Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Nature ; 618(7966): 818-826, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37316669

RESUMO

Correct development and maturation of the enteric nervous system (ENS) is critical for survival1. At birth, the ENS is immature and requires considerable refinement to exert its functions in adulthood2. Here we demonstrate that resident macrophages of the muscularis externa (MMϕ) refine the ENS early in life by pruning synapses and phagocytosing enteric neurons. Depletion of MMϕ before weaning disrupts this process and results in abnormal intestinal transit. After weaning, MMϕ continue to interact closely with the ENS and acquire a neurosupportive phenotype. The latter is instructed by transforming growth factor-ß produced by the ENS; depletion of the ENS and disruption of transforming growth factor-ß signalling result in a decrease in neuron-associated MMϕ associated with loss of enteric neurons and altered intestinal transit. These findings introduce a new reciprocal cell-cell communication responsible for maintenance of the ENS and indicate that the ENS, similarly to the brain, is shaped and maintained by a dedicated population of resident macrophages that adapts its phenotype and transcriptome to the timely needs of the ENS niche.


Assuntos
Sistema Nervoso Entérico , Intestinos , Macrófagos , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/crescimento & desenvolvimento , Sistema Nervoso Entérico/fisiologia , Intestinos/inervação , Linfotoxina-alfa/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiologia , Neurônios/fisiologia , Desmame , Comunicação Celular , Transcriptoma , Fenótipo , Fagocitose , Sinapses , Plasticidade Neuronal , Trânsito Gastrointestinal
2.
J Cell Physiol ; 236(11): 7440-7449, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34041746

RESUMO

Cardiac fibrosis accompanies a number of pathological conditions and results in altered myocardial structure, biomechanical properties and function. The signaling networks leading to fibrosis are complex, contributing to the general lack of progress in identifying effective therapeutic approaches to prevent or reverse this condition. Several studies have shown protective effects of emodin, a plant-derived anthraquinone, in animal models of fibrosis. A number of questions remain regarding the mechanisms whereby emodin impacts fibrosis. Transforming growth factor beta 1 (TGF-ß1) is a potent stimulus of fibrosis and fibroblast activation. In the present study, experiments were performed to evaluate the effects of emodin on activation and function of cardiac fibroblasts following treatment with TGF-ß1. We demonstrate that emodin attenuates TGF-ß1-induced fibroblast activation and collagen accumulation in vitro. Emodin also inhibits activation of several canonical (SMAD2/3) and noncanonical (Erk1/2) TGF-ß signaling pathways, while activating the p38 pathway. These results suggest that emodin may provide an effective therapeutic agent for fibrosis that functions via specific TGF-ß signaling pathways.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Emodina/farmacologia , Fibroblastos/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Animais , Células Cultivadas , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miocárdio/citologia , Ratos Sprague-Dawley , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 40(8): 1870-1890, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32493169

RESUMO

OBJECTIVE: Neointima formation is a primary cause of intermediate to late vein graft (VG) failure. However, the precise source of neointima cells in VGs remains unclear. Approach and Results: Herein we clarify the relative contributions of mature vascular smooth muscle cells (SMCs) and endothelial cells (ECs) to neointima formation in a mouse model of VG remodeling via the genetic-inducible fate mapping approaches. Regardless of the magnitude of neointima formation, the recipient arterial and the donor venous SMCs contributed ≈55% of the neointima cells at the anastomotic regions, whereas only donor venous SMCs donated ≈68% of the neointima cells at the middle bodies. A small portion of the SMC-derived cells became non-SMC cells, most likely vascular stem cells, and constituted 2% to 11% of the cells in each major layer of VGs. In addition, the recipient arterial ECs were the major cellular source of re-endothelialization but did not contribute to neointima formation. The donor venous ECs donated ≈17% neointima cells in the VGs with mild neointima formation and conditional media from ECs after endothelial-to-mesenchymal transition suppressed vascular SMC dedifferentiation. CONCLUSIONS: The recipient arterial and donor venous mature SMCs dominate but contribute distinctly to intimal hyperplasia at the anastomosis and the middle body regions of VGs. The recipient arterial ECs are the major cellular source of re-endothelialization but do not donate neointima formation in VGs. Only the donor venous ECs undergo endothelial-to-mesenchymal transition. Endothelial-to-mesenchymal transition is marginal for generating neointima cells but is likely required for controlling the quality of VG remodeling.


Assuntos
Células Endoteliais/patologia , Veias Jugulares/transplante , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Neointima/patologia , Animais , Hiperplasia , Mesoderma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Remodelação Vascular
4.
J Cell Physiol ; 234(8): 13931-13941, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30609032

RESUMO

An important step in many pathological conditions, particularly tissue and organ fibrosis, is the conversion of relatively quiescent cells into active myofibroblasts. These are highly specialized cells that participate in normal wound healing but also contribute to pathogenesis. These cells possess characteristics of smooth muscle cells and fibroblasts, have enhanced synthetic activity secreting abundant extracellular matrix components, cytokines, and growth factors, and are capable of generating contractile force. As such, these cells have become potential therapeutic targets in a number of disease settings. Transforming growth factor ß (TGF-ß) is a potent stimulus of fibrosis and myofibroblast formation and likewise is an important therapeutic target in several disease conditions. The plant-derived isothiocyanate sulforaphane has been shown to have protective effects in several pathological models including diabetic cardiomyopathy, carcinogenesis, and fibrosis. These studies suggest that sulforaphane may be an attractive preventive agent against disease progression, particularly in conditions involving alterations of the extracellular matrix and activation of myofibroblasts. However, few studies have evaluated the effects of sulforaphane on cardiac fibroblast activation and their interactions with the extracellular matrix. The present studies were carried out to determine the potential effects of sulforaphane on the conversion of quiescent cardiac fibroblasts to an activated myofibroblast phenotype and associated alterations in signaling, expression of extracellular matrix receptors, and cellular physiology following stimulation with TGF-ß1. These studies demonstrate that sulforaphane attenuates TGF-ß1-induced myofibroblast formation and contractile activity. Sulforaphane also reduces expression of collagen-binding integrins and inhibits canonical and noncanonical TGF-ß signaling pathways.


Assuntos
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Isotiocianatos/farmacologia , Miocárdio/citologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Bovinos , Colágeno/farmacologia , Matriz Extracelular/efeitos dos fármacos , Proteínas da Matriz Extracelular/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Hidrogéis/farmacologia , Integrinas/metabolismo , Masculino , Metaloproteinases da Matriz/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sulfóxidos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
5.
Lab Invest ; 99(9): 1363-1375, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31028279

RESUMO

Proper lung development depends on the precise temporal and spatial expression of several morphogenic factors, including Fgf10, Fgf9, Shh, Bmp4, and Tgf-ß. Over- or under-expression of these molecules often leads to aberrant embryonic or postnatal lung development. Herein, we deleted the Tgf-ß1 gene specifically within the lung embryonic mesenchymal compartment at specific gestational stages to determine the contribution of this cytokine to lung development. Mutant embryos developed severe lung hypoplasia and died at birth due to the inability to breathe. Despite the markedly reduced lung size, proliferation and differentiation of the lung epithelium was not affected by the lack of mesenchymal expression of the Tgf-ß1 gene, while apoptosis was significantly increased in the mutant lung parenchyma. Lack of mesenchymal expression of the Tgf-ß1 gene was also associated with reduced lung branching morphogenesis, with accompanying inhibition of the local FGF10 signaling pathway as well as abnormal development of the vascular system. To shed light on the mechanism of lung hypoplasia, we quantified the phosphorylation of 226 proteins in the mutant E12.5 lung compared with control. We identified five proteins, Hrs, Vav2, c-Kit, the regulatory subunit of Pi3k (P85), and Fgfr1, that were over- or under-phosphorylated in the mutant lung, suggesting that they could be indispensable effectors of the TGF-ß signaling program during embryonic lung development. In conclusion, we have uncovered novel roles of the mesenchyme-specific Tgf-ß1 ligand in embryonic mouse lung development and generated a mouse model that may prove helpful to identify some of the key pathogenic mechanisms underlying lung hypoplasia in humans.


Assuntos
Técnicas de Inativação de Genes/métodos , Pulmão/embriologia , Mesoderma/embriologia , Morfogênese/genética , Fator de Crescimento Transformador beta1 , Animais , Animais Recém-Nascidos , Apoptose , Técnicas de Cultura de Células , Feminino , Pulmão/patologia , Pneumopatias/genética , Pneumopatias/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
6.
Dev Biol ; 398(2): 231-41, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25523394

RESUMO

Transforming growth factor-beta3 (TGF-ß3) plays a critical role in palatal epithelial cells by inducing palatal epithelial fusion, failure of which results in cleft palate, one of the most common birth defects in humans. Recent studies have shown that Smad-dependent and Smad-independent pathways work redundantly to transduce TGF-ß3 signaling in palatal epithelial cells. However, detailed mechanisms by which this signaling is mediated still remain to be elucidated. Here we show that TGF-ß activated kinase-1 (Tak1) and Smad4 interact genetically in palatal epithelial fusion. While simultaneous abrogation of both Tak1 and Smad4 in palatal epithelial cells resulted in characteristic defects in the anterior and posterior secondary palate, these phenotypes were less severe than those seen in the corresponding Tgfb3 mutants. Moreover, our results demonstrate that Trim33, a novel chromatin reader and regulator of TGF-ß signaling, cooperates with Smad4 during palatogenesis. Unlike the epithelium-specific Smad4 mutants, epithelium-specific Tak1:Smad4- and Trim33:Smad4-double mutants display reduced expression of Mmp13 in palatal medial edge epithelial cells, suggesting that both of these redundant mechanisms are required for appropriate TGF-ß signal transduction. Moreover, we show that inactivation of Tak1 in Trim33:Smad4 double conditional knockouts leads to the palatal phenotypes which are identical to those seen in epithelium-specific Tgfb3 mutants. To conclude, our data reveal added complexity in TGF-ß signaling during palatogenesis and demonstrate that functionally redundant pathways involving Smad4, Tak1 and Trim33 regulate palatal epithelial fusion.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Palato/embriologia , Palato/metabolismo , Transdução de Sinais , Proteína Smad4/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Animais , Apoptose/genética , Fusão Celular , Proliferação de Células , Cruzamentos Genéticos , Embrião de Mamíferos/metabolismo , Ativação Enzimática , Células Epiteliais/metabolismo , Epitélio/metabolismo , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Camundongos Knockout , Modelos Biológicos , Mutação/genética , Especificidade de Órgãos , Palato/anormalidades , Palato/enzimologia
7.
Genesis ; 52(9): 817-26, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24895296

RESUMO

Transforming growth factor beta2 (TGFß2) is a multifunctional protein which is expressed in several embryonic and adult organs. TGFB2 mutations can cause Loeys Dietz syndrome, and its dysregulation is involved in cardiovascular, skeletal, ocular, and neuromuscular diseases, osteoarthritis, tissue fibrosis, and various forms of cancer. TGFß2 is involved in cell growth, apoptosis, cell migration, cell differentiation, cell-matrix remodeling, epithelial-mesenchymal transition, and wound healing in a highly context-dependent and tissue-specific manner. Tgfb2(-/-) mice die perinatally from congenital heart disease, precluding functional studies in adults. Here, we have generated mice harboring Tgfb2(ßgeo) (knockout-first lacZ-tagged insertion) gene-trap allele and Tgfb2(flox) conditional allele. Tgfb2(ßgeo/ßgeo) or Tgfb2(ßgeo/-) mice died at perinatal stage from the same congenital heart defects as Tgfb2(-/-) mice. ß-galactosidase staining successfully detected Tgfb2 expression in the heterozygous Tgfb2(ßgeo) fetal tissue sections. Tgfb2(flox) mice were produced by crossing the Tgfb2(+/ßgeo) mice with the FLPeR mice. Tgfb2(flox/-) mice were viable. Tgfb2 conditional knockout (Tgfb2(cko/-) ) fetuses were generated by crossing of Tgfb2(flox/-) mice with Tgfb2(+/-) ; EIIaCre mice. Systemic Tgfb2(cko/-) embryos developed cardiac defects which resembled the Tgfb2(ßgeo/ßgeo) , Tgfb2(ßgeo/-) , and Tgfb2(-/-) fetuses. In conclusion, Tgfb2(ßgeo) and Tgfb2(flox) mice are novel mouse strains which will be useful for investigating the tissue specific expression and function of TGFß2 in embryonic development, adult organs, and disease pathogenesis and cancer. genesis


Assuntos
Coração/embriologia , Camundongos Knockout , Fator de Crescimento Transformador beta2/genética , Alelos , Animais , Apoptose , Diferenciação Celular , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Feminino , Coração/crescimento & desenvolvimento , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Fator de Crescimento Transformador beta2/metabolismo , beta-Galactosidase
8.
Circ Res ; 110(11): 1498-512, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22628574

RESUMO

Mouse genetic engineering has revolutionized our understanding of the molecular and genetic basis of heart development and disease. This technology involves conditional tissue-specific and temporal transgenic and gene targeting approaches, as well as introduction of polymorphisms into the mouse genome. These approaches are increasingly used to elucidate the genetic pathways underlying tissue homeostasis, physiology, and pathophysiology of adult heart. They have also led to the development of clinically relevant models of human cardiac diseases. Here, we review the technologies and their limitations in general and the cardiovascular research community in particular.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Cardiopatias/genética , Coração , Processamento Alternativo , Animais , Desoxirribonucleases/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Coração/embriologia , Coração/fisiopatologia , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/patologia , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Crista Neural/metabolismo , Crista Neural/patologia , Pericárdio/metabolismo , Pericárdio/patologia , Polimorfismo de Nucleotídeo Único
9.
J Mol Cell Cardiol ; 65: 137-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24157418

RESUMO

Mature heart valves are complex structures consisting of three highly organized extracellular matrix layers primarily composed of collagens, proteoglycans and elastin. Collectively, these diverse matrix components provide all the necessary biomechanical properties for valve function throughout life. In contrast to healthy valves, myxomatous valve disease is the most common cause of mitral valve prolapse in the human population and is characterized by an abnormal abundance of proteoglycans within the valve tri-laminar structure. Despite the clinical significance, the etiology of this phenotype is not known. Scleraxis (Scx) is a basic-helix-loop-helix transcription factor that we previously showed to be required for establishing heart valve structure during remodeling stages of valvulogenesis. In this study, we report that remodeling heart valves from Scx null mice express decreased levels of proteoglycans, particularly chondroitin sulfate proteoglycans (CSPGs), while overexpression in embryonic avian valve precursor cells and adult porcine valve interstitial cells increases CSPGs. Using these systems we further identify that Scx is positively regulated by canonical Tgfß2 signaling during this process and this is attenuated by MAPK activity. Finally, we show that Scx is increased in myxomatous valves from human patients and mouse models, and overexpression in human mitral valve interstitial cells modestly increases proteoglycan expression consistent with myxomatous mitral valve phenotypes. Together, these studies identify an important role for Scx in regulating proteoglycans in embryonic and mature valve cells and suggest that imbalanced regulation could influence myxomatous pathogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Valvas Cardíacas/metabolismo , Sistema de Sinalização das MAP Quinases , Proteoglicanas/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos , Galinhas , Modelos Animais de Doenças , Valvas Cardíacas/embriologia , Valvas Cardíacas/patologia , Humanos , Camundongos , Valva Mitral/embriologia , Valva Mitral/metabolismo , Valva Mitral/patologia , Células NIH 3T3 , Sus scrofa
10.
J Cardiovasc Dev Dis ; 10(12)2023 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-38132651

RESUMO

The transforming growth factor beta (TGFß) and Hippo signaling pathways are evolutionarily conserved pathways that play a critical role in cardiac fibroblasts during embryonic development, tissue repair, and fibrosis. TGFß signaling and Hippo signaling are also important for cardiac cushion remodeling and septation during embryonic development. Loss of TGFß2 in mice causes cardiac cushion remodeling defects resulting in congenital heart disease. In this study, we used in vitro molecular and pharmacologic approaches in the cushion mesenchymal cell line (tsA58-AVM) and investigated if the Hippo pathway acts as a mediator of TGFß2 signaling. Immunofluorescence staining showed that TGFß2 induced nuclear translocation of activated SMAD3 in the cushion mesenchymal cells. In addition, the results indicate increased nuclear localization of Yes-associated protein 1 (YAP1) following a similar treatment of TGFß2. In collagen lattice formation assays, the TGFß2 treatment of cushion cells resulted in an enhanced collagen contraction compared to the untreated cushion cells. Interestingly, verteporfin, a YAP1 inhibitor, significantly blocked the ability of cushion cells to contract collagen gel in the absence or presence of exogenously added TGFß2. To confirm the molecular mechanisms of the verteporfin-induced inhibition of TGFß2-dependent extracellular matrix (ECM) reorganization, we performed a gene expression analysis of key mesenchymal genes involved in ECM remodeling in heart development and disease. Our results confirm that verteporfin significantly decreased the expression of α-smooth muscle actin (Acta2), collagen 1a1 (Col1a1), Ccn1 (i.e., Cyr61), and Ccn2 (i.e., Ctgf). Western blot analysis indicated that verteporfin treatment significantly blocked the TGFß2-induced activation of SMAD2/3 in cushion mesenchymal cells. Collectively, these results indicate that TGFß2 regulation of cushion mesenchymal cell behavior and ECM remodeling is mediated by YAP1. Thus, the TGFß2 and Hippo pathway integration represents an important step in understanding the etiology of congenital heart disease.

11.
Eur Radiol Exp ; 7(1): 1, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36617620

RESUMO

BACKGROUND: To assess the impact of the new version of a deep learning (DL) spectral reconstruction on image quality of virtual monoenergetic images (VMIs) for contrast-enhanced abdominal computed tomography in the rapid kV-switching platform. METHODS: Two phantoms were scanned with a rapid kV-switching CT using abdomen-pelvic CT examination parameters at dose of 12.6 mGy. Images were reconstructed using two versions of DL spectral reconstruction algorithms (DLSR V1 and V2) for three reconstruction levels. The noise power spectrum (NSP) and task-based transfer function at 50% (TTF50) were computed at 40/50/60/70 keV. A detectability index (d') was calculated for enhanced lesions at low iodine concentrations: 2, 1, and 0.5 mg/mL. RESULTS: The noise magnitude was significantly lower with DLSR V2 compared to DLSR V1 for energy levels between 40 and 60 keV by -36.5% ± 1.4% (mean ± standard deviation) for the standard level. The average NPS frequencies increased significantly with DLSR V2 by 23.7% ± 4.2% for the standard level. The highest difference in TTF50 was observed at the mild level with a significant increase of 61.7% ± 11.8% over 40-60 keV energy with DLSR V2. The d' values were significantly higher for DLSR V2 versus DLSR V1. CONCLUSIONS: The DLSR V2 improves image quality and detectability of low iodine concentrations in VMIs compared to DLSR V1. This suggests a great potential of DLSR V2 to reduce iodined contrast doses.


Assuntos
Aprendizado Profundo , Iodo , Tomografia Computadorizada por Raios X/métodos , Algoritmos , Processamento de Imagem Assistida por Computador/métodos
12.
Genesis ; 50(1): 59-66, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22223248

RESUMO

The transforming growth factor beta (TGFß) pathway is involved in embryonic development and several inherited and acquired human diseases. The gene for TGFß3 (Tgfb3) encodes one of the three ligands for TGFß receptors. It is widely expressed in the embryo and its mutation or misexpression is found in human diseases. Tgfb3-/- mice die at birth from cleft palate, precluding functional studies in adults. Here, we generated mice in which exon 6 of Tgfb3 was flanked with LoxP sites (Tgfb3flox/flox). The adult mice were normal and fertile. EIIa-Cre-mediated deletion of exon 6 in Tgfb3flox/flox mice efficiently generated Tgfb3 conditional knockout (Tgfb3cko/cko) mice which died at birth from the same cleft palate defect as Tgfb3-/- mice, indicating that the conditional and knockout alleles are functionally equivalent. This Tgfb3cko allele will now enable studies of TGFß3 function in different cell or tissue types in embryonic development and during adulthood.


Assuntos
Alelos , Camundongos Knockout , Fator de Crescimento Transformador beta3/genética , Animais , Fissura Palatina/embriologia , Éxons , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Análise de Sequência de DNA , Fator de Crescimento Transformador beta3/metabolismo
13.
Cell Tissue Res ; 347(1): 203-23, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21953136

RESUMO

The majority of children with congenital heart disease now live into adulthood due to the remarkable surgical and medical advances that have taken place over the past half century. Because of this, adults now represent the largest age group with adult cardiovascular diseases. It includes patients with heart diseases that were not detected or not treated during childhood, those whose defects were surgically corrected but now need revision due to maladaptive responses to the procedure, those with exercise problems and those with age-related degenerative diseases. Because adult cardiovascular diseases in this population are relatively new, they are not well understood. It is therefore necessary to understand the molecular and physiological pathways involved if we are to improve treatments. Since there is a developmental basis to adult cardiovascular disease, transforming growth factor beta (TGFß) signaling pathways that are essential for proper cardiovascular development may also play critical roles in the homeostatic, repair and stress response processes involved in adult cardiovascular diseases. Consequently, we have chosen to summarize the current information on a subset of TGFß ligand and receptor genes and related effector genes that, when dysregulated, are known to lead to cardiovascular diseases and adult cardiovascular deficiencies and/or pathologies. A better understanding of the TGFß signaling network in cardiovascular disease and repair will impact genetic and physiologic investigations of cardiovascular diseases in elderly patients and lead to an improvement in clinical interventions.


Assuntos
Reabilitação Cardíaca , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Envelhecimento/fisiologia , Angiotensina II/metabolismo , Animais , Doenças Cardiovasculares/terapia , Transição Epitelial-Mesenquimal/fisiologia , Expressão Gênica , Variação Genética , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo
14.
Cell Tissue Res ; 347(1): 267-77, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22105919

RESUMO

Aortic aneurysm is predominantly found in the ascending aorta in patients with Marfan syndrome (MFS). However, descending aortic disease has emerged as a problem since people are living longer because of improved medical and surgical management of the ascending aorta. Diagnostic procedures before disease onset and the mechanisms involved in the transition of normal aortic tissue to aneurysm remain unclear. We determined signs of descending aortic disease before disease onset in mice with a mutation in the fibrillin 1 gene (Fbn1(+/C1039G)), a validated mouse model of disease susceptibility and progression of aortic aneurysm of MFS. We analyzed a tubular unfixed non-aneurysmal descending thoracic aorta from 8-month-old wild-type and Fbn1(+/C1039G) mice by a tubular biaxial tester that works in conjunction with a two-photon nonlinear microscope. Fbn1(+/C1039G) mouse aorta was more compliant in the circumferential direction. Two-photon imaging showed defective organization of adventitial collagen fibers in the pressurized aortas of Fbn1(+/C1039G) mice. Moreover, disruption in the elastic lamina was noted in the absence of aneurysms in pressurized aortas but not unpressurized aortas of Fbn1(+/C1039G) mice. At the molecular level, this altered tissue behavior in non-aneurysmal descending aortas of Fbn1(+/C1039G) mice was accompanied by an increasing trend of canonical but not noncanonical, transforming growth factor-ß (TGFß) signaling. Finally, assays of in vitro collagen lattice formation in mouse wild-type and TGFß1-deficient embryonic fibroblasts indicate that TGFß1 can regulate collagen organization. The ability to reveal the presence of altered biomechanics and microstructure coupled with subtle changes in TGFß signaling provides a novel surrogate measure of tissue susceptibility to aneurysm before disease onset.


Assuntos
Aorta Torácica/patologia , Modelos Animais de Doenças , Síndrome de Marfan/patologia , Doenças Vasculares/patologia , Animais , Aorta Torácica/fisiologia , Aneurisma da Aorta Torácica/etiologia , Fibrilina-1 , Fibrilinas , Humanos , Síndrome de Marfan/complicações , Síndrome de Marfan/fisiopatologia , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Estresse Mecânico , Doenças Vasculares/etiologia
15.
Dev Dyn ; 240(9): 2127-41, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21780244

RESUMO

Although the function of transforming growth factor beta2 (TGFß2) in epithelial mesenchymal transition (EMT) is well studied, its role in valve remodeling remains to be fully explored. Here, we used histological, morphometric, immunohistochemical and molecular approaches and showed that significant dysregulation of major extracellular matrix (ECM) components contributed to valve remodeling defects in Tgfb2(-/-) embryos. The data indicated that cushion mesenchymal cell differentiation was impaired in Tgfb2(-/-) embryos. Hyaluronan and cartilage link protein-1 (CRTL1) were increased in hyperplastic valves of Tgfb2(-/-) embryos, indicating increased expansion and diversification of cushion mesenchyme into the cartilage cell lineage during heart development. Finally, Western blot and immunohistochemistry analyses indicate that the activation of SMAD2/3 was decreased in Tgfb2(-/-) embryos during valve remodeling. Collectively, the data indicate that TGFß2 promotes valve remodeling and differentiation by inducing matrix organization and suppressing cushion mesenchyme differentiation into cartilage cell lineage during heart development.


Assuntos
Valvas Cardíacas/metabolismo , Coração/embriologia , Fator de Crescimento Transformador beta2/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Matriz Extracelular/metabolismo , Valvas Cardíacas/embriologia , Imuno-Histoquímica , Mesoderma/citologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta2/genética
16.
Front Cardiovasc Med ; 9: 770065, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35928937

RESUMO

Aims: Calcific aortic valve disease (CAVD) is a progressive heart disease that is particularly prevalent in elderly patients. The current treatment of CAVD is surgical valve replacement, but this is not a permanent solution, and it is very challenging for elderly patients. Thus, a pharmacological intervention for CAVD may be beneficial. In this study, we intended to rescue aortic valve (AV) calcification through inhibition of TGFß1 and SMAD3 signaling pathways. Methods and Results: The klotho gene, which was discovered as an aging-suppressor gene, has been observed to play a crucial role in AV calcification. The klotho knockout (Kl -/-) mice have shorter life span (8-12 weeks) and develop severe AV calcification. Here, we showed that increased TGFß1 and TGFß-dependent SMAD3 signaling were associated with AV calcification in Kl -/- mice. Next, we generated Tgfb1- and Smad3-haploinsufficient Kl -/- mice to determine the contribution of TGFß1 and SMAD3 to the AV calcification in Kl -/- mice. The histological and morphometric evaluation suggested a significant reduction of AV calcification in Kl -/-; Tgfb1 ± mice compared to Kl -/- mice. Smad3 heterozygous deletion was observed to be more potent in reducing AV calcification in Kl -/- mice compared to the Kl -/-; Tgfb1 ± mice. We observed significant inhibition of Tgfb1, Pai1, Bmp2, Alk2, Spp1, and Runx2 mRNA expression in Kl -/-; Tgfb1 ± and Kl -/-; Smad3 ± mice compared to Kl -/- mice. Western blot analysis confirmed that the inhibition of TGFß canonical and non-canonical signaling pathways were associated with the rescue of AV calcification of both Kl -/-; Tgfb1 ± and Kl -/-; Smad3 ± mice. Conclusion: Overall, inhibition of the TGFß1-dependent SMAD3 signaling pathway significantly blocks the development of AV calcification in Kl -/- mice. This information is useful in understanding the signaling mechanisms involved in CAVD.

17.
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
18.
J Biomech Eng ; 133(7): 075001, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21823753

RESUMO

Murine models of disease are a powerful tool for researchers to gain insight into disease formation, progression, and therapies. The biomechanical indicators of diseased tissue provide a unique insight into some of these murine models, since the biomechanical properties in scenarios such as aneurysm and Marfan syndrome can dictate tissue failure and mortality. Understanding the properties of the tissue on the macroscopic scale has been shown to be important, as one can then understand the tissue's ability to withstand the high stresses seen in the cardiac pulsatile cycle. Alterations in the biomechanical response can foreshadow prospective mechanical failure of the tissue. These alterations are often seen on the microstructural level, and obtaining detailed information on such changes can offer a better understanding of the phenomena seen on the macroscopic level. Unfortunately, mouse models present problems due to the size and delicate features in the mechanical testing of such tissues. In addition, some smaller arteries in large-animal studies (e.g., coronary and cerebral arteries) can present the same issues, and are sometimes unsuitable for planar biaxial testing. The purpose of this paper is to present a robust method for the investigation of the mechanical properties of small arteries and the classification of the microstructural orientation and degree of fiber alignment. This occurs through the cost-efficient modification of a planar biaxial tester that works in conjunction with a two-photon nonlinear microscope. This system provides a means to further investigate how microstructure and mechanical properties are modified in diseased transgenic animals where the tissue is in small tube form. Several other hard-to-test tubular specimens such as cerebral aneurysm arteries and atherosclerotic coronary arteries can also be tested using the described modular device.


Assuntos
Aorta/citologia , Aorta/fisiologia , Colágeno/fisiologia , Vasos Coronários/citologia , Vasos Coronários/fisiologia , Microscopia/instrumentação , Animais , Fenômenos Biomecânicos , Colágeno/ultraestrutura , Elastina/metabolismo , Elastina/ultraestrutura , Desenho de Equipamento , Humanos , Camundongos , Microscopia/métodos , Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Design de Software , Estresse Mecânico
19.
Microsc Microanal ; 17(2): 167-75, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21226989

RESUMO

The biomechanical response of tissues serves as a valuable marker in the prediction of disease and in understanding the related behavior of the body under various disease and age states. Alterations in the macroscopic biomechanical response of diseased tissues are well documented; however, a thorough understanding of the microstructural events that lead to these changes is poorly understood. In this article we introduce a novel microbiaxial optomechanical device that allows two-photon imaging techniques to be coupled with macromechanical stimulation in hydrated planar tissue specimens. This allows that the mechanical response of the microstructure can be quantified and related to the macroscopic response of the same tissue sample. This occurs without the need to fix tissue in strain states that could introduce a change in the microstructural configuration. We demonstrate the passive realignment of fibrous proteins under various types of loading, which demonstrates the ability of tissue microstructure to reinforce itself in periods of high stress. In addition, the collagen and elastin response of tissue during viscoelastic behavior is reported showing interstitial fluid movement and fiber realignment potentially responsible for the temporal behavior. We also demonstrate that nonhomogeneities in fiber strain exist over biaxial regions of assumed homogeneity.


Assuntos
Vasos Coronários/química , Microscopia/métodos , Animais , Fenômenos Biomecânicos , Colágeno/química , Elasticidade , Elastina/química , Microscopia/instrumentação , Modelos Biológicos , Suínos
20.
ScientificWorldJournal ; 11: 1509-24, 2011 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-21805020

RESUMO

Recent studies have suggested an important role for periostin and transforming growth factor beta (TGF beta) and bone morphogenetic protein (BMP) ligands in heart valve formation and valvular heart diseases. The function of these molecules in cardiovascular development has previously been individually reviewed, but their association has not been thoroughly examined. Here, we summarize the current understanding of the association between periostin and TGF beta and BMP ligands, and discuss the implications of this association in the context of the role of these molecules in heart valve development and valvular homeostasis. Information about hierarchal connections between periostin and TGF beta and BMP ligands in valvulogenesis will increase our understanding of the pathogenesis, progression, and medical treatment of human valve diseases.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Moléculas de Adesão Celular/fisiologia , Doenças das Valvas Cardíacas/embriologia , Valvas Cardíacas/embriologia , Fator de Crescimento Transformador beta/fisiologia , Proteínas Morfogenéticas Ósseas/metabolismo , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Doenças das Valvas Cardíacas/patologia , Valvas Cardíacas/anatomia & histologia , Valvas Cardíacas/metabolismo , Humanos , Modelos Biológicos , Organogênese , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA