Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Elife ; 122024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38180318

RESUMEN

The unlimited expansion of human progenitor cells in vitro could unlock many prospects for regenerative medicine. However, it remains an important challenge as it requires the decoupling of the mechanisms supporting progenitor self-renewal and expansion from those mechanisms promoting their differentiation. This study focuses on the expansion of human pluripotent stem (hPS) cell-derived pancreatic progenitors (PP) to advance novel therapies for diabetes. We obtained mechanistic insights into PP expansion requirements and identified conditions for the robust and unlimited expansion of hPS cell-derived PP cells under GMP-compliant conditions through a hypothesis-driven iterative approach. We show that the combined stimulation of specific mitogenic pathways, suppression of retinoic acid signaling, and inhibition of selected branches of the TGFß and Wnt signaling pathways are necessary for the effective decoupling of PP proliferation from differentiation. This enabled the reproducible, 2000-fold, over 10 passages and 40-45 d, expansion of PDX1+/SOX9+/NKX6-1+ PP cells. Transcriptome analyses confirmed the stabilization of PP identity and the effective suppression of differentiation. Using these conditions, PDX1+/SOX9+/NKX6-1+ PP cells, derived from different, both XY and XX, hPS cell lines, were enriched to nearly 90% homogeneity and expanded with very similar kinetics and efficiency. Furthermore, non-expanded and expanded PP cells, from different hPS cell lines, were differentiated in microwells into homogeneous islet-like clusters (SC-islets) with very similar efficiency. These clusters contained abundant ß-cells of comparable functionality as assessed by glucose-stimulated insulin secretion assays. These findings established the signaling requirements to decouple PP proliferation from differentiation and allowed the consistent expansion of hPS cell-derived PP cells. They will enable the establishment of large banks of GMP-produced PP cells derived from diverse hPS cell lines. This approach will streamline SC-islet production for further development of the differentiation process, diabetes research, personalized medicine, and cell therapies.


Asunto(s)
Diabetes Mellitus , Células Madre Pluripotentes , Humanos , Páncreas , Vía de Señalización Wnt , Bioensayo
2.
Arthroscopy ; 38(9): 2602-2608, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35278599

RESUMEN

PURPOSE: The purpose of this study is to investigate the outcomes of arthroscopic rotator cuff repair in a severely obese population (body mass index [BMI] > 0 kg/m2) compared to a healthy weight population (BMI 18.5-24.9 kg/m2). METHODS: This study is a retrospective review of prospectively collected data examining the outcomes of arthroscopic rotator cuff repair in both severely obese patients and healthy weight patients. Primary outcome measures analyzed include the American Shoulder and Elbow Surgeons (ASES) Score, the Single Assessment Numeric Evaluation (SANE), pain Visual Analog Scale (VAS), range of motion, and complications. RESULTS: A total of 89 patients met inclusion/exclusion criteria: 52 healthy weight patients (BMI 18.5-24.9 kg/m2) and 37 severely obese patients (BMI >40 kg/m2). Patient-reported pain and functional outcomes had significantly improved after surgery in both groups with regard to the visual analog score (VAS) scores, Single Assessment Numeric Evaluation (SANE) scores, and American Shoulder and Elbow Surgeons Shoulder (ASES) scores (P < .0001). When directly comparing the outcomes in the healthy weight group to the severely obese group, the latter had significantly inferior outcomes in VAS scores (P = .0048), SANE scores (P = .0118), ASES scores (P = .0031), and postoperative internal rotation (P =.0132). At large, these outcomes did not have clinically significant differences. The severely obese group also had higher total numbers of comorbid conditions and longer operative times (P =.0041). CONCLUSIONS: Severely obese patients and their associated comorbid conditions pose unique challenges in rotator cuff tear management, but they still achieve overall excellent outcomes after repair and noninferior clinical differences when compared to healthy weight patients. LEVEL OF EVIDENCE: Level III, retrospective comparative study.


Asunto(s)
Obesidad Mórbida , Lesiones del Manguito de los Rotadores , Artroscopía/efectos adversos , Humanos , Obesidad Mórbida/complicaciones , Rango del Movimiento Articular , Estudios Retrospectivos , Manguito de los Rotadores/cirugía , Lesiones del Manguito de los Rotadores/complicaciones , Lesiones del Manguito de los Rotadores/cirugía , Dolor de Hombro/etiología , Resultado del Tratamiento
3.
Stem Cells ; 40(2): 175-189, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35257173

RESUMEN

Hox genes play key roles in the anterior-posterior (AP) specification of all 3 germ layers during different developmental stages. It is only partially understood how they function in widely different developmental contexts, particularly with regards to extracellular signaling, and to what extent their function can be harnessed to guide cell specification in vitro. Here, we addressed the role of Hoxb1 in 2 distinct developmental contexts; in mouse embryonic stem cells (mES)-derived neuromesodermal progenitors (NMPs) and hindbrain neural progenitors. We found that Hoxb1 promotes NMP survival through the upregulation of Fgf8, Fgf17, and other components of Fgf signaling as well as the repression of components of the apoptotic pathway. Additionally, it upregulates other anterior Hox genes suggesting that it plays an active role in the early steps of AP specification. In neural progenitors, Hoxb1 synergizes with shh to repress anterior and dorsal neural markers, promote the expression of ventral neural markers and direct the specification of facial branchiomotorneuron (FBM)-like progenitors. Hoxb1 and shh synergize in regulating the expression of diverse signals and signaling molecules, including the Ret tyrosine kinase receptor. Finally, Hoxb1 synergizes with exogenous Glial cell line-derived neurotrophic factor (GDNF) to strengthen Ret expression and further promote the generation of FBM-like progenitors. Facial branchiomotorneuron-like progenitors survived for at least 6 months and differentiated into postmitotic neurons after orthotopic transplantation near the facial nucleus of adult mice. These results suggested that the patterning activity of Hox genes in combination with downstream signaling molecules can be harnessed for the generation of defined neural populations and transplantations with implications for neurodegenerative diseases.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Rombencéfalo , Animales , Diferenciación Celular/genética , Supervivencia Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Rombencéfalo/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
4.
Front Endocrinol (Lausanne) ; 12: 722250, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421829

RESUMEN

In all forms of diabetes, ß cell mass or function is reduced and therefore the capacity of the pancreatic cells for regeneration or replenishment is a critical need. Diverse lines of research have shown the capacity of endocrine as well as acinar, ductal and centroacinar cells to generate new ß cells. Several experimental approaches using injury models, pharmacological or genetic interventions, isolation and in vitro expansion of putative progenitors followed by transplantations or a combination thereof have suggested several pathways for ß cell neogenesis or regeneration. The experimental results have also generated controversy related to the limitations and interpretation of the experimental approaches and ultimately their physiological relevance, particularly when considering differences between mouse, the primary animal model, and human. As a result, consensus is lacking regarding the relative importance of islet cell proliferation or progenitor differentiation and transdifferentiation of other pancreatic cell types in generating new ß cells. In this review we summarize and evaluate recent experimental approaches and findings related to islet regeneration and address their relevance and potential clinical application in the fight against diabetes.


Asunto(s)
Células Secretoras de Insulina/fisiología , Páncreas/fisiología , Regeneración/fisiología , Adulto , Animales , Recuento de Células , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Transdiferenciación Celular/fisiología , Humanos , Células Secretoras de Insulina/citología , Ratones , Tamaño de los Órganos , Páncreas/citología , Células Madre/fisiología
5.
Elife ; 92020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32804075

RESUMEN

Perturbation of addition of second heart field (SHF) cardiac progenitor cells to the poles of the heart tube results in congenital heart defects (CHD). The transcriptional programs and upstream regulatory events operating in different subpopulations of the SHF remain unclear. Here, we profile the transcriptome and chromatin accessibility of anterior and posterior SHF sub-populations at genome-wide levels and demonstrate that Hoxb1 negatively regulates differentiation in the posterior SHF. Spatial mis-expression of Hoxb1 in the anterior SHF results in hypoplastic right ventricle. Activation of Hoxb1 in embryonic stem cells arrests cardiac differentiation, whereas Hoxb1-deficient mouse embryos display premature cardiac differentiation. Moreover, ectopic differentiation in the posterior SHF of embryos lacking both Hoxb1 and its paralog Hoxa1 results in atrioventricular septal defects. Our results show that Hoxb1 plays a key role in patterning cardiac progenitor cells that contribute to both cardiac poles and provide new insights into the pathogenesis of CHD.


Asunto(s)
Cardiopatías Congénitas/genética , Proteínas de Homeodominio/genética , Células Madre/metabolismo , Transcriptoma , Animales , Cromatina/metabolismo , Genes Homeobox , Cardiopatías Congénitas/embriología , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos
6.
Proc Natl Acad Sci U S A ; 116(41): 20679-20688, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548432

RESUMEN

The presence of progenitor or stem cells in the adult pancreas and their potential involvement in homeostasis and cancer development remain unresolved issues. Here, we show that mouse centroacinar cells can be identified and isolated by virtue of the mitochondrial enzyme Aldh1b1 that they uniquely express. These cells are necessary and sufficient for the formation of self-renewing adult pancreatic organoids in an Aldh1b1-dependent manner. Aldh1b1-expressing centroacinar cells are largely quiescent, self-renew, and, as shown by genetic lineage tracing, contribute to all 3 pancreatic lineages in the adult organ under homeostatic conditions. Single-cell RNA sequencing analysis of these cells identified a progenitor cell population, established its molecular signature, and determined distinct differentiation pathways to early progenitors. A distinct feature of these progenitor cells is the preferential expression of small GTPases, including Kras, suggesting that they might be susceptible to Kras-driven oncogenic transformation. This finding and the overexpression of Aldh1b1 in human and mouse pancreatic cancers, driven by activated Kras, prompted us to examine the involvement of Aldh1b1 in oncogenesis. We demonstrated genetically that ablation of Aldh1b1 completely abrogates tumor development in a mouse model of KrasG12D-induced pancreatic cancer.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/metabolismo , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/patología , Mutación , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Células Madre/patología , Familia de Aldehído Deshidrogenasa 1/genética , Aldehído Deshidrogenasa Mitocondrial/genética , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Células Madre/metabolismo
7.
Stem Cells ; 37(5): 640-651, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30681750

RESUMEN

Understanding the mechanisms that promote the specification of pancreas progenitors and regulate their self-renewal and differentiation will help to maintain and expand pancreas progenitor cells derived from human pluripotent stem (hPS) cells. This will improve the efficiency of current differentiation protocols of hPS cells into ß-cells and bring such cells closer to clinical applications for the therapy of diabetes. Aldehyde dehydrogenase 1b1 (Aldh1b1) is a mitochondrial enzyme expressed specifically in progenitor cells during mouse pancreas development, and we have shown that its functional inactivation leads to accelerated differentiation and deficient ß-cells. In this report, we aimed to identify small molecule inducers of Aldh1b1 expression taking advantage of a mouse embryonic stem (mES) cell Aldh1b1 lacZ reporter line and a pancreas differentiation protocol directing mES cells into pancreatic progenitors. We identified AMI-5, a protein methyltransferase inhibitor, as an Aldh1b1 inducer and showed that it can maintain Aldh1b1 expression in embryonic pancreas explants. This led to a selective reduction in endocrine specification. This effect was due to a downregulation of Ngn3, and it was mediated through Aldh1b1 since the effect was abolished in Aldh1b1 null pancreata. The findings implicated methyltransferase activity in the regulation of endocrine differentiation and showed that methyltransferases can act through specific regulators during pancreas differentiation. Stem Cells 2019;37:640-651.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/genética , Aldehído Deshidrogenasa Mitocondrial/genética , Diferenciación Celular/genética , Diabetes Mellitus/terapia , Células Madre Pluripotentes/trasplante , Proteína Metiltransferasas/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Benzoatos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/enzimología , Proteínas del Tejido Nervioso/genética , Páncreas/efectos de los fármacos , Páncreas/crecimiento & desarrollo , Proteína Metiltransferasas/antagonistas & inhibidores , Xantenos/farmacología
8.
PLoS Biol ; 15(3): e2000949, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28248965

RESUMEN

During development, progenitor expansion, lineage allocation, and implementation of differentiation programs need to be tightly coordinated so that different cell types are generated in the correct numbers for appropriate tissue size and function. Pancreatic dysfunction results in some of the most debilitating and fatal diseases, including pancreatic cancer and diabetes. Several transcription factors regulating pancreas lineage specification have been identified, and Notch signalling has been implicated in lineage allocation, but it remains unclear how these processes are coordinated. Using a combination of genetic approaches, organotypic cultures of embryonic pancreata, and genomics, we found that sphingosine-1-phosphate (S1p), signalling through the G protein coupled receptor (GPCR) S1pr2, plays a key role in pancreas development linking lineage allocation and specification. S1pr2 signalling promotes progenitor survival as well as acinar and endocrine specification. S1pr2-mediated stabilisation of the yes-associated protein (YAP) is essential for endocrine specification, thus linking a regulator of progenitor growth with specification. YAP stabilisation and endocrine cell specification rely on Gαi subunits, revealing an unexpected specificity of selected GPCR intracellular signalling components. Finally, we found that S1pr2 signalling posttranscriptionally attenuates Notch signalling levels, thus regulating lineage allocation. Both S1pr2-mediated YAP stabilisation and Notch attenuation are necessary for the specification of the endocrine lineage. These findings identify S1p signalling as a novel key pathway coordinating cell survival, lineage allocation, and specification and linking these processes by regulating YAP levels and Notch signalling. Understanding lineage allocation and specification in the pancreas will shed light in the origins of pancreatic diseases and may suggest novel therapeutic approaches.


Asunto(s)
Linaje de la Célula , Lisofosfolípidos/metabolismo , Páncreas/citología , Transducción de Señal , Esfingosina/análogos & derivados , Células Acinares/citología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Tipificación del Cuerpo , Proteínas de Ciclo Celular , Diferenciación Celular , Supervivencia Celular , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Ratones , Modelos Biológicos , Fosfoproteínas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Subunidades de Proteína/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Receptores Notch/metabolismo , Esfingosina/metabolismo , Células Madre/citología , Proteínas Señalizadoras YAP
9.
Diabetologia ; 59(1): 139-150, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26518685

RESUMEN

AIMS/HYPOTHESIS: Pancreatic beta cells maintain glucose homeostasis and beta cell dysfunction is a major risk factor in developing diabetes. Therefore, understanding the developmental regulatory networks that define a fully functional beta cell is important for elucidating the genetic origins of the disease. Aldehyde dehydrogenase activity has been associated with stem/progenitor cells and we have previously shown that Aldh1b1 is specifically expressed in pancreas progenitor pools. Here we address the hypothesis that Aldh1b1 may regulate the timing of the appearance and eventual functionality of beta cells. METHODS: We generated an Aldh1b1-knockout mouse line (Aldh1b1 (tm1lacZ)) and used this to study pancreatic development, beta cell functionality and glucose homeostasis in the absence of Aldh1b1 function. RESULTS: Differentiation in the developing pancreas of Aldh1b1 (tm1lacZ) null mice was accelerated. Transcriptome analyses of newborn and adult islets showed misregulation of key beta cell transcription factors and genes crucial for beta cell function. Functional analyses showed that glucose-stimulated insulin secretion was severely compromised in islets isolated from null mice. Several key features of beta cell functionality were affected, including control of oxidative stress, glucose sensing, stimulus-coupling secretion and secretory granule biogenesis. As a result of beta cell dysfunction, homozygous mice developed glucose intolerance and age-dependent hyperglycaemia. CONCLUSIONS/INTERPRETATION: These findings show that Aldh1b1 influences the timing of the transition from the pancreas endocrine progenitor to the committed beta cell and demonstrate that changes in the timing of this transition lead to beta cell dysfunction and thus constitute a diabetes risk factor later in life. Gene Expression Omnibus (GEO) accession: GSE58025.


Asunto(s)
Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/fisiología , Células Secretoras de Insulina/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Aldehído Deshidrogenasa Mitocondrial , Alelos , Animales , Glucemia/análisis , Diferenciación Celular , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucógeno/metabolismo , Homeostasis , Hiperglucemia/metabolismo , Islotes Pancreáticos/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Riesgo , Células Madre/citología , Transcriptoma
10.
Mol Pharmacol ; 84(3): 346-52, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23778362

RESUMEN

G protein-coupled receptors (GPCRs) transduce many important physiological signals and are targets for a large fraction of therapeutic drugs. Members of the largest family of GPCRs (family A) are thought to self-associate as dimers and higher-order oligomers, although the significance of such quaternary structures for signaling or receptor trafficking is known for only a few examples. One outstanding question is the physical stability of family A oligomers in cell membranes. Stable oligomers would be expected to move through cellular compartments and membrane domains as intact groups of protomers. Here, we test this prediction by recruiting subsets of affinity-tagged family A protomers into artificial microdomains on the surface of living cells and asking if untagged protomers move into these domains (are corecruited) at the same time. We find that tagged ß2 adrenergic and µ-opioid protomers are unable to corecruit untagged protomers into microdomains. In contrast, tagged metabotropic glutamate receptor protomers do corecruit untagged protomers into such microdomains, which is consistent with the known covalent mechanism whereby these family C receptors dimerize. These observations suggest that interactions between these family A protomers are too weak to directly influence subcellular location, and that mechanisms that move these receptors between subcellular compartments and domains must operate on individual protomers.


Asunto(s)
Membrana Celular/metabolismo , Regiones Promotoras Genéticas , Receptores Adrenérgicos beta 2/metabolismo , Receptores Opioides mu/metabolismo , Animales , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Células HEK293 , Humanos , Microdominios de Membrana/metabolismo , Multimerización de Proteína , Transporte de Proteínas , Receptores Adrenérgicos beta 2/genética , Receptores Opioides mu/genética
11.
Dev Biol ; 374(1): 153-63, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23142317

RESUMEN

Aldehyde dehydrogenase (ALDH) genes are increasingly associated with stem/progenitor cell status but their role in the maintenance of pluripotency remains uncertain. In a screen conducted for downstream Ngn3 target genes using ES derived pancreas progenitors we identified Aldh1b1, encoding a mitochondrial enzyme, as one of the genes strongly up regulated in response to Ngn3 expression. We found both by in situ hybridization and immunofluorescence using a specific antibody that ALDH1B1 is exclusively expressed in the emerging pancreatic buds of the early embryo (9.5 dpc) in a Pdx1 dependent manner. Around the time of secondary transition, ALDH1B1 expression was restricted in the tip tripotent progenitors of the branching epithelium and in a subset of the trunk epithelium. Expression in the latter was Ngn3 dependent. Subsequently, ALDH1B1 expression persisted only in the tip cells that become restricted to the exocrine lineage and declined rapidly as these cells mature. In the adult pancreas we identified rare ALDH1B1(+) cells that become abundant following pancreas injury in either the caerulein or streptozotocin paradigms. Blocking ALDH catalytic activity in pancreas embryonic explants resulted in reduced size of the explants and accelerated differentiation suggesting for the first time that ALDH activity may be necessary in the developing pancreas for the maintenance and expansion of progenitor pools.


Asunto(s)
Aldehído Deshidrogenasa/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Páncreas/embriología , Aldehído Deshidrogenasa/genética , Familia de Aldehído Deshidrogenasa 1 , Aldehído Deshidrogenasa Mitocondrial , Animales , Catálisis , Genotipo , Hibridación in Situ , Ratones , Microscopía Fluorescente/métodos , Mutación , Oligonucleótidos Antisentido/genética , Células Madre/citología , Factores de Tiempo , Regulación hacia Arriba
12.
Mol Cell Biol ; 31(22): 4442-53, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21911471

RESUMEN

During development pancreatic endocrine cells migrate in a coordinated fashion. This migration is necessary to form fully functional islets, but the mechanisms involved remain unknown. Therapeutic strategies to restore ß-cell mass and islet functionality by reprogramming endogenous exocrine cells would be strengthened from simultaneous treatments that enhance endocrine cell clustering. We found that endocrine progenitors respond to and regulate G protein-coupled receptor (GPCR) signaling in order to cluster in islets. Rgs4, a dedicated regulator of GPCR signaling, was specifically expressed in early epithelial endocrine progenitors of both zebrafish and mouse, and its expression in the mouse endocrine progenitors was strictly dependent upon Ngn3, the key specification gene of the endocrine lineage. Rgs4 loss of function resulted in defects in islet cell aggregation. By genetically inactivating Gα(i)-mediated GPCR signaling in endocrine progenitors, we established its role in islet cell aggregation in both mouse and zebrafish. Finally, we identified sphingosine-1-phosphate (S1P) as a ligand mediating islet cell aggregation in both species acting through distinct but closely related receptors.


Asunto(s)
Islotes Pancreáticos/embriología , Lisofosfolípidos/metabolismo , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Esfingosina/análogos & derivados , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Línea Celular , Movimiento Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Transgénicos , Morfogénesis/genética , Proteínas del Tejido Nervioso/metabolismo , Organogénesis/genética , Filogenia , Proteínas RGS/biosíntesis , Proteínas RGS/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Esfingosina/metabolismo , Células Madre/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
13.
PLoS One ; 6(5): e20197, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21637844

RESUMEN

The evolutionarily conserved Hox family of homeodomain transcription factors plays fundamental roles in regulating cell specification along the anterior posterior axis during development of all bilaterian animals by controlling cell fate choices in a highly localized, extracellular signal and cell context dependent manner. Some studies have established downstream target genes in specific systems but their identification is insufficient to explain either the ability of Hox genes to direct homeotic transformations or the breadth of their patterning potential. To begin delineating Hox gene function in neural development we used a mouse ES cell based system that combines efficient neural differentiation with inducible Hoxb1 expression. Gene expression profiling suggested that Hoxb1 acted as both activator and repressor in the short term but predominantly as a repressor in the long run. Activated and repressed genes segregated in distinct processes suggesting that, in the context examined, Hoxb1 blocked differentiation while activating genes related to early developmental processes, wnt and cell surface receptor linked signal transduction and cell-to-cell communication. To further elucidate aspects of Hoxb1 function we used loss and gain of function approaches in the mouse and chick embryos. We show that Hoxb1 acts as an activator to establish the full expression domain of CRABPI and II in rhombomere 4 and as a repressor to restrict expression of Lhx5 and Lhx9. Thus the Hoxb1 patterning activity includes the regulation of the cellular response to retinoic acid and the delay of the expression of genes that commit cells to neural differentiation. The results of this study show that ES neural differentiation and inducible Hox gene expression can be used as a sensitive model system to systematically identify Hox novel target genes, delineate their interactions with signaling pathways in dictating cell fate and define the extent of functional overlap among different Hox genes.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Proteínas de Homeodominio/metabolismo , Neuronas/citología , Animales , Diferenciación Celular/efectos de los fármacos , Embrión de Pollo , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rombencéfalo/efectos de los fármacos , Rombencéfalo/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Tretinoina/farmacología
14.
Stem Cells ; 29(5): 858-70, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21433221

RESUMEN

Hox genes play a central role in neural crest (NC) patterning particularly in the cranial region of the body. Despite evidence that simultaneous loss of Hoxa1 and Hoxb1 function resulted in NC specification defects, the role of Hox genes in NC specification has remained unclear due to extended genetic redundancy among Hox genes. To circumvent this problem, we expressed anterior Hox genes in the trunk neural tube of the developing chick embryo. This demonstrated that anterior Hox genes play a central role in NC cell specification by rapidly inducing the key transcription factors Snail2 and Msx1/2 and a neural progenitor to NC cell fate switch characterized by cell adhesion changes and an epithelial-to-mesenchymal transition (EMT). Cells delaminated from dorsal and medial neural tube levels and generated ectopic neurons, glia progenitors, and melanocytes. The mobilization of the NC genetic cascade was dependent upon bone morphogenetic protein signaling and optimal levels of Notch signaling. Therefore, anterior Hox patterning genes participate in NC specification and EMT by interacting with NC-inducing signaling pathways and regulating the expression of key genes involved in these processes.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/fisiología , Embrión de Pollo , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Hibridación in Situ , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Ratones , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
PLoS One ; 5(8): e12488, 2010 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-20824215

RESUMEN

BACKGROUND: Present technology uses mostly chimeric proteins as regulators and hormones or antibiotics as signals to induce spatial and temporal gene expression. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that a chromosomally integrated yeast 'Leu3p-alpha-IotaRhoMu' system constitutes a ligand-inducible regulatory "off-on" genetic switch with an extensively dynamic action area. We find that Leu3p acts as an active transcriptional repressor in the absence and as an activator in the presence of alpha-isopropylmalate (alpha-IotaRhoMu) in primary fibroblasts isolated from double transgenic mouse embryos bearing ubiquitously expressing Leu3p and a Leu3p regulated GFP reporter. In the absence of the branched amino acid biosynthetic pathway in animals, metabolically stable alpha-IPM presents an EC(50) equal to 0.8837 mM and fast "OFF-ON" kinetics (t(50)ON = 43 min, t(50)OFF = 2.18 h), it enters the cells via passive diffusion, while it is non-toxic to mammalian cells and to fertilized mouse eggs cultured ex vivo. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that the 'Leu3p-alpha-IotaRhoMu' constitutes a simpler and safer system for inducible gene expression in biomedical applications.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Ingeniería Genética/métodos , Malatos/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Animales , Secuencia de Bases , Femenino , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Embarazo , Saccharomyces cerevisiae/genética
16.
Stem Cells ; 26(8): 1985-97, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18499896

RESUMEN

The directed differentiation of embryonic stem cells (ESCs) into neural stem cells (NSCs) of specific identities and the identification of endogenous pathways that may mediate expansion of NSCs are fundamental goals for the treatment of degenerative disorders and trauma of the nervous system. We report that timely induction of a Hoxb1 transgene in ESC-derived NSCs resulted in the specification of NSCs toward a hindbrain-specific identity through the activation of a rhombomere 4-specific genetic program and the repression of anterior neural identity. This change was accompanied by changes in signaling pathways that pattern the dorsoventral (DV) axis of the nervous system and concomitant changes in the expression of DV neural progenitor markers. Furthermore, Hoxb1 mediated the maintenance and expansion of posterior neural progenitor cells. Hoxb1(+) cells kept proliferating upon mitogen withdrawal and became transiently amplifying progenitors instead of terminally differentiating. This was partially attributed to Hoxb1-dependent activation of the Notch signaling pathway and Notch-dependent STAT3 phosphorylation at Ser 727, thus linking Hox gene function with maintenance of active Notch signaling and the JAK/STAT pathway. Thus, timely expression of specific Hox genes could be used to establish NSCs and neural progenitors of distinct posterior identities. ESC-derived NSCs have a mixed DV identity that is subject to regulation by Hox genes. Finally, these findings set the stage for the elucidation of molecular pathways involved in the expansion of posterior NSCs and neural progenitors. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Neuronas/citología , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
17.
Stem Cells ; 26(1): 3-16, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17932425

RESUMEN

The delineation of regulatory networks involved in early endocrine pancreas specification will play a crucial role in directing the differentiation of embryonic stem cells toward the mature phenotype of beta cells for cell therapy of type 1 diabetes. The transcription factor Ngn3 is required for the specification of the endocrine lineage, but its direct targets and the scope of biological processes it regulates remain elusive. We show that stepwise differentiation of embryonic stem cells using successive in vivo patterning signals can lead to simultaneous induction of Ptf1a and Pdx1 expression. In this cellular context, Ngn3 induction results in upregulation of its known direct target genes within 12 hours. Microarray gene expression profiling at distinct time points following Ngn3 induction suggested novel and diverse roles of Ngn3 in pancreas endocrine cell specification. Induction of Ngn3 expression results in regulation of the Wnt, integrin, Notch, and transforming growth factor beta signaling pathways and changes in biological processes affecting cell motility, adhesion, the cytoskeleton, the extracellular matrix, and gene expression. Furthermore, the combination of in vivo patterning signals and inducible Ngn3 expression enhances ESC differentiation toward the pancreas endocrine lineage. This is shown by strong upregulation of endocrine lineage terminal differentiation markers and strong expression of the hormones glucagon, somatostatin, and insulin. Importantly, all insulin(+) cells are also C-peptide(+), and glucose-dependent insulin release was 10-fold higher than basal levels. These data suggest that bona fide pancreas endocrine cells have been generated and that timely induction of Ngn3 expression can play a decisive role in directing ESC differentiation toward the endocrine lineage.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Islotes Pancreáticos/embriología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Vectores Genéticos , Proteínas de Homeodominio/biosíntesis , Islotes Pancreáticos/citología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis , Transgenes
18.
Nat Neurosci ; 10(11): 1433-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17922007

RESUMEN

In many regions of the developing CNS, distinct cell types are born at different times. The means by which discrete and stereotyped temporal switches in cellular identities are acquired remains poorly understood. To address this, we have examined how visceral motor neurons (VMNs) and serotonergic neurons, two neuronal subtypes, are sequentially generated from a common progenitor pool in the vertebrate hindbrain. We found that the forkhead transcription factor Foxa2, acting in progenitors, is essential for the transition from VMN to serotonergic neurogenesis. Loss-of-function and gain-of-function experiments indicated that Foxa2 activates the switch through a temporal cross-repressive interaction with paired-like homeobox 2b (Phox2b), the VMN progenitor determinant. This mechanism bears a marked resemblance to the cross-repression between neighboring domains of transcription factors that establish discrete progenitor identities along the spatial axes. Moreover, the subsequent differentiation of central serotonergic neurons required both the suppression of VMN neurogenesis and the induction of downstream intrinsic determinants of serotonergic identity by Foxa2.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/fisiología , Serotonina/metabolismo , Células Madre/fisiología , Factores de Transcripción/fisiología , Factores de Edad , Animales , Tipificación del Cuerpo/fisiología , Bromodesoxiuridina/metabolismo , Embrión de Pollo , Electroporación/métodos , Embrión de Mamíferos , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Mutantes , Neuronas/citología , Rombencéfalo/citología , Rombencéfalo/embriología , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología
19.
Mol Cell Biol ; 24(19): 8567-75, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15367676

RESUMEN

Homeodomain containing transcription factors of the Hox family play critical roles in patterning the anteroposterior embryonic body axis, as well as in controlling several steps of organogenesis. Several Hox proteins have been shown to cooperate with members of the Pbx family for the recognition and activation of identified target enhancers. Hox proteins contact Pbx via a conserved hexapeptide motif. Previous biochemical studies provided evidence that critical amino acid substitutions in the hexapeptide sequence of Hoxa1 abolish its interaction with Pbx. As a result, these substitutions also abolish Hoxa1 activity on known target enhancers in cellular models, suggesting that Hoxa1 activity relies on its capacity to interact with Pbx. Here, we show that mice with mutations in the Hoxa1 hexapeptide display hindbrain, cranial nerve, and skeletal defects highly reminiscent of those reported for the Hoxa1 loss of function. Since similar hexapeptide mutations in the mouse Hoxb8 and the Drosophila AbdA proteins result in activity modulation and gain of function, our data demonstrate that the functional importance of the hexapeptide in vivo differs according to the Hox proteins.


Asunto(s)
Proteínas de Homeodominio/genética , Fragmentos de Péptidos/genética , Factores de Transcripción/genética , Sustitución de Aminoácidos , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Nervios Craneales/embriología , Oído/anomalías , Oído/embriología , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Mutación , Cresta Neural/embriología , Hueso Occipital/anomalías , Hueso Occipital/embriología , Fragmentos de Péptidos/metabolismo , Rombencéfalo/embriología , Factores de Transcripción/metabolismo
20.
Development ; 130(23): 5663-79, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14522873

RESUMEN

Hox genes are instrumental in assigning segmental identity in the developing hindbrain. Auto-, cross- and para-regulatory interactions help establish and maintain their expression. To understand to what extent such regulatory interactions shape neuronal patterning in the hindbrain, we analysed neurogenesis, neuronal differentiation and motoneuron migration in Hoxa1, Hoxb1 and Hoxb2 mutant mice. This comparison revealed that neurogenesis and differentiation of specific neuronal subpopulations in r4 was impaired in a similar fashion in all three mutants, but with different degrees of severity. In the Hoxb1 mutants, neurons derived from the presumptive r4 territory were re-specified towards an r2-like identity. Motoneurons derived from that territory resembled trigeminal motoneurons in both their migration patterns and the expression of molecular markers. Both migrating motoneurons and the resident territory underwent changes consistent with a switch from an r4 to r2 identity. Abnormally migrating motoneurons initially formed ectopic nuclei that were subsequently cleared. Their survival could be prolonged through the introduction of a block in the apoptotic pathway. The Hoxa1 mutant phenotype is consistent with a partial misspecification of the presumptive r4 territory that results from partial Hoxb1 activation. The Hoxb2 mutant phenotype is a hypomorph of the Hoxb1 mutant phenotype, consistent with the overlapping roles of these genes in facial motoneuron specification. Therefore, we have delineated the functional requirements in hindbrain neuronal patterning that follow the establishment of the genetic regulatory hierarchy between Hoxa1, Hoxb1 and Hoxb2.


Asunto(s)
Tipificación del Cuerpo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , Rombencéfalo/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Movimiento Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Factor de Transcripción GATA2 , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Rombencéfalo/anomalías , Rombencéfalo/citología , Rombencéfalo/fisiología , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...