Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Am J Respir Cell Mol Biol ; 66(4): 402-414, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35045271

RESUMO

Oxygen supplementation in preterm infants disrupts alveolar epithelial type 2 (AT2) cell proliferation through poorly understood mechanisms. Here, newborn mice are used to understand how hyperoxia stimulates an early aberrant wave of AT2 cell proliferation that occurs between Postnatal Days (PNDs) 0 and 4. RNA-sequencing analysis of AT2 cells isolated from PND4 mice revealed hyperoxia stimulates expression of mitochondrial-specific methylenetetrahydrofolate dehydrogenase 2 and other genes involved in mitochondrial one-carbon coupled folate metabolism and serine synthesis. The same genes are induced when AT2 cells normally proliferate on PND7 and when they proliferate in response to the mitogen fibroblast growth factor 7. However, hyperoxia selectively stimulated their expression via the stress-responsive activating transcription factor 4 (ATF4). Administration of the mitochondrial superoxide scavenger mitoTEMPO during hyperoxia suppressed ATF4 and thus early AT2 cell proliferation, but it had no effect on normative AT2 cell proliferation seen on PND7. Because ATF4 and methylenetetrahydrofolate dehydrogenase are detected in hyperplastic AT2 cells of preterm infant humans and baboons with bronchopulmonary dysplasia, dampening mitochondrial oxidative stress and ATF4 activation may provide new opportunities for controlling excess AT2 cell proliferation in neonatal lung disease.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Hiperóxia , Fator 4 Ativador da Transcrição/genética , Animais , Animais Recém-Nascidos , Proliferação de Células , Ácido Fólico/farmacologia , Hiperóxia/metabolismo , Recém-Nascido Prematuro , Camundongos
2.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L846-L859, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29345197

RESUMO

Supplemental oxygen given to preterm infants has been associated with permanently altering postnatal lung development. Now that these individuals are reaching adulthood, there is growing concern that early life oxygen exposure may also promote cardiovascular disease through poorly understood mechanisms. We previously reported that adult mice exposed to 100% oxygen between postnatal days 0 and 4 develop pulmonary hypertension, defined pathologically by capillary rarefaction, dilation of arterioles and veins, cardiac failure, and a reduced lifespan. Here, Affymetrix Gene Arrays are used to identify early transcriptional changes that take place in the lung before pulmonary capillary rarefaction. We discovered neonatal hyperoxia reduced expression of cardiac muscle genes, including those involved in contraction, calcium signaling, mitochondrial respiration, and vasodilation. Quantitative RT-PCR, immunohistochemistry, and genetic lineage mapping using Myh6CreER; Rosa26RmT/mG mice revealed this reflected loss of pulmonary vein cardiomyocytes. The greatest loss of cadiomyocytes was seen within the lung followed by a graded loss beginning at the hilum and extending into the left atrium. Loss of these cells was seen by 2 wk of age in mice exposed to ≥80% oxygen and was attributed, in part, to reduced proliferation. Administering mitoTEMPO, a scavenger of mitochondrial superoxide during neonatal hyperoxia prevented loss of these cells. Since pulmonary vein cardiomyocytes help pump oxygen-rich blood out of the lung, their early loss following neonatal hyperoxia may contribute to cardiovascular disease seen in these mice, and perhaps in people who were born preterm.


Assuntos
Biomarcadores/metabolismo , Hiperóxia/fisiopatologia , Hipertensão Pulmonar/patologia , Mitocôndrias/química , Miócitos Cardíacos/patologia , Oxigênio/metabolismo , Veias Pulmonares/patologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Perfilação da Expressão Gênica , Hipertensão Pulmonar/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Oxirredução , Veias Pulmonares/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(34): 12444-9, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25114215

RESUMO

Changing the morphology of a simple epithelial tube to form a highly ramified branching network requires changes in cell behavior that lead to tissue-wide changes in organ shape. How epithelial cells in branched organs modulate their shape and behavior to promote bending and sculpting of the epithelial sheet is not well understood, and the mechanisms underlying this process remain obscure. We show that the Wnt receptor Frizzled 2 (Fzd2) is required for domain branch formation during the initial establishment of the respiratory tree. Live imaging and transcriptome analysis of lung-branching morphogenesis demonstrate that Fzd2 promotes changes in epithelial cell length and shape. These changes in cell morphology deform the developing epithelial tube to generate and maintain new domain branches. Fzd2 controls branch formation and the shape of the epithelial tube by regulating Rho signaling and by the localization of phospho-myosin light chain 2, in turn controlling the changes in the shape of epithelial cells during morphogenesis. This study demonstrates the importance of Wnt/Fzd2 signaling in promoting and maintaining changes in epithelial cell shape that affect development of a branching network.


Assuntos
Receptores Frizzled/metabolismo , Pulmão/embriologia , Animais , Forma Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Receptores Frizzled/deficiência , Receptores Frizzled/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Ligantes , Pulmão/citologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Morfogênese , Gravidez , Transdução de Sinais , Via de Sinalização Wnt , beta Catenina/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
4.
Dev Biol ; 398(1): 80-96, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25482987

RESUMO

Wnt proteins regulate cell behavior via a canonical signaling pathway that induces ß-catenin dependent transcription. It is now appreciated that Wnt/ß-catenin signaling promotes the expansion of the second heart field (SHF) progenitor cells that ultimately give-rise to the majority of cardiomyocytes. However, activating ß-catenin can also cause the loss of SHF progenitors, highlighting the necessity of precise control over ß-catenin signaling during heart development. We recently reported that two non-canonical Wnt ligands, Wnt5a and Wnt11, act cooperatively to attenuate canonical Wnt signaling that would otherwise disrupt the SHF. While these data reveal the essential role of this anti-canonical Wnt5a/Wnt11 signaling in SHF development, the mechanisms by which these ligands inhibit the canonical Wnt pathway are unclear. Wnt11 was previously shown to inhibit ß-catenin and promote cardiomyocyte maturation by activating a novel apoptosis-independent function of Caspases. Consistent with these data, we now show that Wnt5a and Wnt11 are capable of inducing Caspase activity in differentiating embryonic stem (ES) cells and that hearts from Wnt5a(-/-); Wnt11(-/-) embryos have diminished Caspase 3 (Casp3) activity. Furthermore, SHF markers are reduced in Casp3 mutant ES cells while the treatment of wild type ES cells with Caspase inhibitors blocked the ability of Wnt5a and Wnt11 to promote SHF gene expression. This finding was in agreement with our in vivo studies in which injecting pregnant mice with Caspase inhibitors reduced SHF marker expression in their gestating embryos. Caspase inhibition also blocked other Wnt5a/Wnt11 induced effects, including the suppression of ß-catenin protein expression and activity. Interestingly, Wnt5a/Wnt11 treatment of differentiating ES cells reduced both phosphorylated and total Akt through a Caspase-dependent mechanism and phosphorylated Akt levels were increased in the hearts Caspase inhibitor treated. Surprisingly, inhibition of either Akt or PI3K in ES cells was an equally effective means of increasing SHF markers compared to treatment with Wnt5a/Wnt11. Moreover, Akt inhibition restored SHF gene expression in Casp3 mutant ES cells. Taken together, these findings suggest that Wnt5a/Wnt11 inhibit ß-catenin to promote SHF development through Caspase-dependent Akt degradation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Caspases/metabolismo , Linhagem Celular , Células-Tronco Embrionárias/citologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Miocárdio/citologia , Fosforilação , Reação em Cadeia da Polimerase , Gravidez , Prenhez , Transdução de Sinais , Proteína Wnt-5a
5.
Dev Biol ; 408(1): 126-39, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26526197

RESUMO

Wnt ligands regulate heart morphogenesis but the underlying mechanisms remain unclear. Two Formin-related proteins, DAAM1 and 2, were previously found to bind the Wnt effector Disheveled. Here, since DAAM1 and 2 nucleate actin and mediate Wnt-induced cytoskeletal changes, a floxed-allele of Daam1 was used to disrupt its function specifically in the myocardium and investigate Wnt-associated pathways. Homozygous Daam1 conditional knockout (CKO) mice were viable but had misshapen hearts and poor cardiac function. The defects in Daam1 CKO mice were observed by mid-gestation and were associated with a loss of protrusions from cardiomyocytes invading the outflow tract. Further, these mice exhibited noncompaction cardiomyopathy (NCM) and deranged cardiomyocyte polarity. Interestingly, Daam1 CKO mice that were also homozygous for an insertion disrupting Daam2 (DKO) had stronger NCM, severely reduced cardiac function, disrupted sarcomere structure, and increased myocardial proliferation, suggesting that DAAM1 and DAAM2 have redundant functions. While RhoA was unaffected in the hearts of Daam1/2 DKO mice, AKT activity was lower than in controls, raising the issue of whether DAAM1/2 are only mediating Wnt signaling. Daam1-floxed mice were thus bred to Wnt5a null mice to identify genetic interactions. The hearts of Daam1 CKO mice that were also heterozygous for the null allele of Wnt5a had stronger NCM and more severe loss of cardiac function than Daam1 CKO mice, consistent with DAAM1 and Wnt5a acting in a common pathway. However, deleting Daam1 further disrupted Wnt5a homozygous-null hearts, suggesting that DAAM1 also has Wnt5a-independent roles in cardiac development.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Miocárdio/metabolismo , Sarcômeros/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Adesão Celular , Proliferação de Células , Citoesqueleto/metabolismo , Embrião de Mamíferos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Quinase 3 da Glicogênio Sintase/metabolismo , Testes de Função Cardíaca , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Heterozigoto , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Morfogênese , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Especificidade de Órgãos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas Wnt , Proteína Wnt-5a , Proteínas rho de Ligação ao GTP/deficiência , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
6.
Development ; 139(11): 1931-40, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22569553

RESUMO

Wnt/ß-catenin has a biphasic effect on cardiogenesis, promoting the induction of cardiac progenitors but later inhibiting their differentiation. Second heart field progenitors and expression of the second heart field transcription factor Islet1 are inhibited by the loss of ß-catenin, indicating that Wnt/ß-catenin signaling is necessary for second heart field development. However, expressing a constitutively active ß-catenin with Islet1-Cre also inhibits endogenous Islet1 expression, reflecting the inhibitory effect of prolonged Wnt/ß-catenin signaling on second heart field development. We show that two non-canonical Wnt ligands, Wnt5a and Wnt11, are co-required to regulate second heart field development in mice. Loss of Wnt5a and Wnt11 leads to a dramatic loss of second heart field progenitors in the developing heart. Importantly, this loss of Wnt5a and Wnt11 is accompanied by an increase in Wnt/ß-catenin signaling, and ectopic Wnt5a/Wnt11 inhibits ß-catenin signaling and promotes cardiac progenitor development in differentiating embryonic stem cells. These data show that Wnt5a and Wnt11 are essential regulators of the response of second heart field progenitors to Wnt/ß-catenin signaling and that they act by restraining Wnt/ß-catenin signaling during cardiac development.


Assuntos
Coração/embriologia , Miocárdio/citologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Proteínas Wnt/fisiologia , Animais , Western Blotting , Técnicas Histológicas , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Wnt/metabolismo , Proteína Wnt-5a , beta Catenina/metabolismo
7.
Proc Natl Acad Sci U S A ; 109(38): 15348-53, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22949635

RESUMO

Endoderm-mesenchyme cross-talk is a central process in the development of foregut-derived organs. How signaling pathways integrate the activity of multiple ligands to guide organ development is poorly understood. We show that two Wnt ligands, Wnt2 and Wnt7b, cooperatively induce Wnt signaling without affecting the stabilization of the Wnt canonical effector ß-catenin despite it being necessary for Wnt2-Wnt7b cooperativity. Wnt2-Wnt7b cooperation is specific for mesenchymal cell lineages and the combined loss of Wnt2 and Wnt7b leads to more severe developmental defects in the lung than loss of Wnt2 or Wnt7b alone. High-throughput small-molecule screens and biochemical assays reveal that the Pdgf pathway is required for cooperative Wnt2-Wnt7b signaling. Inhibition of Pdgf signaling in cell culture reduces Wnt2-Wnt7b cooperative signaling. Moreover, inhibition of Pdgf signaling in lung explant cultures results in decreased Wnt signaling and lung smooth-muscle development. These data suggest a model in which Pdgf signaling potentiates Wnt2-Wnt7b signaling to promote high levels of Wnt activity in mesenchymal progenitors that is required for proper development of endoderm-derived organs, such as the lung.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mucosa Intestinal/metabolismo , Intestinos/embriologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt2/metabolismo , Animais , Linhagem Celular , Linhagem da Célula , Epitélio/metabolismo , Humanos , Ligantes , Pulmão/metabolismo , Mesoderma/metabolismo , Camundongos , Miócitos de Músculo Liso/metabolismo , Organogênese/genética , Transdução de Sinais
8.
Circ Res ; 109(6): 616-28, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21778429

RESUMO

RATIONALE: Integrin-linked kinase (ILK) is located at focal adhesions and links the extracellular matrix (ECM) to the actin cytoskeleton via ß1- and ß3-integrins. ILK plays a role in the activation of kinases including protein kinase B/Akt and glycogen synthase kinase 3ß and regulates cell proliferation, motility, and survival. OBJECTIVE: To determine the function of ILK in vascular smooth muscle cells (SMCs) in vivo. METHODS AND RESULTS: SM22Cre(+)Ilk(Fl/Fl) conditional mutant mice were generated in which the Ilk gene was selectively ablated in SMCs. SM22Cre(+)Ilk(Fl/Fl) conditional mutant mice survive to birth but die in the perinatal period exhibiting multiple vascular pathologies including aneurysmal dilatation of the aorta and patent ductus arteriosus (PDA). Defects in morphogenetic development of the aorta were observed as early as E12.5 in SM22Cre(+)Ilk(Fl/Fl) mutant embryos. By late gestation (E16.5 to 18.5), striking expansion of the thoracic aorta was observed in ILK mutant embryos. Histological analyses revealed that the structural organization of the arterial tunica media is severely disrupted with profound derangements in SMC morphology, cell-cell, and cell-matrix relationships, including disruption of the elastic lamellae. ILK deletion in primary aortic SMCs results in alterations of RhoA/cytoskeletal signaling transduced through aberrant localization of myocardin-related transcription factor (MRTF)-A repressing the transcription and expression of SMC genes, which are required for the maintenance of the contractile SMC phenotype. CONCLUSIONS: These data identify a molecular pathway linking ILK signaling to the contractile SMC gene program. Activation of this pathway is required for morphogenetic development of the aorta and ductus arteriosus during embryonic and postnatal survival.


Assuntos
Aneurisma Aórtico/enzimologia , Deleção de Genes , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Animais , Aneurisma Aórtico/patologia , Células Cultivadas , Feminino , Marcação de Genes/métodos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/embriologia , Miócitos de Músculo Liso/citologia , Gravidez
9.
Dev Biol ; 356(2): 541-52, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21704027

RESUMO

Smooth muscle in the lung is thought to derive from the developing lung mesenchyme. Smooth muscle formation relies upon coordination of both autocrine and paracrine signaling between the budding epithelium and adjacent mesenchyme to govern its proliferation and differentiation. However, the pathways initiating the earliest aspects of smooth muscle specification and differentiation in the lung are poorly understood. Here, we identify the Wnt2 ligand as a critical regulator of the earliest aspects of lung airway smooth muscle development. Using Wnt2 loss and gain of function models, we show that Wnt2 signaling is necessary and sufficient for activation of a transcriptional and signaling network critical for smooth muscle specification and differentiation including myocardin/Mrtf-B and the signaling factor Fgf10. These studies place Wnt2 high in a hierarchy of signaling molecules that promote the earliest aspects of lung airway smooth muscle development.


Assuntos
Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Pulmão/embriologia , Músculo Liso/embriologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Proteína Wnt2/fisiologia , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/metabolismo , Mesoderma/embriologia , Camundongos , Músculo Liso/metabolismo , Reação em Cadeia da Polimerase , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , beta Catenina/fisiologia
10.
JCI Insight ; 6(5)2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33507880

RESUMO

Preterm birth increases the risk for pulmonary hypertension and heart failure in adulthood. Oxygen therapy can damage the immature cardiopulmonary system and may be partially responsible for the cardiovascular disease in adults born preterm. We previously showed that exposing newborn mice to hyperoxia causes pulmonary hypertension by 1 year of age that is preceded by a poorly understood loss of pulmonary vein cardiomyocyte proliferation. We now show that hyperoxia also reduces cardiomyocyte proliferation and survival in the left atrium and causes diastolic heart failure by disrupting its filling of the left ventricle. Transcriptomic profiling showed that neonatal hyperoxia permanently suppressed fatty acid synthase (Fasn), stearoyl-CoA desaturase 1 (Scd1), and other fatty acid synthesis genes in the atria of mice, the HL-1 line of mouse atrial cardiomyocytes, and left atrial tissue explanted from human infants. Suppressing Fasn or Scd1 reduced HL-1 cell proliferation and increased cell death, while overexpressing these genes maintained their expansion in hyperoxia, suggesting that oxygen directly inhibits atrial cardiomyocyte proliferation and survival by repressing Fasn and Scd1. Pharmacologic interventions that restore Fasn, Scd1, and other fatty acid synthesis genes in atrial cardiomyocytes may, thus, provide a way of ameliorating the adverse effects of supplemental oxygen on preterm infants.


Assuntos
Ácido Graxo Sintases/metabolismo , Ácidos Graxos/biossíntese , Átrios do Coração/citologia , Miócitos Cardíacos/metabolismo , Oxigênio/efeitos adversos , Nascimento Prematuro , Estearoil-CoA Dessaturase/metabolismo , Animais , Animais Recém-Nascidos , Morte Celular , Proliferação de Células , Modelos Animais de Doenças , Ácido Graxo Sintases/antagonistas & inibidores , Feminino , Átrios do Coração/patologia , Humanos , Hiperóxia , Recém-Nascido , Recém-Nascido Prematuro , Lipogênese , Masculino , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/patologia , Oxigênio/administração & dosagem , Terapia Respiratória , Estearoil-CoA Dessaturase/antagonistas & inibidores , Transcriptoma
11.
Nat Commun ; 12(1): 4155, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34230480

RESUMO

The organization of an integrated coronary vasculature requires the specification of immature endothelial cells (ECs) into arterial and venous fates based on their localization within the heart. It remains unclear how spatial information controls EC identity and behavior. Here we use single-cell RNA sequencing at key developmental timepoints to interrogate cellular contributions to coronary vessel patterning and maturation. We perform transcriptional profiling to define a heterogenous population of epicardium-derived cells (EPDCs) that express unique chemokine signatures. We identify a population of Slit2+ EPDCs that emerge following epithelial-to-mesenchymal transition (EMT), which we term vascular guidepost cells. We show that the expression of guidepost-derived chemokines such as Slit2 are induced in epicardial cells undergoing EMT, while mesothelium-derived chemokines are silenced. We demonstrate that epicardium-specific deletion of myocardin-related transcription factors in mouse embryos disrupts the expression of key guidance cues and alters EPDC-EC signaling, leading to the persistence of an immature angiogenic EC identity and inappropriate accumulation of ECs on the epicardial surface. Our study suggests that EC pathfinding and fate specification is controlled by a common mechanism and guided by paracrine signaling from EPDCs linking epicardial EMT to EC localization and fate specification in the developing heart.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , Pericárdio/citologia , Pericárdio/metabolismo , Animais , Quimiocinas , Vasos Coronários/metabolismo , Embrião de Mamíferos , Transição Epitelial-Mesenquimal , Expressão Gênica , Coração , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso , Proteínas Nucleares , Pericárdio/embriologia , Fator de Resposta Sérica , Transdução de Sinais , Transativadores , Fatores de Transcrição/metabolismo , Transcriptoma
12.
J Clin Invest ; 117(7): 1794-804, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17607356

RESUMO

The anterior heart field (AHF), which contributes to the outflow tract and right ventricle of the heart, is defined in part by expression of the LIM homeobox transcription factor Isl-1. The importance of Isl-1-positive cells in cardiac development and homeostasis is underscored by the finding that these cells are required for cardiac development and act as cardiac stem/progenitor cells within the postnatal heart. However, the molecular pathways regulating these cells' expansion and differentiation are poorly understood. We show that Isl-1-positive AHF progenitor cells in mice were responsive to Wnt/beta-catenin signaling, and these responsive cells contributed to the outflow tract and right ventricle of the heart. Loss of Wnt/beta-catenin signaling in the AHF caused defective outflow tract and right ventricular development with a decrease in Isl-1-positive progenitors and loss of FGF signaling. Conversely, Wnt gain of function in these cells led to expansion of Isl-1-positive progenitors with a concomitant increase in FGF signaling through activation of a specific set of FGF ligands including FGF3, FGF10, FGF16, and FGF20. These data reveal what we believe to be a novel Wnt-FGF signaling axis required for expansion of Isl-1-positive AHF progenitors and suggest future therapies to increase the number and function of these cells for cardiac regeneration.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Homeodomínio/metabolismo , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Marcadores Genéticos , Coração/embriologia , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM , Camundongos , Fatores de Transcrição , beta Catenina/deficiência , beta Catenina/genética
13.
Pediatr Cardiol ; 31(3): 342-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19967349

RESUMO

Cardiac development is comprised of a series of morphological events tightly controlled both spatially and temporally. The molecular pathways controlling early cardiac differentiation are poorly understood, but Wnt signaling is emerging as a critical pathway for multiple aspects of early cardiovascular development. The Wnt pathway plays multiple roles in regulating cellular behavior including proliferation, differentiation, cell migration, and cell polarity. Recent data have demonstrated that Wnt activity is important for early precardiac mesoderm differentiation but must be inhibited in subsequent steps for cardiomyocyte differentiation to proceed. Given the important role that Wnt signaling plays in both the differentiation of cardiomyocytes from pluripotential stem cells and tissue regeneration in general, an increased understanding of this pathway is likely to enhance our knowledge about both cardiovascular development and reparative mechanisms.


Assuntos
Sistema Cardiovascular/embriologia , Transdução de Sinais , Proteína Wnt1/fisiologia , Proteína Wnt2/fisiologia , beta Catenina/fisiologia , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Humanos , Modelos Animais , Células-Tronco Pluripotentes , Proteína Wnt1/genética , Proteína Wnt2/genética , beta Catenina/genética
14.
PLoS One ; 8(1): e55782, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383281

RESUMO

Previous studies have demonstrated that certain Wnt ligands can promote high levels of cooperative signaling in a cell type specific manner. To explore the underlying mechanism of this cooperative Wnt signaling, we performed a high-throughput screen of more than 14,000 cDNAs to identify genes that promote cooperative Wnt signaling in the context of a single Wnt ligand, Wnt2. This screen identified several homeobox factors including Msx2, Nkx5.2, and Esx1, in addition to other factors known to promote Wnt signaling including Pias4. Generation of dominant-active or dominant-negative forms of Msx2 indicate that the mechanism by which homeobox factors cooperatively promote Wnt signaling is through their ability to repress gene transcription. These data identify a broad homeobox code, which acts to increase Wnt signaling through transcriptional repression.


Assuntos
Genômica , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Linhagem Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genômica/métodos , Ensaios de Triagem em Larga Escala , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Ligantes , Ligação Proteica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
15.
Dev Cell ; 18(2): 275-87, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20159597

RESUMO

Little is understood about the molecular mechanisms underlying the morphogenesis of the posterior pole of the heart. Here we show that Wnt2 is expressed specifically in the developing inflow tract mesoderm, which generates portions of the atria and atrio-ventricular canal. Loss of Wnt2 results in defective development of the posterior pole of the heart, resulting in a phenotype resembling the human congenital heart syndrome complete common atrio-ventricular canal. The number and proliferation of posterior second heart field progenitors is reduced in Wnt2(-/-) mutants. Moreover, these defects can be rescued in a temporally restricted manner through pharmacological inhibition of Gsk-3beta. We also show that Wnt2 works in a feedforward transcriptional loop with Gata6 to regulate posterior cardiac development. These data reveal a molecular pathway regulating the posterior cardiac mesoderm and demonstrate that cardiovascular defects caused by loss of Wnt signaling can be rescued pharmacologically in vivo.


Assuntos
Coração Fetal/embriologia , Coração Fetal/fisiologia , Fator de Transcrição GATA6/fisiologia , Proteína Wnt2/fisiologia , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Coração Fetal/efeitos dos fármacos , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/fisiopatologia , Humanos , Cloreto de Lítio/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Modelos Cardiovasculares , Fenótipo , Gravidez , Transdução de Sinais , Proteína Wnt2/deficiência , Proteína Wnt2/genética
16.
J Clin Invest ; 119(9): 2538-49, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19690384

RESUMO

Paracrine signaling from lung epithelium to the surrounding mesenchyme is important for lung SMC development and function and is a contributing factor in an array of pulmonary diseases such as bronchopulmonary dysplasia, pulmonary hypertension, and asthma. Wnt7b, which is exclusively expressed in the lung epithelium, is important for lung vascular smooth muscle integrity, but the underlying mechanism by which Wnt signaling regulates lung SMC development is unclear. In this report, we have demonstrated that Wnt7b regulates a program of mesenchymal differentiation in the mouse lung that is essential for SMC development. Genetic loss-of-function studies showed that Wnt7b and beta-catenin were required for expression of Pdgfralpha and Pdgfrbeta and proliferation in pulmonary SMC precursors. In contrast, gain-of-function studies showed that activation of Wnt signaling increased the expression of both Pdgfralpha and Pdgfrbeta as well as the proliferation of SMC precursors. We further showed that the effect on Pdgfr expression was, in part, mediated by direct transcriptional regulation of the ECM protein tenascin C (Tnc), which was necessary and sufficient for Pdgfralpha/beta expression in lung explants. Moreover, this pathway was highly upregulated in a mouse model of asthma and in lung tissue from patients with pulmonary hypertension. Together, these data define a Wnt/Tnc/Pdgfr signaling axis that is critical for smooth muscle development and disease progression in the lung.


Assuntos
Pulmão/embriologia , Pulmão/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Tenascina/metabolismo , Proteínas Wnt/metabolismo , Animais , Asma/metabolismo , Proliferação de Células , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Humanos , Hipertensão Pulmonar/metabolismo , Pulmão/irrigação sanguínea , Pulmão/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Modelos Biológicos , Miócitos de Músculo Liso/citologia , Gravidez , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Proteínas Wnt/deficiência , Proteínas Wnt/genética , beta Catenina/deficiência , beta Catenina/genética , beta Catenina/metabolismo
17.
Dev Cell ; 17(2): 290-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19686689

RESUMO

Patterning of the primitive foregut promotes appropriate organ specification along its anterior-posterior axis. However, the molecular pathways specifying foregut endoderm progenitors are poorly understood. We show here that Wnt2/2b signaling is required to specify lung endoderm progenitors within the anterior foregut. Embryos lacking Wnt2/2b expression exhibit complete lung agenesis and do not express Nkx2.1, the earliest marker of the lung endoderm. In contrast, other foregut endoderm-derived organs, including the thyroid, liver, and pancreas, are correctly specified. The phenotype observed is recapitulated by an endoderm-restricted deletion of beta-catenin, demonstrating that Wnt2/2b signaling through the canonical Wnt pathway is required to specify lung endoderm progenitors within the foregut. Moreover, activation of canonical Wnt/beta-catenin signaling results in the reprogramming of esophagus and stomach endoderm to a lung endoderm progenitor fate. Together, these data reveal that canonical Wnt2/2b signaling is required for the specification of lung endoderm progenitors in the developing foregut.


Assuntos
Sistema Digestório , Pulmão , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Proteínas Wnt/metabolismo , Proteína Wnt2/metabolismo , beta Catenina/metabolismo , Animais , Padronização Corporal/fisiologia , Sistema Digestório/anatomia & histologia , Sistema Digestório/embriologia , Endoderma/citologia , Endoderma/fisiologia , Pulmão/anatomia & histologia , Pulmão/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco/citologia , Proteínas Wnt/genética , Proteína Wnt2/genética , beta Catenina/genética
18.
Development ; 135(5): 789-98, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18263841

RESUMO

Emerging evidence indicates that Wnt signaling regulates crucial aspects of cardiovascular biology (including cardiac morphogenesis, and the self-renewal and differentiation of cardiac progenitor cells). The ability of Wnt signaling to regulate such diverse aspects of cardiovascular development rests on the multifarious downstream and tangential targets affected by this pathway. Here, we discuss the roles for Wnt signaling in cardiac and vascular development, and focus on the emerging role of Wnt signaling in cardiovascular morphogenesis and progenitor cell self-renewal.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Morfogênese/fisiologia , Células-Tronco/fisiologia , Proteínas Wnt/fisiologia , Animais , Feminino , Coração Fetal/fisiologia , Camundongos , Placenta/fisiologia , Gravidez , Transdução de Sinais , Células-Tronco/citologia
19.
Nat Genet ; 40(7): 862-70, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18536717

RESUMO

Epithelial organs, including the lung, are known to possess regenerative abilities through activation of endogenous stem cell populations, but the molecular pathways regulating stem cell expansion and regeneration are not well understood. Here we show that Gata6 regulates the temporal appearance and number of bronchioalveolar stem cells (BASCs) in the lung, its absence in Gata6-null lung epithelium leading to the precocious appearance of BASCs and concurrent loss in epithelial differentiation. This expansion of BASCs was the result of a pronounced increase in canonical Wnt signaling in lung epithelium upon loss of Gata6. Expression of the noncanonical Wnt receptor Fzd2 was downregulated in Gata6 mutants and increased Fzd2 or decreased beta-catenin expression rescued, in part, the lung epithelial defects in Gata6 mutants. During lung epithelial regeneration, canonical Wnt signaling was activated in the niche containing BASCs and forced activation of Wnt signaling led to a large increase in BASC numbers. Moreover, Gata6 was required for proper lung epithelial regeneration, and postnatal loss of Gata6 led to increased BASC expansion and decreased differentiation. Together, these data demonstrate that Gata6-regulated Wnt signaling controls the balance between progenitor expansion and epithelial differentiation required for both lung development and regeneration.


Assuntos
Fator de Transcrição GATA6/fisiologia , Regeneração/genética , Mucosa Respiratória/fisiologia , Células-Tronco/fisiologia , Proteínas Wnt/fisiologia , Animais , Proliferação de Células , Embrião de Mamíferos , Células Epiteliais/fisiologia , Receptores Frizzled/fisiologia , Fator de Transcrição GATA6/genética , Perfilação da Expressão Gênica , Pulmão/embriologia , Pulmão/metabolismo , Pulmão/fisiologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Acoplados a Proteínas G/fisiologia , Regeneração/fisiologia , Mucosa Respiratória/metabolismo , Fenômenos Fisiológicos Respiratórios , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , beta Catenina/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA