Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 150(3): 590-605, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863011

RESUMEN

Endothelium in embryonic hematopoietic tissues generates hematopoietic stem/progenitor cells; however, it is unknown how its unique potential is specified. We show that transcription factor Scl/Tal1 is essential for both establishing the hematopoietic transcriptional program in hemogenic endothelium and preventing its misspecification to a cardiomyogenic fate. Scl(-/-) embryos activated a cardiac transcriptional program in yolk sac endothelium, leading to the emergence of CD31+Pdgfrα+ cardiogenic precursors that generated spontaneously beating cardiomyocytes. Ectopic cardiogenesis was also observed in Scl(-/-) hearts, where the disorganized endocardium precociously differentiated into cardiomyocytes. Induction of mosaic deletion of Scl in Scl(fl/fl)Rosa26Cre-ER(T2) embryos revealed a cell-intrinsic, temporal requirement for Scl to prevent cardiomyogenesis from endothelium. Scl(-/-) endothelium also upregulated the expression of Wnt antagonists, which promoted rapid cardiomyocyte differentiation of ectopic cardiogenic cells. These results reveal unexpected plasticity in embryonic endothelium such that loss of a single master regulator can induce ectopic cardiomyogenesis from endothelial cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Endotelio Vascular/embriología , Corazón/embriología , Proteínas Proto-Oncogénicas/metabolismo , Animales , Cadherinas/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Mesodermo/metabolismo , Ratones , Miocitos Cardíacos/citología , Placenta/irrigación sanguínea , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/metabolismo , Saco Vitelino/irrigación sanguínea
2.
Semin Cell Dev Biol ; 122: 44-49, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34083115

RESUMEN

Direct cardiac reprogramming, which refers to somatic cell (i.e. fibroblast) fate conversion to cardiomyocyte-like cell without transitioning through an intermediate pluripotent state, provides a novel therapeutic strategy for heart regeneration by converting resident cardiac fibroblasts to cardiomyocytes in situ. However, several limitations need to be addressed prior to clinical translation of this technology. They include low efficiency of reprogramming, heterogeneity of starting fibroblasts, functional immaturity of induced cardiomyocytes (iCMs), virus immunogenicity and toxicity, incomplete understanding of changes in the epigenetic landscape as fibroblasts undergo reprogramming, and the environmental factors that influence fate conversion. Several studies have demonstrated that a combination of enforced expression of cardiac transcription factors along with certain cytokines and growth factors in the presence of favorable environmental cues (including extracellular matrix, topography, and mechanical properties) enhance the efficiency and quality of direct reprogramming. This paper reviews the literature on the influence of the microenvironment on direct cardiac reprogramming in vitro and in vivo.


Asunto(s)
Reprogramación Celular/fisiología , Miocitos Cardíacos/metabolismo , Animales , Exposición a Riesgos Ambientales , Humanos , Ratones
3.
EMBO J ; 34(6): 759-77, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25564442

RESUMEN

Scl/Tal1 confers hemogenic competence and prevents ectopic cardiomyogenesis in embryonic endothelium by unknown mechanisms. We discovered that Scl binds to hematopoietic and cardiac enhancers that become epigenetically primed in multipotent cardiovascular mesoderm, to regulate the divergence of hematopoietic and cardiac lineages. Scl does not act as a pioneer factor but rather exploits a pre-established epigenetic landscape. As the blood lineage emerges, Scl binding and active epigenetic modifications are sustained in hematopoietic enhancers, whereas cardiac enhancers are decommissioned by removal of active epigenetic marks. Our data suggest that, rather than recruiting corepressors to enhancers, Scl prevents ectopic cardiogenesis by occupying enhancers that cardiac factors, such as Gata4 and Hand1, use for gene activation. Although hematopoietic Gata factors bind with Scl to both activated and repressed genes, they are dispensable for cardiac repression, but necessary for activating genes that enable hematopoietic stem/progenitor cell development. These results suggest that a unique subset of enhancers in lineage-specific genes that are accessible for regulators of opposing fates during the time of the fate decision provide a platform where the divergence of mutually exclusive fates is orchestrated.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Elementos de Facilitación Genéticos/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/citología , Mesodermo/embriología , Mioblastos Cardíacos/citología , Proteínas Proto-Oncogénicas/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Biblioteca de Genes , Células Madre Hematopoyéticas/fisiología , Humanos , Mesodermo/metabolismo , Análisis por Micromatrices , Modelos Biológicos , Datos de Secuencia Molecular , Mioblastos Cardíacos/fisiología , Análisis de Secuencia de ARN , Proteína 1 de la Leucemia Linfocítica T Aguda
4.
Development ; 143(3): 473-82, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26674310

RESUMEN

The elucidation of mechanisms in semilunar valve development might enable the development of new therapies for congenital heart disorders. Here, we found differences in proliferation-associated genes and genes repressed by VEGF between human semilunar valve leaflets from first and second trimester hearts. The proliferation of valve interstitial cells and ventricular valve endothelial cells (VECs) and cellular density declined from the first to the second trimester. Cytoplasmic expression of NFATC1 was detected in VECs (4 weeks) and, later, cells in the leaflet/annulus junction mesenchyme expressing inactive NFATC1 (5.5-9 weeks) were detected, indicative of endocardial-to-mesenchymal transformation (EndMT) in valvulogenesis. At this leaflet/annulus junction, CD44(+) cells clustered during elongation (11 weeks), extending toward the tip along the fibrosal layer in second trimester leaflets. Differing patterns of maturation in the fibrosa and ventricularis were detected via increased fibrosal periostin content, which tracked the presence of the CD44(+) cells in the second trimester. We revealed that spatiotemporal NFATC1 expression actively regulates EndMT during human valvulogenesis, as early as 4 weeks. Additionally, CD44(+) cells play a role in leaflet maturation toward the trilaminar structure, possibly via migration of VECs undergoing EndMT, which subsequently ascend from the leaflet/annulus junction.


Asunto(s)
Endocardio/embriología , Válvulas Cardíacas/citología , Válvulas Cardíacas/embriología , Mesodermo/citología , Mesodermo/embriología , Moléculas de Adhesión Celular/metabolismo , Recuento de Células , Diferenciación Celular , Proliferación Celular , Células Endoteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Receptores de Hialuranos/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Embarazo , Segundo Trimestre del Embarazo , Análisis Espacio-Temporal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Ann Rheum Dis ; 77(5): 760-769, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29436471

RESUMEN

OBJECTIVE: Human adult articular cartilage (AC) has little capacity for repair, and joint surface injuries often result in osteoarthritis (OA), characterised by loss of matrix, hypertrophy and chondrocyte apoptosis. Inflammation mediated by interleukin (IL)-6 family cytokines has been identified as a critical driver of proarthritic changes in mouse and human joints, resulting in a feed-forward process driving expression of matrix degrading enzymes and IL-6 itself. Here we show that signalling through glycoprotein 130 (gp130), the common receptor for IL-6 family cytokines, can have both context-specific and cytokine-specific effects on articular chondrocytes and that a small molecule gp130 modulator can bias signalling towards anti-inflammatory and antidegenerative outputs. METHODS: High throughput screening of 170 000 compounds identified a small molecule gp130 modulator termed regulator of cartilage growth and differentiation (RCGD 423) that promotes atypical homodimeric signalling in the absence of cytokine ligands, driving transient increases in MYC and pSTAT3 while suppressing oncostatin M- and IL-6-mediated activation of ERK and NF-κB via direct competition for gp130 occupancy. RESULTS: This small molecule increased proliferation while reducing apoptosis and hypertrophic responses in adult chondrocytes in vitro. In a rat partial meniscectomy model, RCGD 423 greatly reduced chondrocyte hypertrophy, loss and degeneration while increasing chondrocyte proliferation beyond that observed in response to injury. Moreover, RCGD 423 improved cartilage healing in a rat full-thickness osteochondral defect model, increasing proliferation of mesenchymal cells in the defect and also inhibiting breakdown of cartilage matrix in de novo generated cartilage. CONCLUSION: These results identify a novel strategy for AC remediation via small molecule-mediated modulation of gp130 signalling.


Asunto(s)
Enfermedades de los Cartílagos/tratamiento farmacológico , Cartílago Articular/metabolismo , Receptor gp130 de Citocinas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Genes myc/efectos de los fármacos , Ratas , Factor de Transcripción STAT3/metabolismo
6.
BMC Genomics ; 18(1): 655, 2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28835208

RESUMEN

BACKGROUND: Rice (Oryza sativa) is critical for human nutrition worldwide. Due to a growing population, cultivars that produce high yields in high salinity soil are of major importance. Here we describe the discovery and molecular characterization of a novel sea water adapted rice strain, Sea Rice 86 (SR86). RESULTS: SR86 can produce nutritious grains when grown in high salinity soil. Compared to a salt resistant rice cultivar, Yanfen 47 (YF47), SR86 grows in environments with up to 3X the salt content, and produces grains with significantly higher nutrient content in 12 measured components, including 2.9X calcium and 20X dietary fiber. Whole genome sequencing demonstrated that SR86 is a relatively ancient indica subspecies, phylogenetically close to the divergence point of the major rice varietals. SR86 has 12 chromosomes with a total genome size of 373,130,791 bps, slightly smaller than other sequenced rice genomes. Via comparison with 3000 rice genomes, we identified 42,359 putative unique, high impact variants in SR86. Transcriptome analysis of SR86 grown under normal and high saline conditions identified a large number of differentially expressed and salt-induced genes. Many of those genes fall into several gene families that have established or suggested roles in salt tolerance, while others represent potentially novel mediators of salt adaptation. CONCLUSIONS: Whole genome sequencing and transcriptome analysis of SR86 has laid a foundation for further molecular characterization of several desirable traits in this novel rice cultivar. A number of candidate genes related to salt adaptation identified in this study will be valuable for further functional investigation.


Asunto(s)
Adaptación Fisiológica/genética , Perfilación de la Expresión Génica , Oryza/genética , Oryza/fisiología , Sales (Química)/farmacología , Agua de Mar/química , Secuenciación Completa del Genoma , Adaptación Fisiológica/efectos de los fármacos , Mapeo Cromosómico , Cromosomas de las Plantas/genética , Genes de Plantas/genética , Marcadores Genéticos/genética , Inmersión , Oryza/efectos de los fármacos
7.
Haematologica ; 101(4): 417-26, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26611474

RESUMEN

Thymic stromal lymphopoietin (TSLP) stimulates in-vitro proliferation of human fetal B-cell precursors. However, its in-vivo role during normal human B lymphopoiesis is unknown. Genetic alterations that cause overexpression of its receptor component, cytokine receptor-like factor 2 (CRLF2), lead to high-risk B-cell acute lymphoblastic leukemia implicating this signaling pathway in leukemogenesis. We show that mouse thymic stromal lymphopoietin does not stimulate the downstream pathways (JAK/STAT5 and PI3K/AKT/mTOR) activated by the human cytokine in primary high-risk leukemia with overexpression of the receptor component. Thus, the utility of classic patient-derived xenografts for in-vivo studies of this pathway is limited. We engineered xenograft mice to produce human thymic stromal lymphopoietin (+T mice) by injection with stromal cells transduced to express the cytokine. Control (-T) mice were produced using stroma transduced with control vector. Normal levels of human thymic stromal lymphopoietin were achieved in sera of +T mice, but were undetectable in -T mice. Patient-derived xenografts generated from +T as compared to -T mice showed a 3-6-fold increase in normal human B-cell precursors that was maintained through later stages of B-cell development. Gene expression profiles in high-risk B-cell acute lymphoblastic leukemia expanded in +T mice indicate increased mTOR pathway activation and are more similar to the original patient sample than those from -T mice. +T/-T xenografts provide a novel pre-clinical model for understanding this pathway in B lymphopoiesis and identifying treatments for high-risk B-cell acute lymphoblastic leukemia with overexpression of cytokine-like factor receptor 2.


Asunto(s)
Xenoinjertos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Receptores de Citocinas/metabolismo , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Xenoinjertos/inmunología , Humanos , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Recuento de Linfocitos , Linfopoyesis/genética , Linfopoyesis/inmunología , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Citocinas/genética , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Transgenes , Trasplante Heterólogo
8.
J Virol ; 87(4): 2094-108, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23192877

RESUMEN

Sophisticated retargeting systems for lentiviral vectors have been developed in recent years. Most seek to suppress the viral envelope's natural tropism while modifying the receptor-binding domain such that its tropism is determined by the specificity of the engineered ligand-binding motif. Here we took advantage of the natural tropism of Nipah virus (NiV), whose attachment envelope glycoprotein has picomolar affinity for ephrinB2, a molecule proposed as a molecular marker of "stemness" (present on embryonic, hematopoietic, and neural stem cells) as well as being implicated in tumorigenesis of specific cancers. NiV entry requires both the fusion (F) and attachment (G) glycoproteins. Truncation of the NiV-F cytoplasmic tail (T5F) alone, combined with full-length NiV-G, resulted in optimal titers of NiV-pseudotyped particles (NiVpp) (∼10(6) IU/ml), even without ultracentrifugation. To further enhance the infectivity of NiVpp, we engineered a hyperfusogenic NiV-F protein lacking an N-linked glycosylation site (T5FΔN3). T5FΔN3/wt G particles exhibited enhanced infectivity on less permissive cell lines and efficiently targeted ephrinB2(+) cells even in a 1,000-fold excess of ephrinB2-negative cells, all without any loss of specificity, as entry was abrogated by soluble ephrinB2. NiVpp also transduced human embryonic, hematopoietic, and neural stem cell populations in an ephrinB2-dependent manner. Finally, intravenous administration of the luciferase reporter NiVpp-T5FΔN3/G to mice resulted in signals being detected in the spleen and lung but not in the liver. Bypassing the liver sink is a critical barrier for targeted gene therapy. The extraordinary specificity of NiV-G for ephrinB2 holds promise for targeting specific ephrinB2(+) populations in vivo or in vitro.


Asunto(s)
Efrina-B2/metabolismo , Vectores Genéticos , Lentivirus/genética , Virus Nipah/fisiología , Receptores Virales/metabolismo , Células Madre/virología , Internalización del Virus , Animales , Células Cultivadas , Humanos , Ratones , Biología Molecular/métodos , Virus Nipah/genética , Transducción Genética
9.
Blood ; 117(7): 2157-65, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21163928

RESUMEN

MicroRNAs (miRs) play an important role in cell differentiation and maintenance of cell identity, but relatively little is known of their functional role in modulating human hematopoietic lineage differentiation. Human embryonic stem cells (hESCs) provide a model system to study early human hematopoiesis. We differentiated hESCs by embryoid body (EB) formation and compared the miR expression profile of undifferentiated hESCs to CD34(+) EB cells. miRs-126/126* were the most enriched of the 7 miRs that were up-regulated in CD34(+) cells, and their expression paralleled the kinetics of hematopoietic transcription factors RUNX1, SCL, and PU.1. To define the role of miRs-126/126* in hematopoiesis, we created hESCs overexpressing doxycycline-regulated miRs-126/126* and analyzed their hematopoietic differentiation. Induction of miRs-126/126* during both EB differentiation and colony formation reduced the number of erythroid colonies, suggesting an inhibitory role of miRs-126/126* in erythropoiesis. Protein tyrosine phosphatase, nonreceptor type 9 (PTPN9), a protein tyrosine phosphatase that is required for growth and expansion of erythroid cells, is one target of miR-126. PTPN9 restoration partially relieved the suppressed erythropoiesis caused by miRs-126/126*. Our results define an important function of miRs-126/126* in negative regulation of erythropoiesis, providing the first evidence for a role of miR in hematopoietic differentiation of hESCs.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Eritropoyesis/genética , Eritropoyesis/fisiología , MicroARNs/genética , Antígenos CD34/metabolismo , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Cinética , MicroARNs/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/fisiología
10.
Blood ; 116(17): 3321-30, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20628147

RESUMEN

Embryonic hematopoiesis starts via the generation of primitive red blood cells (RBCs) that satisfy the embryo's immediate oxygen needs. Although primitive RBCs were thought to retain their nuclei, recent studies have shown that primitive RBCs in mice enucleate in the fetal liver. It has been unknown whether human primitive RBCs enucleate, and what hematopoietic site might support this process. Our data indicate that the terminal maturation and enucleation of human primitive RBCs occurs in first trimester placental villi. Extravascular ζ-globin(+) primitive erythroid cells were found in placental villi between 5-7 weeks of development, at which time the frequency of enucleated RBCs was higher in the villous stroma than in circulation. RBC enucleation was further evidenced by the presence of primitive reticulocytes and pyrenocytes (ejected RBC nuclei) in the placenta. Extravascular RBCs were found to associate with placental macrophages, which contained ingested nuclei. Clonogenic macrophage progenitors of fetal origin were present in the chorionic plate of the placenta before the onset of fetoplacental circulation, after which macrophages had migrated to the villi. These findings indicate that placental macrophages may assist the enucleation process of primitive RBCs in placental villi, implying an unexpectedly broad role for the placenta in embryonic hematopoiesis.


Asunto(s)
Células Eritroides/citología , Eritropoyesis , Placenta/citología , Primer Trimestre del Embarazo , Vellosidades Coriónicas/ultraestructura , Femenino , Feto/irrigación sanguínea , Feto/citología , Humanos , Macrófagos/citología , Placenta/irrigación sanguínea , Placenta/ultraestructura , Embarazo
11.
Front Cell Dev Biol ; 10: 954028, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36092701

RESUMEN

Complex injury and open reconstructive surgeries of the knee often lead to joint dysfunction that may alter the normal biomechanics of the joint. Two major complications that often arise are excessive deposition of fibrotic tissue and acquired heterotopic endochondral ossification. Knee arthrofibrosis is a fibrotic joint disorder where aberrant buildup of scar tissue and adhesions develop around the joint. Heterotopic ossification is ectopic bone formation around the periarticular tissues. Even though arthrofibrosis and heterotopic ossification pose an immense clinical problem, limited studies focus on their cellular and molecular mechanisms. Effective cell-targeted therapeutics are needed, but the cellular origin of both knee disorders remains elusive. Moreover, all the current animal models of knee arthrofibrosis and stiffness are developed in rats and rabbits, limiting genetic experiments that would allow us to explore the contribution of specific cellular targets to these knee pathologies. Here, we present a novel mouse model where surgically induced injury and hyperextension of the knee lead to excessive deposition of disorganized collagen in the meniscus, synovium, and joint capsule in addition to formation of extra-skeletal bone in muscle and soft tissues within the joint capsule. As a functional outcome, arthrofibrosis and acquired heterotopic endochondral ossification coupled with a significant increase in total joint stiffness were observed. By employing this injury model and genetic lineage tracing, we also demonstrate that Gli1+ mesenchymal progenitors proliferate after joint injury and contribute to the pool of fibrotic cells in the synovium and ectopic osteoblasts within the joint capsule. These findings demonstrate that Gli1+ cells are a major cellular contributor to knee arthrofibrosis and acquired heterotopic ossification that manifest after knee injury. Our data demonstrate that genetic manipulation of Gli1+ cells in mice may offer a platform for identification of novel therapeutic targets to prevent knee joint dysfunction after chronic injury.

12.
Commun Biol ; 5(1): 64, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35039652

RESUMEN

Growth of long bones and vertebrae is maintained postnatally by a long-lasting pool of progenitor cells. Little is known about the molecular mechanisms that regulate the output and maintenance of the cells that give rise to mature cartilage. Here we demonstrate that postnatal chondrocyte-specific deletion of a transcription factor Stat3 results in severely reduced proliferation coupled with increased hypertrophy, growth plate fusion, stunting and signs of progressive dysfunction of the articular cartilage. This effect is dimorphic, with females more strongly affected than males. Chondrocyte-specific deletion of the IL-6 family cytokine receptor gp130, which activates Stat3, phenocopied Stat3-deletion; deletion of Lifr, one of many co-receptors that signals through gp130, resulted in a milder phenotype. These data define a molecular circuit that regulates chondrogenic cell maintenance and output and reveals a pivotal positive function of IL-6 family cytokines in the skeletal system with direct implications for skeletal development and regeneration.


Asunto(s)
Condrocitos/metabolismo , Receptor gp130 de Citocinas/genética , Placa de Crecimiento/metabolismo , Ratones/genética , Factor de Transcripción STAT3/genética , Animales , Proliferación Celular/genética , Receptor gp130 de Citocinas/metabolismo , Homeostasis/genética , Ratones/crecimiento & desarrollo , Factor de Transcripción STAT3/metabolismo
13.
Curr Opin Hematol ; 17(4): 313-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20571394

RESUMEN

PURPOSE OF REVIEW: The lifelong stream of all blood cells originates from the pool of hematopoietic stem cells (HSCs) generated during embryogenesis. Given that the placenta has been recently unveiled as a major hematopoietic organ that supports HSC development, the purpose of this review is to present current advances in defining the origin and regulation of placental HSCs. RECENT FINDINGS: The mouse placenta has been shown to have the potential to generate multipotential myelo-lymphoid hematopoietic stem/progenitor cells de novo. The cellular origin of HSCs generated in the placenta and other sites has been tracked to the hemogenic endothelium by using novel genetic and imaging-based cell-tracing approaches. Transplantable, myelo-lymphoid hematopoietic stem/progenitor cells have also been recovered from the human placenta throughout gestation. SUMMARY: The discovery of the placenta as a major organ that generates HSCs and maintains them in an undifferentiated state provides a valuable model to further elucidate regulatory mechanisms governing HSC emergence and expansion during mouse and human development. Concurrent efforts to optimize protocols for placental banking and HSC harvesting may increase the therapeutic utility of the human placenta as a source of transplantable HSCs.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Placenta/citología , Placenta/fisiología , Animales , Linaje de la Célula , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Ratones , Embarazo
14.
Front Cell Dev Biol ; 9: 725854, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733842

RESUMEN

Cartilage tissue is comprised of extracellular matrix and chondrocytes, a cell type with very low cellular turnover in adults, providing limited capacity for regeneration. However, in development a significant number of chondrocytes actively proliferate and remodel the surrounding matrix. Uncoupling the microenvironmental influences that determine the balance between clonogenic potential and terminal differentiation of these cells is essential for the development of novel approaches for cartilage regeneration. Unfortunately, most of the existing methods are not applicable for the analysis of functional properties of chondrocytes at a single cell resolution. Here we demonstrate that a novel 3D culture method provides a long-term and permissive in vitro niche that selects for highly clonogenic, colony-forming chondrocytes which maintain cartilage-specific matrix production, thus recapitulating the in vivo niche. As a proof of concept, clonogenicity of Sox9 IRES-EGFP mouse chondrocytes is almost exclusively found in the highest GFP+ fraction known to be enriched for chondrocyte progenitor cells. Although clonogenic chondrocytes are very rare in adult cartilage, we have optimized this system to support large, single cell-derived chondrogenic organoids with complex zonal architecture and robust chondrogenic phenotype from adult pig and human articular chondrocytes. Moreover, we have demonstrated that growth trajectory and matrix biosynthesis in these organoids respond to a pro-inflammatory environment. This culture method offers a robust, defined and controllable system that can be further used to interrogate the effects of various microenvironmental signals on chondrocytes, providing a high throughput platform to assess genetic and environmental factors in development and disease.

15.
NPJ Regen Med ; 6(1): 77, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815400

RESUMEN

Osteoarthritis (OA) impacts hundreds of millions of people worldwide, with those affected incurring significant physical and financial burdens. Injuries such as focal defects to the articular surface are a major contributing risk factor for the development of OA. Current cartilage repair strategies are moderately effective at reducing pain but often replace damaged tissue with biomechanically inferior fibrocartilage. Here we describe the development, transcriptomic ontogenetic characterization and quality assessment at the single cell level, as well as the scaled manufacturing of an allogeneic human pluripotent stem cell-derived articular chondrocyte formulation that exhibits long-term functional repair of porcine articular cartilage. These results define a new potential clinical paradigm for articular cartilage repair and mitigation of the associated risk of OA.

16.
Nat Commun ; 12(1): 2595, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972536

RESUMEN

Tissue regeneration is a process that recapitulates and restores organ structure and function. Although previous studies have demonstrated wound-induced hair neogenesis (WIHN) in laboratory mice (Mus), the regeneration is limited to the center of the wound unlike those observed in African spiny (Acomys) mice. Tissue mechanics have been implicated as an integral part of tissue morphogenesis. Here, we use the WIHN model to investigate the mechanical and molecular responses of laboratory and African spiny mice, and report these models demonstrate opposing trends in spatiotemporal morphogenetic field formation with association to wound stiffness landscapes. Transcriptome analysis and K14-Cre-Twist1 transgenic mice show the Twist1 pathway acts as a mediator for both epidermal-dermal interactions and a competence factor for periodic patterning, differing from those used in development. We propose a Turing model based on tissue stiffness that supports a two-scale tissue mechanics process: (1) establishing a morphogenetic field within the wound bed (mm scale) and (2) symmetry breaking of the epidermis and forming periodically arranged hair primordia within the morphogenetic field (µm scale). Thus, we delineate distinct chemo-mechanical events in building a Turing morphogenesis-competent field during WIHN of laboratory and African spiny mice and identify its evo-devo advantages with perspectives for regenerative medicine.


Asunto(s)
Epidermis/anatomía & histología , Epidermis/metabolismo , Folículo Piloso/metabolismo , Morfogénesis/fisiología , Regeneración/fisiología , Proteína 1 Relacionada con Twist/metabolismo , Cicatrización de Heridas/fisiología , Animales , Epidermis/fisiología , Perfilación de la Expresión Génica , Folículo Piloso/anatomía & histología , Folículo Piloso/fisiología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Microscopía de Fuerza Atómica , Modelos Psicológicos , Morfogénesis/genética , Murinae , RNA-Seq , Regeneración/genética , Medicina Regenerativa , Transducción de Señal/genética , Transducción de Señal/fisiología , Análisis Espacio-Temporal , Proteína 1 Relacionada con Twist/genética , Cicatrización de Heridas/genética
17.
Cell Stem Cell ; 27(1): 158-176.e10, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32396864

RESUMEN

The developmental trajectory of human skeletal myogenesis and the transition between progenitor and stem cell states are unclear. We used single-cell RNA sequencing to profile human skeletal muscle tissues from embryonic, fetal, and postnatal stages. In silico, we identified myogenic as well as other cell types and constructed a "roadmap" of human skeletal muscle ontogeny across development. In a similar fashion, we also profiled the heterogeneous cell cultures generated from multiple human pluripotent stem cell (hPSC) myogenic differentiation protocols and mapped hPSC-derived myogenic progenitors to an embryonic-to-fetal transition period. We found differentially enriched biological processes and discovered co-regulated gene networks and transcription factors present at distinct myogenic stages. This work serves as a resource for advancing our knowledge of human myogenesis. It also provides a tool for a better understanding of hPSC-derived myogenic progenitors for translational applications in skeletal muscle-based regenerative medicine.


Asunto(s)
Desarrollo de Músculos , Células Madre Pluripotentes , Diferenciación Celular , Humanos , Músculo Esquelético , Factores de Transcripción
18.
Int J Nanomedicine ; 14: 5831-5848, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31534327

RESUMEN

PURPOSE: In order to accelerate the tendon-bone healing processes and achieve the efficient osteointegration between the tendon graft and bone tunnel, we aim to design bioactive electrospun nanofiber membranes combined with tendon stem/progenitor cells (TSPCs) to promote osteogenic regeneration of the tendon and bone interface. METHODS: In this study, nanofiber membranes of polycaprolactone (PCL), PCL/collagen I (COL-1) hybrid nanofiber membranes, poly(dopamine) (PDA)-coated PCL nanofiber membranes and PDA-coated PCL/COL-1 hybrid nanofiber membranes were successfully fabricated by electrospinning. The biochemical characteristics and nanofibrous morphology of the membranes, as well as the characterization of rat TSPCs, were defined in vitro. After co-culture with different types of electrospun nanofiber membranes in vitro, cell proliferation, viability, adhesion and osteogenic differentiation of TSPCs were evaluated at different time points. RESULTS: Among all the membranes, the performance of the PCL/COL-1 (volume ratio: 2:1 v/v) group was superior in terms of its ability to support the adhesion, proliferation, and osteogenic differentiation of TSPCs. No benefit was found in this study to include PDA coating on cell adhesion, proliferation and osteogenic differentiation of TSPCs. CONCLUSION: The PCL/COL-1 hybrid electrospun nanofiber membranes are biocompatible, biomimetic, easily fabricated, and are capable of supporting cell adhesion, proliferation, and osteogenic differentiation of TSPCs. These bioactive electrospun nanofiber membranes may act as a suitable functional biomimetic scaffold in tendon-bone tissue engineering applications to enhance tendon-bone healing abilities.


Asunto(s)
Materiales Biocompatibles/farmacología , Huesos/fisiología , Membranas Artificiales , Nanofibras/química , Células Madre/citología , Tendones/citología , Ingeniería de Tejidos/métodos , Animales , Huesos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Nanofibras/ultraestructura , Osteogénesis , Ratas Sprague-Dawley , Células Madre/efectos de los fármacos
20.
Nat Commun ; 9(1): 3634, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30194383

RESUMEN

Tissue-specific gene expression defines cellular identity and function, but knowledge of early human development is limited, hampering application of cell-based therapies. Here we profiled 5 distinct cell types at a single fetal stage, as well as chondrocytes at 4 stages in vivo and 2 stages during in vitro differentiation. Network analysis delineated five tissue-specific gene modules; these modules and chromatin state analysis defined broad similarities in gene expression during cartilage specification and maturation in vitro and in vivo, including early expression and progressive silencing of muscle- and bone-specific genes. Finally, ontogenetic analysis of freshly isolated and pluripotent stem cell-derived articular chondrocytes identified that integrin alpha 4 defines 2 subsets of functionally and molecularly distinct chondrocytes characterized by their gene expression, osteochondral potential in vitro and proliferative signature in vivo. These analyses provide new insight into human musculoskeletal development and provide an essential comparative resource for disease modeling and regenerative medicine.


Asunto(s)
Condrocitos/metabolismo , Condrogénesis , Mioblastos/metabolismo , Osteoblastos/metabolismo , Tenocitos/metabolismo , Animales , Biomarcadores/metabolismo , Epigénesis Genética , Desarrollo Fetal , Perfilación de la Expresión Génica , Código de Histonas , Humanos , Ratones , Análisis de Secuencia de ARN , Porcinos , Transcripción Genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda