiales , Microscopía Electrónica de Rastreo , Minerales/química , Nanofibras/ultraestructura , Osteogénesis , Poliésteres/química
20.
Genet Mol Res ; 13(1): 220-7, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24446314

RESUMEN

Although ginkgo biloba extract (GBE) was shown to have antioxidant effects, little has been reported on the ability to GBE to help endothelial progenitor cells (EPCs) resist oxidative stress. The present study evaluated the influence of different concentrations of GBE on superoxide dismutase (SOD) and apoptosis of diabetic peripheral blood EPCs. Twenty-five diabetic patients without any vascular complications were included in the experimental group, while 15 healthy adults made up the control group. Peripheral blood mononuclear cells were isolated with density gradient centrifugation, and, after in vitro differentiation, were determined to be EPCs using FITC-UEA-I and Dil-Ac-LDL dual staining. After the colony and fusiform adherent cells were observed, on day 7, various concentrations of ginkgo biloba extract (0, 10, 25, 50 mg/L) were added to the culture medium for a 24-h incubation. EPC-SOD activity and apoptosis were subsequently detected. We found that within the experimental group, GBE significantly improved SOD activity within EPCs and reduced the rate of apoptosis. These effects became more obvious with increasing GBE concentrations (25 mg/L, P < 0.05; 50 mg/L, P < 0.01). GBE also improved SOD activity and reduced the rate of apoptosis within EPCs of the control group; however, the changes were not statistically significant. We conclude that GBE can improve SOD activity and reduce the rate of apoptosis of EPCs within the peripheral blood of diabetic patients, effects that are dose-dependent.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Ginkgo biloba/química , Extractos Vegetales/farmacología , Superóxido Dismutasa/metabolismo , Adulto , Células Madre Adultas/enzimología , Células Madre Adultas/fisiología , Estudios de Casos y Controles , Diabetes Mellitus/sangre , Células Endoteliales/enzimología , Células Endoteliales/fisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...