Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Int J Mol Sci ; 25(15)2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39125771

RESUMO

Cardiomyopathy is the predominant defect in Barth syndrome (BTHS) and is caused by a mutation of the X-linked Tafazzin (TAZ) gene, which encodes an enzyme responsible for remodeling mitochondrial cardiolipin. Despite the known importance of mitochondrial dysfunction in BTHS, how specific TAZ mutations cause diverse BTHS heart phenotypes remains poorly understood. We generated a patient-tailored CRISPR/Cas9 knock-in mouse allele (TazPM) that phenocopies BTHS clinical traits. As TazPM males express a stable mutant protein, we assessed cardiac metabolic dysfunction and mitochondrial changes and identified temporally altered cardioprotective signaling effectors. Specifically, juvenile TazPM males exhibit mild left ventricular dilation in systole but have unaltered fatty acid/amino acid metabolism and normal adenosine triphosphate (ATP). This occurs in concert with a hyperactive p53 pathway, elevation of cardioprotective antioxidant pathways, and induced autophagy-mediated early senescence in juvenile TazPM hearts. However, adult TazPM males exhibit chronic heart failure with reduced growth and ejection fraction, cardiac fibrosis, reduced ATP, and suppressed fatty acid/amino acid metabolism. This biphasic changeover from a mild-to-severe heart phenotype coincides with p53 suppression, downregulation of cardioprotective antioxidant pathways, and the onset of terminal senescence in adult TazPM hearts. Herein, we report a BTHS genotype/phenotype correlation and reveal that absent Taz acyltransferase function is sufficient to drive progressive cardiomyopathy.


Assuntos
Aciltransferases , Síndrome de Barth , Cardiomiopatias , Síndrome de Barth/genética , Síndrome de Barth/metabolismo , Síndrome de Barth/patologia , Animais , Camundongos , Aciltransferases/genética , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Masculino , Humanos , Mutação Puntual , Modelos Animais de Doenças , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fenótipo
2.
BMC Public Health ; 23(1): 2000, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37833710

RESUMO

BACKGROUND: Chinese urban residents consume more salt from meals prepared outside home than in the past. The purpose of this study is to understand Chinese consumer demand for salt reduction as expressed through their orders on meal delivery apps (MDAs), restaurants' willingness to promote salt reduction, and the extent to which restaurants comply with reduced salt requests. METHODS: We analyzed consumer comments extracted from 718 restaurants on a Chinese MDA called ELEME for orders made in the July-December 2020 timeframe. A self-designed questionnaire was distributed to the restaurant managers to assess restaurants' attitude towards salt reduction upon signing up for the study, and laboratory validation was conducted to test whether dishes ordered with reduced salt requests by consumers actually contained less salt. RESULTS: A total of 25,982 (0.7%) orders out of 3,630,798 orders contained consumer comments. Of the consumer comments, 40.6% (10,549) were about requests for less salt in dishes. Totally 91.5% of 421 surveyed restaurants showed a willingness to respond to consumers' reduced salt requests. The median sodium content measured in the reduced-salt dishes by the laboratory was significantly lower than that in their regular salt counterparts (P < 0.05). CONCLUSIONS: We observed substantial consumer demand for salt reduction while ordering meals on the MDA and that restaurants did, in response, reduce the sodium content in the meals they provided. As meals delivered via MDAs comprise an increasing proportion of outside foods consumed, there is an opportunity for public health experts and policy makers to work with MDAs and restaurants to promote healthier food selections. TRIAL REGISTRATION: ChiCTR2100047729.


Assuntos
Refeições , Restaurantes , Humanos , Cloreto de Sódio na Dieta , Preferências Alimentares , Sódio
3.
Am J Respir Cell Mol Biol ; 64(3): 318-330, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33264084

RESUMO

Pulmonary angiogenesis is a key driver of alveolarization. Our prior studies showed that NF-κB promotes pulmonary angiogenesis during early alveolarization. However, the mechanisms regulating temporal-specific NF-κB activation in the pulmonary vasculature are unknown. To identify mechanisms that activate proangiogenic NF-κB signaling in the developing pulmonary vasculature, proteomic analysis of the lung secretome was performed using two-dimensional difference gel electrophoresis. NF-κB activation and angiogenic function was assessed in primary pulmonary endothelial cells (PECs) and TGFBI (transforming growth factor-ß-induced protein)-regulated genes identified using RNA sequencing. Alveolarization and pulmonary angiogenesis was assessed in wild-type and Tgfbi null mice exposed to normoxia or hyperoxia. Lung TGFBI expression was determined in premature lambs supported by invasive and noninvasive respiratory support. Secreted factors from the early alveolar, but not the late alveolar or adult lung, promoted proliferation and migration in quiescent, adult PECs. Proteomic analysis identified TGFBI as one protein highly expressed by the early alveolar lung that promoted PEC migration by activating NF-κB via αvß3 integrins. RNA sequencing identified Csf3 as a TGFBI-regulated gene that enhances nitric oxide production in PECs. Loss of TGFBI in mice exaggerated the impaired pulmonary angiogenesis induced by chronic hyperoxia, and TGFBI expression was disrupted in premature lambs with impaired alveolarization. Our studies identify TGFBI as a developmentally regulated protein that promotes NF-κB-mediated angiogenesis during early alveolarization by enhancing nitric oxide production. We speculate that dysregulation of TGFBI expression may contribute to diseases marked by impaired alveolar and vascular growth.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Pulmão/irrigação sanguínea , Pulmão/crescimento & desenvolvimento , NF-kappa B/metabolismo , Neovascularização Fisiológica , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos , Movimento Celular , Fatores Estimuladores de Colônias/metabolismo , Células Endoteliais/metabolismo , Integrina alfaVbeta3/metabolismo , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Nascimento Prematuro , Alvéolos Pulmonares/metabolismo , Ovinos
5.
Am J Pathol ; 186(4): 777-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26878215

RESUMO

Septation of the gas-exchange saccules of the morphologically immature mouse lung requires regulated timing, spatial direction, and dosage of transforming growth factor (TGF)-ß signaling. We found that neonatal hyperoxia acutely initially diminished saccular TGF-ß signaling coincident with alveolar simplification. However, sustained hyperoxia resulted in a biphasic response and subsequent up-regulation of TGF-ß signaling, ultimately resulting in bronchopulmonary dysplasia. Significantly, we found that the TGF-ß-induced matricellular protein (TGFBI) was similarly biphasically altered in response to hyperoxia. Moreover, genetic ablation revealed that TGFBI was required for normal alveolar structure and function. Although the phenotype was not neonatal lethal, Tgfbi-deficient lungs were morphologically abnormal. Mutant septal tips were stunted, lacked elastin-positive tips, exhibited reduced proliferation, and contained abnormally persistent alveolar α-smooth muscle actin myofibroblasts. In addition, Tgfbi-deficient lungs misexpressed TGF-ß-responsive follistatin and serpine 1, and transiently suppressed myofibroblast platelet-derived growth factor α differentiation marker. Finally, despite normal lung volume, Tgfbi-null lungs displayed diminished elastic recoil and gas exchange efficiency. Combined, these data demonstrate that initial suppression of the TGF-ß signaling apparatus, as well as loss of key TGF-ß effectors (like TGFBI), underlies early alveolar structural defects, as well as long-lasting functional deficits routinely observed in chronic lung disease of infancy patients. These studies underline the complex (and often contradictory) role of TGF-ß and indicate a need to design studies to associate alterations with initial appearance of phenotypical changes suggestive of bronchopulmonary dysplasia.


Assuntos
Displasia Broncopulmonar/etiologia , Displasia Broncopulmonar/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Hiperóxia/metabolismo , Pulmão/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos , Camundongos , Miofibroblastos/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 111(14): E1374-82, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24706815

RESUMO

Autonomic innervation is an essential component of cardiovascular regulation that is first established from the neural crest (NC) lineage in utero and continues developing postnatally. Although in vitro studies have indicated that SH2-containing protein tyrosine phosphatase 2 (SHP-2) is a signaling factor critical for regulating sympathetic neuron differentiation, this has yet to be shown in the complex in vivo environment of cardiac autonomic innervation. Targeting SHP-2 within postmigratory NC lineages resulted in a fully penetrant mouse model of diminished sympathetic cardiac innervation and concomitant bradycardia. Immunohistochemistry of the sympathetic nerve marker tyrosine hydroxylase revealed a progressive loss of adrenergic ganglionic neurons and reduction of cardiac sympathetic axon density in Shp2 cKOs. Molecularly, Shp2 cKOs exhibit lineage-specific suppression of activated phospo-ERK1/2 signaling but not of other downstream targets of SHP-2 such as pAKT. Genetic restoration of the phosphorylated-extracellular signal-regulated kinase (pERK) deficiency via lineage-specific expression of constitutively active MEK1 was sufficient to rescue the sympathetic innervation deficit and its physiological consequences. These data indicate that SHP-2 signaling specifically through pERK in postmigratory NC lineages is essential for development and maintenance of sympathetic cardiac innervation postnatally.


Assuntos
Coração/inervação , Crista Neural/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Sistema Nervoso Simpático/fisiologia , Animais , Bradicardia/fisiopatologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Camundongos Knockout , Crista Neural/citologia , Neuritos , Proteína Tirosina Fosfatase não Receptora Tipo 11/química , Transdução de Sinais
7.
Development ; 140(9): 1946-57, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23571217

RESUMO

Trabeculation and compaction of the embryonic myocardium are morphogenetic events crucial for the formation and function of the ventricular walls. Fkbp1a (FKBP12) is a ubiquitously expressed cis-trans peptidyl-prolyl isomerase. Fkbp1a-deficient mice develop ventricular hypertrabeculation and noncompaction. To determine the physiological function of Fkbp1a in regulating the intercellular and intracellular signaling pathways involved in ventricular trabeculation and compaction, we generated a series of Fkbp1a conditional knockouts. Surprisingly, cardiomyocyte-restricted ablation of Fkbp1a did not give rise to the ventricular developmental defect, whereas endothelial cell-restricted ablation of Fkbp1a recapitulated the ventricular hypertrabeculation and noncompaction observed in Fkbp1a systemically deficient mice, suggesting an important contribution of Fkbp1a within the developing endocardia in regulating the morphogenesis of ventricular trabeculation and compaction. Further analysis demonstrated that Fkbp1a is a novel negative modulator of activated Notch1. Activated Notch1 (N1ICD) was significantly upregulated in Fkbp1a-ablated endothelial cells in vivo and in vitro. Overexpression of Fkbp1a significantly reduced the stability of N1ICD and direct inhibition of Notch signaling significantly reduced hypertrabeculation in Fkbp1a-deficient mice. Our findings suggest that Fkbp1a-mediated regulation of Notch1 plays an important role in intercellular communication between endocardium and myocardium, which is crucial in controlling the formation of the ventricular walls.


Assuntos
Endocárdio/metabolismo , Ventrículos do Coração/patologia , Miocárdio/metabolismo , Receptor Notch1/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Linhagem da Célula , Células Cultivadas , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Desenvolvimento Embrionário , Endocárdio/embriologia , Endocárdio/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout/embriologia , Camundongos Knockout/metabolismo , Miocárdio/patologia , Crista Neural/metabolismo , Crista Neural/patologia , Fenótipo , Receptor Notch1/genética , Transdução de Sinais , Proteínas de Ligação a Tacrolimo/genética , Transfecção
8.
Circ Res ; 114(9): 1422-34, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24650916

RESUMO

RATIONALE: Cardiac fibroblasts are critical to proper heart function through multiple interactions with the myocardial compartment, but appreciation of their contribution has suffered from incomplete characterization and lack of cell-specific markers. OBJECTIVE: To generate an unbiased comparative gene expression profile of the cardiac fibroblast pool, identify and characterize the role of key genes in cardiac fibroblast function, and determine their contribution to myocardial development and regeneration. METHODS AND RESULTS: High-throughput cell surface and intracellular profiling of cardiac and tail fibroblasts identified canonical mesenchymal stem cell and a surprising number of cardiogenic genes, some expressed at higher levels than in whole heart. While genetically marked fibroblasts contributed heterogeneously to interstitial but not cardiomyocyte compartments in infarcted hearts, fibroblast-restricted depletion of one highly expressed cardiogenic marker, T-box 20, caused marked myocardial dysmorphology and perturbations in scar formation on myocardial infarction. CONCLUSIONS: The surprising transcriptional identity of cardiac fibroblasts, the adoption of cardiogenic gene programs, and direct contribution to cardiac development and repair provoke alternative interpretations for studies on more specialized cardiac progenitors, offering a novel perspective for reinterpreting cardiac regenerative therapies.


Assuntos
Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/genética , Miocárdio/metabolismo , Regeneração/genética , Animais , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/patologia , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA não Traduzido/genética , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
9.
Cancer Cell ; 13(2): 129-40, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18242513

RESUMO

Neurofibromatosis is caused by the loss of neurofibromin (Nf1), leading to peripheral nervous system (PNS) tumors, including neurofibromas and malignant peripheral nerve sheath tumors (MPNSTs). A long-standing question has been whether these tumors arise from neural crest stem cells (NCSCs) or differentiated glia. Germline or conditional Nf1 deficiency caused a transient increase in NCSC frequency and self-renewal in most regions of the fetal PNS. However, Nf1-deficient NCSCs did not persist postnatally in regions of the PNS that developed tumors and could not form tumors upon transplantation into adult nerves. Adult P0a-Cre+Nf1(fl/-) mice developed neurofibromas, and Nf1(+/-)Ink4a/Arf(-/-) and Nf1/p53(+/-) mice developed MPNSTs, but NCSCs did not persist postnatally in affected locations in these mice. Tumors appeared to arise from differentiated glia, not NCSCs.


Assuntos
Neoplasias/patologia , Crista Neural/citologia , Neurofibromina 1/deficiência , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Mutação/genética , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/patologia , Neoplasias de Bainha Neural/patologia , Crista Neural/efeitos dos fármacos , Neurofibroma Plexiforme/patologia , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Sistema Nervoso Periférico/efeitos dos fármacos , Sistema Nervoso Periférico/embriologia , Sistema Nervoso Periférico/metabolismo , Células de Schwann/efeitos dos fármacos , Células de Schwann/patologia , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Proteínas ras/metabolismo
10.
Dev Dyn ; 244(3): 457-67, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25428115

RESUMO

BACKGROUND: Multiple bone morphogenetic protein (BMP) genes are expressed in the developing heart from the initiation to late-differentiation stages, and play pivotal roles in cardiovascular development. In this study, we investigated the requirement of BMP activity in heart development by transgenic over-expression of extracellular BMP antagonist Noggin. RESULTS: Using Nkx2.5-Cre to drive lineage-restricted Noggin within cardiomyocyte progenitors, we show persistent Noggin arrests cardiac development at the linear heart stage. This is coupled with a significantly reduced cell proliferation rate, subsequent cardiomyocyte programmed cell death and reduction of downstream intracellular pSMAD1/5/8 expression. Noggin mutants exhibit reduced heartbeat which likely results in subsequent fully penetrant in utero lethality. Significantly, confocal and electron micrographic examination revealed considerably fewer contractile elements, as well as a lack of maturation of actin-myosin microfilaments. Molecular analysis demonstrated that ectopic Noggin-expressing regions in the early heart's pacemaker region, failed to express the potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4 (Hcn4), resulting in an overall decrease in Hcn4 levels. CONCLUSIONS: Combined, our results reveal a novel role for BMP signaling in the progression of heart development from the tubular heart stage to the looped stage by means of regulation of proliferation and promotion of maturation of the in utero heart's contractile apparatus and pacemaker.


Assuntos
Proteínas de Transporte , Morte Fetal , Regulação da Expressão Gênica no Desenvolvimento , Contração Miocárdica/genética , Miócitos Cardíacos , Células-Tronco , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Feminino , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Gravidez , Células-Tronco/metabolismo , Células-Tronco/patologia
11.
J Neurosci ; 34(17): 6112-22, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24760871

RESUMO

Schwann cells, the myelinating glia of the peripheral nervous system (PNS), originate from multipotent neural crest cells that also give rise to other cells, including neurons, melanocytes, chondrocytes, and smooth muscle cells. The transcription factor Sox10 is required for peripheral glia specification. However, all neural crest cells express Sox10 and the mechanisms directing neural crest cells into a specific lineage are poorly understood. We show here that histone deacetylases 1 and 2 (HDAC1/2) are essential for the specification of neural crest cells into Schwann cell precursors and satellite glia, which express the early determinants of their lineage myelin protein zero (P0) and/or fatty acid binding protein 7 (Fabp7). In neural crest cells, HDAC1/2 induced expression of the transcription factor Pax3 by binding and activating the Pax3 promoter. In turn, Pax3 was required to maintain high Sox10 levels and to trigger expression of Fabp7. In addition, HDAC1/2 were bound to the P0 promoter and activated P0 transcription. Consistently, in vivo genetic deletion of HDAC1/2 in mouse neural crest cells led to strongly decreased Sox10 expression, no detectable Pax3, virtually no satellite glia, and no Schwann cell precursors in dorsal root ganglia and peripheral nerves. Similarly, in vivo ablation of Pax3 in the mouse neural crest resulted in strongly reduced expression of Sox10 and Fabp7. Therefore, by controlling the expression of Pax3 and the concerted action of Pax3 and Sox10 on their target genes, HDAC1/2 direct the specification of neural crest cells into peripheral glia.


Assuntos
Diferenciação Celular/fisiologia , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Células de Schwann/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Camundongos , Crista Neural/citologia , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Células de Schwann/citologia
12.
J Immunol ; 186(8): 4959-66, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21402898

RESUMO

Periostin is a 90-kDa member of the fasciclin-containing family and functions as part of the extracellular matrix. Periostin is expressed in a variety of tissues and expression is increased in airway epithelial cells from asthmatic patients. Recent studies have implicated a role for periostin in allergic eosinophilic esophagitis. To further define a role for periostin in Th2-mediated inflammatory diseases such as asthma, we studied the development of allergic pulmonary inflammation in periostin-deficient mice. Sensitization and challenge of periostin-deficient mice with OVA resulted in increased peripheral Th2 responses compared with control mice. In the lungs, periostin deficiency resulted in increased airway resistance and significantly enhanced mucus production by goblet cells concomitant with increased expression of Gob5 and Muc5ac compared with wild type littermates. Periostin also inhibited the expression of Gob5, a putative calcium-activated chloride channel involved in the regulation of mucus production, in primary murine airway epithelial cells. Our studies suggest that periostin may be part of a negative-feedback loop regulating allergic inflammation that could be therapeutic in the treatment of atopic disease.


Assuntos
Moléculas de Adesão Celular/imunologia , Células Caliciformes/imunologia , Hipersensibilidade/imunologia , Pneumonia/imunologia , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Western Blotting , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Humanos , Hipersensibilidade/genética , Hipersensibilidade/metabolismo , Imuno-Histoquímica , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Metaplasia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muco/imunologia , Muco/metabolismo , Pneumonia/genética , Pneumonia/metabolismo , Baço/citologia , Baço/imunologia , Baço/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
13.
Dev Biol ; 356(2): 308-22, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21600894

RESUMO

Systemic loss-of-function studies have demonstrated that Pax3 transcription factor expression is essential for dorsal neural tube, early neural crest and muscle cell lineage morphogenesis. Cardiac neural crest cells participate in both remodeling of the pharyngeal arch arteries and outflow tract septation during heart development, but the lineage specific role of Pax3 in neural crest function has not yet been determined. To gain insight into the requirement of Pax3 within the neural crest, we conditionally deleted Pax3 in both the premigratory and migratory neural crest populations via Wnt1-Cre and Ap2α-Cre and via P0-Cre in only the migratory neural crest, and compared these phenotypes to the pulmonary atresia phenotype observed following the systemic loss of Pax3. Surprisingly, using Wnt1-Cre deletion there are no resultant heart defects despite the loss of Pax3 from the premigratory and migratory neural crest. In contrast, earlier premigratory and migratory Ap2α-Cre mediated deletion resulted in double outlet right ventricle alignment heart defects. In order to assess the tissue-specific contribution of neural crest to heart development, genetic ablation of neural crest lineage using a Wnt1-Cre-activated diphtheria toxin fragment-A cell-killing system was employed. Significantly, ablation of Wnt1-Cre-expressing neural crest cells resulted in fully penetrant persistent truncus arteriosus malformations. Combined, the data show that Pax3 is essential for early neural crest progenitor formation, but is not required for subsequent cardiac neural crest progeny morphogenesis involving their migration to the heart or septation of the outflow tract.


Assuntos
Coração/embriologia , Morfogênese , Miocárdio/metabolismo , Crista Neural/embriologia , Fatores de Transcrição Box Pareados/fisiologia , Animais , Linhagem da Célula , Movimento Celular , Feminino , Integrases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia , Miócitos Cardíacos/citologia , Fator de Transcrição PAX3 , Proteína Wnt1/fisiologia
14.
Dev Biol ; 344(1): 233-47, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20457144

RESUMO

Smad7 is a negative regulator of TGFbeta superfamily signaling. Using a three-component triple transgenic system, expression of the inhibitory Smad7 was induced via doxycycline within the NCC lineages at pre- and post-migratory stages. Consistent with its role in negatively regulating both TGFbeta and BMP signaling in vitro, induction of Smad7 within the NCC significantly suppressed phosphorylation levels of both Smad1/5/8 and Smad2/3 in vivo, resulting in subsequent loss of NCC-derived craniofacial, pharyngeal and cardiac OFT cushion cells. At the cellular level, increased cell death was observed in pharyngeal arches. However, cell proliferation and NCC-derived smooth muscle differentiation were unaltered. NCC lineage mapping demonstrated that cardiac NCC emigration and initial migration were not affected, but subsequent colonization of the OFT was significantly reduced. Induction of Smad7 in post-migratory NCC resulted in interventricular septal chamber septation defects, suggesting that TGFbeta superfamily signaling is also essential for cardiac NCC at post-migratory stages to govern normal cardiac development. Taken together, the data illustrate that tightly regulated TGFbeta superfamily signaling plays an essential role during craniofacial and cardiac NCC colonization and cell survival in vivo.


Assuntos
Regulação da Expressão Gênica , Crista Neural/metabolismo , Proteína Smad7/genética , Proteína Smad7/fisiologia , Animais , Região Branquial/anormalidades , Doenças Cardiovasculares/congênito , Doenças Cardiovasculares/genética , Linhagem da Célula , Sobrevivência Celular , Anormalidades Craniofaciais/genética , Camundongos , Modelos Biológicos , Modelos Genéticos , Miocárdio/metabolismo , Ativação Transcricional , Transgenes
15.
Circ Res ; 105(10): 934-47, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19893021

RESUMO

Cardiac fibroblasts are the most populous nonmyocyte cell type within the mature heart and are required for extracellular matrix synthesis and deposition, generation of the cardiac skeleton, and to electrically insulate the atria from the ventricles. Significantly, cardiac fibroblasts have also been shown to play an important role in cardiomyocyte growth and expansion of the ventricular chambers during heart development. Although there are currently no cardiac fibroblast-restricted molecular markers, it is generally envisaged that the majority of the cardiac fibroblasts are derived from the proepicardium via epithelial-to-mesenchymal transformation. However, still relatively little is known about when and where the cardiac fibroblasts cells are generated, the lineage of each cell, and how cardiac fibroblasts move to reside in their final position throughout all four cardiac chambers. In this review, we summarize the present understanding regarding the function of Periostin, a useful marker of the noncardiomyocyte lineages, and its role during cardiac morphogenesis. Characterization of the cardiac fibroblast lineage and identification of the signals that maintain, expand and regulate their differentiation will be required to improve our understanding of cardiac function in both normal and pathophysiological states.


Assuntos
Antígenos de Diferenciação/metabolismo , Moléculas de Adesão Celular/metabolismo , Matriz Extracelular/genética , Miocárdio/citologia , Miocárdio/metabolismo , Pericárdio/citologia , Pericárdio/embriologia , Animais , Linhagem da Célula/fisiologia , Fibroblastos , Átrios do Coração/citologia , Átrios do Coração/embriologia , Ventrículos do Coração/citologia , Ventrículos do Coração/embriologia , Humanos , Morfogênese/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo
16.
Birth Defects Res A Clin Mol Teratol ; 91(6): 551-64, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21557455

RESUMO

Although Patch mutants show severe abnormalities in many neural crest-derived structures including the face and the heart, there is a paucity of information characterizing the mechanisms underlying these congenital defects. Via manipulating the genetic background to circumvent early embryonic lethality, our results revealed that Patch phenotypes are most likely due to a significant decrease in migratory neural crest lineage due to diminished neural crest survival and elevated apoptosis. Homozygous mutant neural crest precursors can undergo typical expansion within the neural tube, epithelial-to-mesenchymal transformation, and initiate normal neural crest emigration. Moreover, in vitro explant culture demonstrated that when isolated from the surrounding mesenchyme, Patch mutant neural crest cells (NCCs) can migrate appropriately. Additionally, Patch foregut, notochord and somitic morphogenesis, and Sonic hedgehog expression profiles were all perturbed. Significantly, the timing of lethality and extent of apoptosis correlated with the degree of severity of Patch mutant foregut, notochord, and somite dysfunction. Finally, analysis of Balb/c-enriched surviving Patch mutants revealed that not all the neural crest subpopulations are affected and that Patch mutant neural crest-derived sympathetic ganglia and dorsal root ganglia were unaffected. We hypothesize that loss of normal coordinated signaling from the notochord, foregut, and somites underlies the diminished survival of the neural crest lineage within Patch mutants resulting in subsequent neural crest-deficient phenotypes.


Assuntos
Apoptose , Trato Gastrointestinal/anormalidades , Coração/embriologia , Crista Neural/citologia , Crista Neural/embriologia , Notocorda/anormalidades , Animais , Movimento Celular , Transição Epitelial-Mesenquimal , Gânglios Espinais/embriologia , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Proteínas Hedgehog/biossíntese , Camundongos , Crista Neural/metabolismo , Tubo Neural/embriologia , Notocorda/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais
17.
Cells ; 10(8)2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-34440898

RESUMO

BACKGROUND & AIMS: Liver fibrosis is a pathological healing process resulting from hepatic stellate cell (HSC) activation and the generation of myofibroblasts from activated HSCs. The precise underlying mechanisms of liver fibrogenesis are still largely vague due to lack of understanding the functional heterogeneity of activated HSCs during liver injury. Approach and Results: In this study, to define the mechanism of HSC activation, we performed the transcriptomic analysis at single-cell resolution (scRNA-seq) on HSCs in mice treated with carbon tetrachloride (CCl4). By employing LRAT-Cre:Rosa26mT/mG mice, we were able to isolate an activated GFP-positive HSC lineage derived cell population by fluorescence-activated cell sorter (FACS). A total of 8 HSC subpopulations were identified based on an unsupervised analysis. Each HSC cluster displayed a unique transcriptomic profile, despite all clusters expressing common mouse HSC marker genes. We demonstrated that one of the HSC subpopulations expressed high levels of mitosis regulatory genes, velocity, and monocle analysis indicated that these HSCs are at transitioning and proliferating phases at the beginning of HSCs activation and will eventually give rise to several other HSC subtypes. We also demonstrated cell clusters representing HSC-derived mature myofibroblast populations that express myofibroblasts hallmark genes with unique contractile properties. Most importantly, we found a novel HSC cluster that is likely to be critical in liver regeneration, immune reaction, and vascular remodeling, in which the unique profiles of genes such as Rgs5, Angptl6, and Meg3 are highly expressed. Lastly, we demonstrated that the heterogeneity of HSCs in the injured mouse livers is closely similar to that of cirrhotic human livers. CONCLUSIONS: Collectively, our scRNA-seq data provided insight into the landscape of activated HSC populations and the dynamic transitional pathway from HSC to myofibroblasts in response to liver injury.


Assuntos
Células Estreladas do Fígado/metabolismo , Fígado/metabolismo , Animais , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Componente Principal , Análise de Célula Única , Transcriptoma/genética
18.
J Biol Chem ; 284(40): 27265-72, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19661061

RESUMO

The NCX1 (sodium-calcium exchanger) is up-regulated in human heart failure and in many animal models of heart failure. The potential benefits and risks of therapeutically blocking NCX1 in heart failure and during ischemia-reperfusion are being actively investigated. In this study, we demonstrate that prolonged administration of the NCX1 inhibitor KB-R7943 resulted in the up-regulation of Ncx1 gene expression in both isolated adult cardiomyocytes and intact mouse hearts. Ncx1 up-regulation is mediated by the activation of p38. Importantly, p38 is not activated by KB-R7943 treatment in heart tubes from Ncx1(-/-) mice at 9.5 days postcoitum but is activated in heart tubes from Ncx1(+/+) mice. p38 activation does not appear to be in response to changes in cytosolic calcium concentration, [Ca(2+)](i). Interestingly, chronic KB-R7943 treatment in mice leads to the formation of an NCX1-p38 complex. Our study demonstrates for the first time that the electrogenic sarcolemma membrane cardiac NCX1 can act as a regulator of "activity-dependent signal transduction" leading to changes in gene expression.


Assuntos
Coração/efeitos dos fármacos , Miocárdio/metabolismo , Trocador de Sódio e Cálcio/genética , Tioureia/análogos & derivados , Regulação para Cima/efeitos dos fármacos , Antagonistas Adrenérgicos beta/administração & dosagem , Antagonistas Adrenérgicos beta/farmacologia , Animais , Antiarrítmicos/administração & dosagem , Antiarrítmicos/farmacologia , Bloqueadores dos Canais de Cálcio/administração & dosagem , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Gatos , Ativação Enzimática/efeitos dos fármacos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Tioureia/administração & dosagem , Tioureia/farmacologia , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Circ Res ; 102(7): 752-60, 2008 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-18296617

RESUMO

The secreted periostin protein, which marks mesenchymal cells in endocardial cushions following epithelial-mesenchymal transformation and in mature valves following remodeling, is a putative valvulogenesis target molecule. Indeed, periostin is expressed throughout cardiovascular morphogenesis and in all 4 adult mice valves (annulus and leaflets). Additionally, periostin is expressed throughout the fibrous cardiac skeleton and endocardial cushions in the developing heart but is absent from both normal and/or pathological mouse cardiomyocytes. Periostin (peri(lacZ)) knockout mice exhibit viable valve disease, with neonatal lethality in a minority and latent disease with leaflet abnormalities in the viable majority. Surviving peri(lacZ)-null leaflets are truncated, contain ectopic cardiomyocytes and smooth muscle, misexpress the cartilage proteoglycan aggrecan, demonstrate disorganized matrix stratification, and exhibit reduced transforming growth factor-beta signaling. Neonatal peri(lacZ) nulls that die (14%) display additional defects, including leaflet discontinuities, delamination defects, and deposition of acellular extracellular matrix. Assessment of collagen production, 3D lattice formation ability, and transforming growth factor-beta responsiveness indicate periostin-deficient fibroblasts are unable to support normal valvular remodeling and establishment of a mature cardiac skeleton. Furthermore, pediatric stenotic bicuspid aortic valves that have lost normal extracellular matrix trilaminar stratification have greatly reduced periostin. This suggests that loss of periostin results in inappropriate differentiation of mesenchymal cushion cells and valvular abnormalities via a transforming growth factor-beta-dependent pathway during establishment of the mature heart. Thus, peri(lacZ) knockouts provide a new model of viable latent valve disease.


Assuntos
Moléculas de Adesão Celular/metabolismo , Matriz Extracelular/metabolismo , Mesoderma/metabolismo , Mesoderma/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Modelos Animais de Doenças , Endocárdio/metabolismo , Endocárdio/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Doenças das Valvas Cardíacas/metabolismo , Doenças das Valvas Cardíacas/patologia , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fator de Crescimento Transformador beta/metabolismo
20.
J Dev Biol ; 8(3)2020 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-32962040

RESUMO

Testicular development starts in utero and maturation continues postnatally, requiring a cascade of gene activation and differentiation into different cell types, with each cell type having its own specific function. As we had previously reported that the Capping protein inhibiting regulator of actin (Cracd) gene was expressed in the adult mouse testis, herein we examine when and where the ß-catenin associated Cracd is initially expressed during postnatal testis development. Significantly, Cracd mRNA is present in both the immature postnatal and adult testis in round spermatid cells, with highest level of expression occurring during the first wave of meiosis and spermatogenesis. In the juvenile testes, Cracd is initially expressed within the innermost region but as maturation occurs, Cracd mRNA switches to a more peripheral location. Thereafter, Cracd is downregulated to maintenance levels in the haploid male germ cell lineage. As Cracd mRNA was expressed within developing round spermatids, we tested its effectiveness as a biomarker of non-obstructive azoospermia using transgenic knockout mice models. Meaningfully, Cracd expression was absent in Deleted in azoospermia like (Dazl) null testis, which exhibit a dramatic germ cell loss. Moreover, Cracd was abnormally regulated and ectopically mis-expressed in Polypyrimidine tract binding protein-2 (Ptbp2) conditional germ cell restricted knockout testis, which exhibit a block during spermatid differentiation and a reduction in the number of late stage spermatocytes coincident with reduced ß-catenin expression. Combined, these data suggest that Cracd is a useful first wave of spermatogenesis biomarker of azoospermia phenotypes, even prior to an overt phenotype being evident.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA