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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 144(12): 947-960, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34264749

RESUMEN

BACKGROUND: Ischemia-reperfusion injury (IRI) is one of the major risk factors implicated in morbidity and mortality associated with cardiovascular disease. During cardiac ischemia, the buildup of acidic metabolites results in decreased intracellular and extracellular pH, which can reach as low as 6.0 to 6.5. The resulting tissue acidosis exacerbates ischemic injury and significantly affects cardiac function. METHODS: We used genetic and pharmacologic methods to investigate the role of acid-sensing ion channel 1a (ASIC1a) in cardiac IRI at the cellular and whole-organ level. Human induced pluripotent stem cell-derived cardiomyocytes as well as ex vivo and in vivo models of IRI were used to test the efficacy of ASIC1a inhibitors as pre- and postconditioning therapeutic agents. RESULTS: Analysis of human complex trait genetics indicates that variants in the ASIC1 genetic locus are significantly associated with cardiac and cerebrovascular ischemic injuries. Using human induced pluripotent stem cell-derived cardiomyocytes in vitro and murine ex vivo heart models, we demonstrate that genetic ablation of ASIC1a improves cardiomyocyte viability after acute IRI. Therapeutic blockade of ASIC1a using specific and potent pharmacologic inhibitors recapitulates this cardioprotective effect. We used an in vivo model of myocardial infarction and 2 models of ex vivo donor heart procurement and storage as clinical models to show that ASIC1a inhibition improves post-IRI cardiac viability. Use of ASIC1a inhibitors as preconditioning or postconditioning agents provided equivalent cardioprotection to benchmark drugs, including the sodium-hydrogen exchange inhibitor zoniporide. At the cellular and whole organ level, we show that acute exposure to ASIC1a inhibitors has no effect on cardiac ion channels regulating baseline electromechanical coupling and physiologic performance. CONCLUSIONS: Our data provide compelling evidence for a novel pharmacologic strategy involving ASIC1a blockade as a cardioprotective therapy to improve the viability of hearts subjected to IRI.


Asunto(s)
Canales Iónicos Sensibles al Ácido/biosíntesis , Canales Iónicos Sensibles al Ácido/genética , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Preparación de Corazón Aislado/métodos , Masculino , Ratones , Ratones Noqueados , Isquemia Miocárdica/terapia , Daño por Reperfusión Miocárdica/terapia , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Polimorfismo de Nucleótido Simple/fisiología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Venenos de Araña/farmacología
2.
Genome Res ; 28(7): 1053-1066, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29752298

RESUMEN

Heterogeneity of cell states represented in pluripotent cultures has not been described at the transcriptional level. Since gene expression is highly heterogeneous between cells, single-cell RNA sequencing can be used to identify how individual pluripotent cells function. Here, we present results from the analysis of single-cell RNA sequencing data from 18,787 individual WTC-CRISPRi human induced pluripotent stem cells. We developed an unsupervised clustering method and, through this, identified four subpopulations distinguishable on the basis of their pluripotent state, including a core pluripotent population (48.3%), proliferative (47.8%), early primed for differentiation (2.8%), and late primed for differentiation (1.1%). For each subpopulation, we were able to identify the genes and pathways that define differences in pluripotent cell states. Our method identified four transcriptionally distinct predictor gene sets composed of 165 unique genes that denote the specific pluripotency states; using these sets, we developed a multigenic machine learning prediction method to accurately classify single cells into each of the subpopulations. Compared against a set of established pluripotency markers, our method increases prediction accuracy by 10%, specificity by 20%, and explains a substantially larger proportion of deviance (up to threefold) from the prediction model. Finally, we developed an innovative method to predict cells transitioning between subpopulations and support our conclusions with results from two orthogonal pseudotime trajectory methods.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , ARN/genética , Diferenciación Celular/genética , Línea Celular , Análisis por Conglomerados , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Expresión Génica/genética , Heterogeneidad Genética , Marcadores Genéticos/genética , Humanos , Análisis de Secuencia de ARN/métodos , Transcripción Genética/genética
3.
Dev Cell ; 59(1): 91-107.e6, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38091997

RESUMEN

Genomic regulation of cardiomyocyte differentiation is central to heart development and function. This study uses genetic loss-of-function human-induced pluripotent stem cell-derived cardiomyocytes to evaluate the genomic regulatory basis of the non-DNA-binding homeodomain protein HOPX. We show that HOPX interacts with and controls cardiac genes and enhancer networks associated with diverse aspects of heart development. Using perturbation studies in vitro, we define how upstream cell growth and proliferation control HOPX transcription to regulate cardiac gene programs. We then use cell, organoid, and zebrafish regeneration models to demonstrate that HOPX-regulated gene programs control cardiomyocyte function in development and disease. Collectively, this study mechanistically links cell signaling pathways as upstream regulators of HOPX transcription to control gene programs underpinning cardiomyocyte identity and function.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Animales , Humanos , Miocitos Cardíacos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Pez Cebra/metabolismo , Diferenciación Celular/genética , Proliferación Celular
4.
Stem Cell Reports ; 18(1): 159-174, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36493778

RESUMEN

Vascular endothelial cells are a mesoderm-derived lineage with many essential functions, including angiogenesis and coagulation. The gene-regulatory mechanisms underpinning endothelial specialization are largely unknown, as are the roles of chromatin organization in regulating endothelial cell transcription. To investigate the relationships between chromatin organization and gene expression, we induced endothelial cell differentiation from human pluripotent stem cells and performed Hi-C and RNA-sequencing assays at specific time points. Long-range intrachromosomal contacts increase over the course of differentiation, accompanied by widespread heteroeuchromatic compartment transitions that are tightly associated with transcription. Dynamic topologically associating domain boundaries strengthen and converge on an endothelial cell state, and function to regulate gene expression. Chromatin pairwise point interactions (DNA loops) increase in frequency during differentiation and are linked to the expression of genes essential to vascular biology. Chromatin dynamics guide transcription in endothelial cell development and promote the divergence of endothelial cells from cardiomyocytes.


Asunto(s)
Cromatina , Células Endoteliales , Humanos , Diferenciación Celular/genética , Regulación de la Expresión Génica
5.
Nat Commun ; 14(1): 3240, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37296104

RESUMEN

The mechanisms by which DNA alleles contribute to disease risk, drug response, and other human phenotypes are highly context-specific, varying across cell types and different conditions. Human induced pluripotent stem cells are uniquely suited to study these context-dependent effects but cell lines from hundreds or thousands of individuals are required. Village cultures, where multiple induced pluripotent stem lines are cultured and differentiated in a single dish, provide an elegant solution for scaling induced pluripotent stem experiments to the necessary sample sizes required for population-scale studies. Here, we show the utility of village models, demonstrating how cells can be assigned to an induced pluripotent stem line using single-cell sequencing and illustrating that the genetic, epigenetic or induced pluripotent stem line-specific effects explain a large percentage of gene expression variation for many genes. We demonstrate that village methods can effectively detect induced pluripotent stem line-specific effects, including sensitive dynamics of cell states.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Línea Celular , Diferenciación Celular/genética , Fenotipo
6.
Genesis ; 50(9): 711-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22511315

RESUMEN

Crim1 is a developmentally expressed, transmembrane protein essential for normal embryonic development. We generated mice engineered to contain a Crim1 conditional null allele by flanking exons three and four of Crim1 with unidirectional LoxP sites. After crossing Crim1+/FLOX mice with a CMV-Cre line, a Crim1+/Δflox colony was established after germline transmission of the deleted allele. We then analyzed genomic DNA, mRNA transcripts, and protein expression from Crim1Δflox/Δflox null mice to confirm the nature of the genomic lesion. Crim1Δflox/Δflox mice displayed phenotypes similar to those previously described for a Crim1 gene-trap mutant, Crim1KST264/KST264, including perinatal lethality, digit syndactyly, eye, and kidney abnormalities, with varying penetrance and severity. The production of a conditional mutant allele represents a valuable resource for the study of the tissue-specific roles for Crim1, and for understanding the pleimorphic phenotypes associated with Crim1 mutation.


Asunto(s)
Anomalías Múltiples/embriología , Receptores de Proteínas Morfogenéticas Óseas/genética , Desarrollo Embrionario/genética , Ingeniería Genética/métodos , Anomalías Múltiples/genética , Alelos , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Quimera , Cruzamientos Genéticos , Exones , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Genotipo , Integrasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Especificidad de Órganos , Organogénesis/genética , Fenotipo , Embarazo , Recombinación Genética
7.
Dev Biol ; 344(2): 1071-87, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20510229

RESUMEN

Here we describe the first detailed catalog of gene expression in the developing lower urinary tract (LUT), including epithelial and mesenchymal portions of the developing bladder, urogenital sinus, urethra, and genital tubercle (GT) at E13 and E14. Top compartment-specific genes implicated by the microarray data were validated using whole-mount in situ hybridization (ISH) over the entire LUT. To demonstrate the potential of this resource to implicate developmentally critical features, we focused on gene expression patterns and pathways in the sexually indeterminate, androgen-independent GT. GT expression patterns reinforced the proposed similarities between development of GT, limb, and craniofacial prominences. Comparison of spatial expression patterns predicted a network of Wnt7a-associated GT-enriched epithelial genes, including Gjb2, Dsc3, Krt5, and Sostdc1. Known from other contexts, these genes are associated with normal epidermal differentiation, with disruptions in Dsc3 and Gjb2 showing palmo-plantar keratoderma in the limb. We propose that this gene network contributes to normal foreskin, scrotum, and labial development. As several of these genes are known to be regulated by, or contain cis elements responsive to retinoic acid, estrogen, or androgen, this implicates this pathway in the later androgen-dependent development of the GT.


Asunto(s)
Expresión Génica , Redes Reguladoras de Genes , Sistema Urogenital/embriología , Andrógenos/genética , Animales , Diferenciación Celular/genética , Embrión de Mamíferos , Epidermis , Extremidades , Genitales Masculinos/embriología , Masculino , Ratones , Organogénesis/genética , Uretra/embriología
8.
J Nanosci Nanotechnol ; 11(12): 10633-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22408963

RESUMEN

The U.S. Food and Drug Administration allows a maximum of 72 mg of caffeine per 12 oz. serving (6 mg/oz). Consuming 400 mg of caffeine 3 times daily for 7 days may develop sleep disruption effects. However, it is still very hard for people to estimate how much caffeine is intake daily. Moreover, (-)epigallocatechin gallate (EGCG) is studied a potent antioxidant that may have therapeutic properties for anti-aging and cancer. Conventionally, both caffeine and EGCG could be measured by the protocols of high performance liquid chromatography; however, high precision instruments are required. In this work, the caffeine and EGCG are used as the template molecules and imprinted into poly(ethylene-co-vinyl alcohol), EVAL, via solvent evaporation. The EVAL membrane is then used as the sensing element for electrochemical analysis after templates removal. From the cyclic voltammetry measurement of the caffeine, the peak oxidation potential is shifted from 0.36 to 0.46 V when the final concentration of caffeine is from 0.01 to 1 mg/mL, and the highest current density is about 0.18 microA/cm2. The caffeine and EGCG concentrations measured in three real samples are about 0.10-0.13 mg/mL and 0.49-1.74 mg/mL, respectively. This molecularly imprinted polymeric coated electrode is potential employed as a home-care system.


Asunto(s)
Cafeína/análisis , Catequina/análogos & derivados , Electroquímica/instrumentación , Polivinilos/química , Catequina/análisis
9.
Genome Biol ; 22(1): 76, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33673841

RESUMEN

BACKGROUND: The discovery that somatic cells can be reprogrammed to induced pluripotent stem cells (iPSCs) has provided a foundation for in vitro human disease modelling, drug development and population genetics studies. Gene expression plays a critical role in complex disease risk and therapeutic response. However, while the genetic background of reprogrammed cell lines has been shown to strongly influence gene expression, the effect has not been evaluated at the level of individual cells which would provide significant resolution. By integrating single cell RNA-sequencing (scRNA-seq) and population genetics, we apply a framework in which to evaluate cell type-specific effects of genetic variation on gene expression. RESULTS: Here, we perform scRNA-seq on 64,018 fibroblasts from 79 donors and map expression quantitative trait loci (eQTLs) at the level of individual cell types. We demonstrate that the majority of eQTLs detected in fibroblasts are specific to an individual cell subtype. To address if the allelic effects on gene expression are maintained following cell reprogramming, we generate scRNA-seq data in 19,967 iPSCs from 31 reprogramed donor lines. We again identify highly cell type-specific eQTLs in iPSCs and show that the eQTLs in fibroblasts almost entirely disappear during reprogramming. CONCLUSIONS: This work provides an atlas of how genetic variation influences gene expression across cell subtypes and provides evidence for patterns of genetic architecture that lead to cell type-specific eQTL effects.


Asunto(s)
Reprogramación Celular/genética , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Sitios de Carácter Cuantitativo , RNA-Seq/métodos , Análisis de la Célula Individual , Biología Computacional/métodos , Fibroblastos/citología , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Especificidad de Órganos/genética , Análisis de la Célula Individual/métodos
10.
Dev Biol ; 332(2): 273-86, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19501082

RESUMEN

While nephron formation is known to be initiated by a mesenchyme-to-epithelial transition of the cap mesenchyme to form a renal vesicle (RV), the subsequent patterning of the nephron and fusion with the ureteric component of the kidney to form a patent contiguous uriniferous tubule has not been fully characterized. Using dual section in situ hybridization (SISH)/immunohistochemistry (IHC) we have revealed distinct distal/proximal patterning of Notch, BMP and Wnt pathway components within the RV stage nephron. Quantitation of mitoses and Cyclin D1 expression indicated that cell proliferation was higher in the distal RV, reflecting the differential developmental programs of the proximal and distal populations. A small number of RV genes were also expressed in the early connecting segment of the nephron. Dual ISH/IHC combined with serial section immunofluorescence and 3D reconstruction revealed that fusion occurs between the late RV and adjacent ureteric tip via a process that involves loss of the intervening ureteric epithelial basement membrane and insertion of cells expressing RV markers into the ureteric tip. Using Six2-eGFPCre x R26R-lacZ mice, we demonstrate that these cells are derived from the cap mesenchyme and not the ureteric epithelium. Hence, both nephron patterning and patency are evident at the late renal vesicle stage.


Asunto(s)
Proliferación Celular , Riñón/anatomía & histología , Riñón/embriología , Mesodermo/fisiología , Morfogénesis/fisiología , Nefronas/embriología , Uréter , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Calbindinas , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Epitelio/fisiología , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Riñón/fisiología , Proteínas con Homeodominio LIM , Laminina/genética , Laminina/metabolismo , Ratones , Nefronas/anatomía & histología , Nefronas/fisiología , Embarazo , Receptores Notch/genética , Receptores Notch/metabolismo , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo , Factores de Transcripción , Uréter/anatomía & histología , Uréter/embriología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
11.
Genome Biol ; 20(1): 290, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856883

RESUMEN

A variety of methods have been developed to demultiplex pooled samples in a single cell RNA sequencing (scRNA-seq) experiment which either require hashtag barcodes or sample genotypes prior to pooling. We introduce scSplit which utilizes genetic differences inferred from scRNA-seq data alone to demultiplex pooled samples. scSplit also enables mapping clusters to original samples. Using simulated, merged, and pooled multi-individual datasets, we show that scSplit prediction is highly concordant with demuxlet predictions and is highly consistent with the known truth in cell-hashing dataset. scSplit is ideally suited to samples without external genotype information and is available at: https://github.com/jon-xu/scSplit.


Asunto(s)
Análisis de Secuencia de ARN , Análisis de la Célula Individual , Programas Informáticos , Humanos
12.
Cell Stem Cell ; 23(4): 586-598.e8, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30290179

RESUMEN

Cardiac differentiation of human pluripotent stem cells (hPSCs) requires orchestration of dynamic gene regulatory networks during stepwise fate transitions but often generates immature cell types that do not fully recapitulate properties of their adult counterparts, suggesting incomplete activation of key transcriptional networks. We performed extensive single-cell transcriptomic analyses to map fate choices and gene expression programs during cardiac differentiation of hPSCs and identified strategies to improve in vitro cardiomyocyte differentiation. Utilizing genetic gain- and loss-of-function approaches, we found that hypertrophic signaling is not effectively activated during monolayer-based cardiac differentiation, thereby preventing expression of HOPX and its activation of downstream genes that govern late stages of cardiomyocyte maturation. This study therefore provides a key transcriptional roadmap of in vitro cardiac differentiation at single-cell resolution, revealing fundamental mechanisms underlying heart development and differentiation of hPSC-derived cardiomyocytes.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Homeodominio/genética , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Análisis de la Célula Individual , Transcriptoma , Proteínas Supresoras de Tumor/genética , Animales , Células Cultivadas , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas Supresoras de Tumor/metabolismo
13.
J Mol Histol ; 48(1): 53-61, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27803996

RESUMEN

Endothelial cells form a critical component of the coronary vasculature, yet the factors regulating their development remain poorly defined. Here we reveal a novel role for the transmembrane protein CRIM1 in mediating cardiac endothelial cell development. In the absence of Crim1 in vivo, the coronary vasculature is malformed, the number of endothelial cells reduced, and the canonical BMP pathway dysregulated. Moreover, we reveal that CRIM1 can bind IGFs, and regulate IGF signalling within endothelial cells. Finally, loss of CRIM1 from human cardiac endothelial cells results in misregulation of endothelial genes, predicted by pathway analysis to be involved in an increased inflammatory response and cytolysis, reminiscent of endothelial cell dysfunction in cardiovascular disease pathogenesis. Collectively, these findings implicate CRIM1 in endothelial cell development and homeostasis in the coronary vasculature.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Vasos Coronarios/citología , Células Endoteliales/metabolismo , Homeostasis , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Supervivencia Celular/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Mutación , Transducción de Señal
14.
Sci Signal ; 10(508)2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-29208680

RESUMEN

Oxytocin and vasopressin mediate various physiological functions that are important for osmoregulation, reproduction, cardiovascular function, social behavior, memory, and learning through four G protein-coupled receptors that are also implicated in high-profile disorders. Targeting these receptors is challenging because of the difficulty in obtaining ligands that retain selectivity across rodents and humans for translational studies. We identified a selective and more stable oxytocin receptor (OTR) agonist by subtly modifying the pharmacophore framework of human oxytocin and vasopressin. [Se-Se]-oxytocin-OH displayed similar potency to oxytocin but improved selectivity for OTR, an effect that was retained in mice. Centrally infused [Se-Se]-oxytocin-OH potently reversed social fear in mice, confirming that this action was mediated by OTR and not by V1a or V1b vasopressin receptors. In addition, [Se-Se]-oxytocin-OH produced a more regular contraction pattern than did oxytocin in a preclinical labor induction and augmentation model using myometrial strips from cesarean sections. [Se-Se]-oxytocin-OH had no activity in human cardiomyocytes, indicating a potentially improved safety profile and therapeutic window compared to those of clinically used oxytocin. In conclusion, [Se-Se]-oxytocin-OH is a novel probe for validating OTR as a therapeutic target in various biological systems and is a promising new lead for therapeutic development. Our medicinal chemistry approach may also be applicable to other peptidergic signaling systems with similar selectivity issues.


Asunto(s)
Ansiedad/tratamiento farmacológico , Receptores de Oxitocina/agonistas , Animales , Células COS , Química Farmacéutica , Chlorocebus aethiops , Condicionamiento Psicológico/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Infusiones Intraventriculares , Ligandos , Masculino , Ratones , Ratas
15.
Sci Rep ; 6: 19832, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26821812

RESUMEN

The epicardium has a critical role during embryonic development, contributing epicardium-derived lineages to the heart, as well as providing regulatory and trophic signals necessary for myocardial development. Crim1 is a unique trans-membrane protein expressed by epicardial and epicardially-derived cells but its role in cardiogenesis is unknown. Using knockout mouse models, we observe that loss of Crim1 leads to congenital heart defects including epicardial defects and hypoplastic ventricular compact myocardium. Epicardium-restricted deletion of Crim1 results in increased epithelial-to-mesenchymal transition and invasion of the myocardium in vivo, and an increased migration of primary epicardial cells. Furthermore, Crim1 appears to be necessary for the proliferation of epicardium-derived cells (EPDCs) and for their subsequent differentiation into cardiac fibroblasts. It is also required for normal levels of cardiomyocyte proliferation and apoptosis, consistent with a role in regulating epicardium-derived trophic factors that act on the myocardium. Mechanistically, Crim1 may also modulate key developmentally expressed growth factors such as TGFßs, as changes in the downstream effectors phospho-SMAD2 and phospho-ERK1/2 are observed in the absence of Crim1. Collectively, our data demonstrates that Crim1 is essential for cell-autonomous and paracrine aspects of heart development.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Cardiopatías Congénitas/genética , Corazón/crecimiento & desarrollo , Organogénesis/genética , Animales , Diferenciación Celular/genética , Desarrollo Embrionario/genética , Transición Epitelial-Mesenquimal/genética , Cardiopatías Congénitas/patología , Humanos , Ratones , Ratones Noqueados , Miocardio/metabolismo , Comunicación Paracrina/genética , Pericardio/embriología , Proteína Smad2/genética , Factor de Crecimiento Transformador beta1/genética
16.
Methods Mol Biol ; 886: 223-39, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22639265

RESUMEN

Studies into the molecular basis of morphogenesis frequently begin with investigations into gene expression across time and cell type in that organ. One of the most anatomically informative approaches to such studies is the use of in situ hybridization, either of intact or histologically sectioned tissues. Here, we describe the optimization of this approach for use in the temporal and spatial analysis of gene expression in the urogenital system, from embryonic development to the postnatal period. The methods described are applicable for high throughput analysis of large gene sets. As such, ISH has become a powerful technique for gene expression profiling and is valuable for the validation of profiling analyses performed using other approaches such as microarrays.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ/métodos , ARN Mensajero/genética , Sistema Urogenital/crecimiento & desarrollo , Animales , Riñón/embriología , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Ratones , ARN Mensajero/aislamiento & purificación , Sistema Urogenital/embriología , Sistema Urogenital/metabolismo
17.
Front Neurosci ; 6: 130, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22988430

RESUMEN

Relative positions of neurons within mature murine pelvic ganglia based on expression of neurotransmitters have been described. However the spatial organization of developing innervation in the murine urogenital tract (UGT) and the gene networks that regulate specification and maturation of neurons within the pelvic ganglia of the lower urinary tract (LUT) are unknown. We used whole-mount immunohistochemistry and histochemical stains to localize neural elements in 15.5 days post coitus (dpc) fetal mice. To identify potential regulatory factors expressed in pelvic ganglia, we surveyed expression patterns for known or probable transcription factors (TF) annotated in the mouse genome by screening a whole-mount in situ hybridization library of fetal UGTs. Of the 155 genes detected in pelvic ganglia, 88 encode TFs based on the presence of predicted DNA-binding domains. Neural crest (NC)-derived progenitors within the LUT were labeled by Sox10, a well-known regulator of NC development. Genes identified were categorized based on patterns of restricted expression in pelvic ganglia, pelvic ganglia and urethral epithelium, or pelvic ganglia and urethral mesenchyme. Gene expression patterns and the distribution of Sox10+, Phox2b+, Hu+, and PGP9.5+ cells within developing ganglia suggest previously unrecognized regional segregation of Sox10+ progenitors and differentiating neurons in early development of pelvic ganglia. Reverse transcription-PCR of pelvic ganglia RNA from fetal and post-natal stages demonstrated that multiple TFs maintain post-natal expression, although Pax3 is extinguished before weaning. Our analysis identifies multiple potential regulatory genes including TFs that may participate in segregation of discrete lineages within pelvic ganglia. The genes identified here are attractive candidate disease genes that may now be further investigated for their roles in malformation syndromes or in LUT dysfunction.

18.
PLoS One ; 6(2): e17286, 2011 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-21386911

RESUMEN

The development of the mammalian kidney is well conserved from mouse to man. Despite considerable temporal and spatial data on gene expression in mammalian kidney development, primarily in rodent species, there is a paucity of genes whose expression is absolutely specific to a given anatomical compartment and/or developmental stage, defined here as 'anchor' genes. We previously generated an atlas of gene expression in the developing mouse kidney using microarray analysis of anatomical compartments collected via laser capture microdissection. Here, this data is further analysed to identify anchor genes via stringent bioinformatic filtering followed by high resolution section in situ hybridisation performed on 200 transcripts selected as specific to one of 11 anatomical compartments within the midgestation mouse kidney. A total of 37 anchor genes were identified across 6 compartments with the early proximal tubule being the compartment richest in anchor genes. Analysis of minimal and evolutionarily conserved promoter regions of this set of 25 anchor genes identified enrichment of transcription factor binding sites for Hnf4a and Hnf1b, RbpJ (Notch signalling), PPARγ:RxRA and COUP-TF family transcription factors. This was reinforced by GO analyses which also identified these anchor genes as targets in processes including epithelial proliferation and proximal tubular function. As well as defining anchor genes, this large scale validation of gene expression identified a further 92 compartment-enriched genes able to subcompartmentalise key processes during murine renal organogenesis spatially or ontologically. This included a cohort of 13 ureteric epithelial genes revealing previously unappreciated compartmentalisation of the collecting duct system and a series of early tubule genes suggesting that segmentation into proximal tubule, loop of Henle and distal tubule does not occur until the onset of glomerular vascularisation. Overall, this study serves to illuminate previously ill-defined stages of patterning and will enable further refinement of the lineage relationships within mammalian kidney development.


Asunto(s)
Compartimento Celular/genética , Regulación del Desarrollo de la Expresión Génica , Genes del Desarrollo/fisiología , Riñón/embriología , Organogénesis/genética , Animales , Análisis por Conglomerados , Perfilación de la Expresión Génica , Riñón/metabolismo , Ratones , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/genética , Transducción de Señal/fisiología , Distribución Tisular/genética , Estudios de Validación como Asunto
19.
Histochem Cell Biol ; 130(5): 927-42, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18618131

RESUMEN

The kidney is the most complex organ within the urogenital system. The adult mouse kidney contains in excess of 8,000 mature nephrons, each of which can be subdivided into a renal corpuscle and 14 distinct tubular segments. The histological complexity of this organ can make the clarification of the site of gene expression by in situ hybridisation difficult. We have defined a panel of seven antibodies capable of identifying the six stages of early nephron development, the tubular nephron segments and the components of the renal corpuscle within the embryonic and adult mouse kidney. We have analysed in detail the protein expression of Wt1, Calb1 Aqp1, Aqp2 and Umod using these antibodies. We have then coupled immunohistochemistry with RNA in situ hybridisation in order to precisely identify the expression pattern of different genes, including Wnt4, Umod and Spp1. This technique will be invaluable for examining at high resolution, the structure of both the developing and mature nephron where standard in situ hybridisation and histological techniques are insufficient. The use of this technique will enhance the expression analyses of genes which may be involved in nephron formation and the function of the mature nephron in the mouse.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Nefronas/química , ARN Mensajero/análisis , Animales , Acuaporina 1/análisis , Acuaporina 2/análisis , Calbindina 1 , Calbindinas , Proteínas de Unión al Calcio/análisis , Femenino , Masculino , Ratones , Mucoproteínas/análisis , Mucoproteínas/genética , Nefronas/embriología , Nefronas/crecimiento & desarrollo , Proteínas del Tejido Nervioso/análisis , Osteopontina/genética , Proteína G de Unión al Calcio S100 , Uromodulina , Proteínas WT1/análisis , Proteínas Wnt/genética , Proteína Wnt4
20.
Gene Expr Patterns ; 7(6): 680-99, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17452023

RESUMEN

Cataloguing gene expression during development of the genitourinary tract will increase our understanding not only of this process but also of congenital defects and disease affecting this organ system. We have developed a high-resolution ontology with which to describe the subcompartments of the developing murine genitourinary tract. This ontology incorporates what can be defined histologically and begins to encompass other structures and cell types already identified at the molecular level. The ontology is being used to annotate in situ hybridisation data generated as part of the Genitourinary Development Molecular Anatomy Project (GUDMAP), a publicly available data resource on gene and protein expression during genitourinary development. The GUDMAP ontology encompasses Theiler stage (TS) 17-27 of development as well as the sexually mature adult. It has been written as a partonomic, text-based, hierarchical ontology that, for the embryological stages, has been developed as a high-resolution expansion of the existing Edinburgh Mouse Atlas Project (EMAP) ontology. It also includes group terms for well-characterised structural and/or functional units comprising several sub-structures, such as the nephron and juxtaglomerular complex. Each term has been assigned a unique identification number. Synonyms have been used to improve the success of query searching and maintain wherever possible existing EMAP terms relating to this organ system. We describe here the principles and structure of the ontology and provide representative diagrammatic, histological, and whole mount and section RNA in situ hybridisation images to clarify the terms used within the ontology. Visual examples of how terms appear in different specimen types are also provided.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Ratones/genética , Sistema Urogenital/crecimiento & desarrollo , Animales , Clítoris/crecimiento & desarrollo , Endodermo/fisiología , Femenino , Masculino , Mesodermo/fisiología , Ratones/embriología , Ratones/crecimiento & desarrollo , Nefronas/embriología , Nefronas/crecimiento & desarrollo , Pene/crecimiento & desarrollo , Escroto/crecimiento & desarrollo , Maduración Sexual , Sistema Urogenital/anatomía & histología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA