Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
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.

2.
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
3.
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
4.
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.

5.
J Biomech ; 125: 110543, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34174532

RESUMO

Transforming growth factor-beta (TGFß-1, -2, -3) ligands act through a common receptor complex yet each is expressed in a unique and overlapping fashion throughout development. TGFß plays a role in extra-cellular matrix composition with mutations to genes encoding TGFß and TGFß signaling molecules contributing to diverse and deadly thoracic aortopathies common in Loeys-Dietz syndrome (LDS). In this investigation, we studied the TGFß ligand-specific mechanical phenotype of ascending thoracic aortas (ATA) taken from 4-to-6 months-old Tgfb1+/-, Tgfb2+/-, and Tgfb3+/- mice, their wild-type (WT) controls, and an elastase infusion model representative of severe elastolysis. Heterozygous mice were studied at an age without dilation to elucidate potential pre-aortopathic mechanical cues. Our findings indicate that ATAs from Tgfb2+/- mice demonstrated significant wall thickening, a corresponding decrease in biaxial stress, decreased biaxial stiffness, and a decrease in stored energy. These results were unlike the pathological elastase model where decreases in biaxial stretch were found along with increases in diameter, biaxial stress, and biaxial stiffness. ATAs from Tgfb1+/- and Tgfb3+/-, on the other hand, had few mechanical differences when compared to wild-type controls. Although aortopathy generally occurs later in development, our findings reveal that in 4-to-6 month-old animals, only Tgfb2+/- mice demonstrate a significant phenotype that fails to model ubiquitous elastolysis.


Assuntos
Síndrome de Loeys-Dietz , Elastase Pancreática , Animais , Aorta , Camundongos , Mutação , Fator de Crescimento Transformador beta2/genética
6.
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
7.
J Cardiovasc Dev Dis ; 8(3)2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33801433

RESUMO

Among the three transforming growth factor beta (TGFß) ligands, TGFß2 is essential for heart development and is produced by multiple cell types, including myocardium. Heterozygous mutations in TGFB2 in patients of connective tissue disorders result in congenital heart defects and adult valve malformations, including mitral valve prolapse (MVP) with or without regurgitation. Tgfb2 germline knockout fetuses exhibit multiple cardiac defects but the role of myocardial-TGFß2 in heart development is yet to be elucidated. Here, myocardial Tgfb2 conditional knockout (CKO) embryos were generated by crossing Tgfb2flox mice with Tgfb2+/-; cTntCre mice. Tgfb2flox/- embryos were normal, viable. Cell fate mapping was done using dual-fluorescent mT/mG+/- mice. Cre-mediated Tgfb2 deletion was assessed by genomic PCR. RNAscope in situ hybridization was used to detect the loss of myocardial Tgfb2 expression. Histological, morphometric, immunohistochemical, and in situ hybridization analyses of CKOs and littermate controls at different stages of heart development (E12.5-E18.5) were used to determine the role of myocardium-derived TGFß2 in atrioventricular (AV) cushion remodeling and myocardial development. CKOs exhibit a thin ventricular myocardium, AV cushion remodeling defects and developed incomplete AV septation defects. The loss of myocardial Tgfb2 resulted in impaired cushion maturation and dysregulated cell death. Phosphorylated SMAD2, a surrogate for TGFß signaling, was "paradoxically" increased in both AV cushion mesenchyme and ventricular myocardium in the CKOs. Our results indicate that TGFß2 produced by cardiomyocytes acting as cells autonomously on myocardium and via paracrine signaling on AV cushions are required for heart development.

8.
Nat Cancer ; 2(3): 327-339, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-34993493

RESUMO

In the bone marrow (BM) microenvironment, where breast cancer (BC) disseminated tumour cells (DTCs) can remain dormant for decades, NG2+/Nestin+ mesenchymal stem cells (MSCs) promote hematopoietic stem cell quiescence. Here, we reveal that periarteriolar BM-resident NG2+/Nestin+ MSCs can also instruct BC DTCs to enter dormancy. NG2+/Nestin+ MSCs produce TGFß2 and BMP7 and activate a quiescence pathway dependent on TGFBRIII and BMPRII, which via p38-kinase result in p27 induction. Genetic depletion of MSCs or conditional knock-out of TGFß2 in MSCs using an NG2-CreER driver led to bone metastatic outgrowth of otherwise dormant p27+/Ki67- DTCs. Also ER+ BC patients without systemic recurrence displayed higher frequency of TGFß2 and BMP7 detection in the BM. Our results provide a direct proof that HSC dormancy niches control BC DTC dormancy and suggest that aging or extrinsic factors that affect the NG2+/Nestin+ MSC niche homeostasis may result in a break from dormancy and BC bone relapse.


Assuntos
Neoplasias da Mama , Células-Tronco Mesenquimais , Medula Óssea/metabolismo , Neoplasias da Mama/genética , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Recidiva Local de Neoplasia/metabolismo , Nestina/metabolismo , Microambiente Tumoral
9.
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
10.
J Cardiovasc Dev Dis ; 7(2)2020 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-32456345

RESUMO

Transforming growth factor beta3 (TGFB3) gene mutations in patients of arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD1) and Loeys-Dietz syndrome-5 (LDS5)/Rienhoff syndrome are associated with cardiomyopathy, cardiac arrhythmia, cardiac fibrosis, cleft palate, aortic aneurysms, and valvular heart disease. Although the developing heart of embryos express Tgfb3, its overarching role remains unclear in cardiovascular development and disease. We used histological, immunohistochemical, and molecular analyses of Tgfb3-/- fetuses and compared them to wildtype littermate controls. The cardiovascular phenotypes were diverse with approximately two thirds of the Tgfb3-/- fetuses having one or more cardiovascular malformations, including abnormal ventricular myocardium (particularly of the right ventricle), outflow tract septal and alignment defects, abnormal aortic and pulmonary trunk walls, and thickening of semilunar and/or atrioventricular valves. Ventricular septal defects (VSD) including the perimembranous VSDs were observed in Tgfb3-/- fetuses with myocardial defects often accompanied by the muscular type VSD. In vitro studies using TGFß3-deficient fibroblasts in 3-D collagen lattice formation assays indicated that TGFß3 was required for collagen matrix reorganization. Biochemical studies indicated the 'paradoxically' increased activation of canonical (SMAD-dependent) and noncanonical (MAP kinase-dependent) pathways. TGFß3 is required for cardiovascular development to maintain a balance of canonical and noncanonical TGFß signaling pathways.

11.
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
12.
Nat Metab ; 1(2): 291-303, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-31032475

RESUMO

Exercise improves health and well-being across diverse organ systems, and elucidating mechanisms underlying the beneficial effects of exercise can lead to new therapies. Here, we show that transforming growth factor-ß2 (TGF-ß2) is secreted from adipose tissue in response to exercise and improves glucose tolerance in mice. We identify TGF-ß2 as an exercise-induced adipokine in a gene expression analysis of human subcutaneous adipose tissue biopsies after exercise training. In mice, exercise training increases TGF-ß2 in scWAT, serum, and its secretion from fat explants. Transplanting scWAT from exercise-trained wild type mice, but not from adipose tissue-specific Tgfb2-/- mice, into sedentary mice improves glucose tolerance. TGF-ß2 treatment reverses the detrimental metabolic effects of high fat feeding in mice. Lactate, a metabolite released from muscle during exercise, stimulates TGF-ß2 expression in human adipocytes. Administration of the lactate-lowering agent dichloroacetate during exercise training in mice decreases circulating TGF-ß2 levels and reduces exercise-stimulated improvements in glucose tolerance. Thus, exercise training improves systemic metabolism through inter-organ communication with fat via a lactate-TGF-ß2-signaling cycle.


Assuntos
Adipocinas/metabolismo , Ácidos Graxos/metabolismo , Glucose/metabolismo , Condicionamento Físico Animal , Fator de Crescimento Transformador beta2/metabolismo , Tecido Adiposo/metabolismo , Animais , Camundongos
13.
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
14.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-780685

RESUMO

Aims@#Dendrobiums are majorly affected by Fusarium proliferatum and F. oxysporum. The aim of this research was to utilise the mycotoxin, fusaric acid (FA) on Dendrobium hybrid to produce cultivars that are resistant towards these fungi. @*Methodology and results@#FA of concentrations 0.05, 0.10, 0.15 and 0.20 mM were transferred to sterilised half-strength Murashige and Skoog (MS) medium and inoculated with four weeks old thin cell layer (TCL) of protocorm-like bodies (PLBs) for eight weeks. It was deduced that PLBs treated with 0.10 mM of FA resulted in highest survival and shoot regeneration rate but the survival and regeneration rate began to decline as the concentrations of FA were increased. Histology and scanning electron microscopy (SEM) observation showed prominent cell damage and stomatal closure in PLBs treated with FA. Direct amplification of minisatellite DNA (DAMD) markers showed polymorphism in the FA treated PLBs compared to the control PLBs. In the leaf bridge bioassay, plantlets treated with 0.05 mM of FA showed most resistance towards both fungal species. @*Conclusion, significance and impact of study@#Therefore, this research is a preliminary screening study where the optimum concentration of FA was selected based on the reaction of treated TCL of PLBs towards these mutagens.

16.
Clin Perinatol ; 43(1): 39-53, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26876120

RESUMO

Cardiovascular malformations (CVMs) are the most common birth defect, occurring in 1% to 5% of all live births. Genetic, epigenetic, and environmental factors all influence the development of CVMs, and an improved understanding of the causation of CVMs is a prerequisite for prevention. Cardiac development is a complex, multistep process of morphogenesis that is under genetic regulation. Although the genetic contribution to CVMs is well recognized, the genetic causes of human CVMs are still identified infrequently. This article discusses the key genetic concepts characterizing human CVMs, their developmental basis, and the critical developmental and genetic concepts underlying their pathogenesis.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/genética , Anormalidades Cardiovasculares/embriologia , Anormalidades Cardiovasculares/genética , Dosagem de Genes , Genótipo , Cardiopatias Congênitas/embriologia , Humanos , Fenótipo
17.
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
18.
Front Physiol ; 5: 258, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25071603

RESUMO

Tgfb3 is strongly and specifically expressed in the epithelial tips of pre-fusion palatal shelves where it plays a critical non-redundant role in palatal fusion in both medial edge epithelial (MEE) cells and in a thin layer of flattened peridermal cells that covers the MEE. It is not known how Tgfb3 expression is regulated in these specific cell types. Using comparative genomics and transgenic reporter assays, we have identified cis-regulatory elements that could control Tgfb3 expression during palatogenesis. Our results show that a 61-kb genomic fragment encompassing the Tgfb3 gene drives remarkably specific reporter expression in the MEE and adjacent periderm. Within this fragment, we identified two small, non-coding, evolutionarily conserved regions in intron 2 of the neighboring Ift43 gene, and a larger region in the intervening sequence between the Ift43 and Tgfb3 genes, each of which could target reporter activity to the tips of pre-fusion/fusing palatal shelves. Identification of the cis-regulatory sequences controlling spatio-temporal Tgfb3 expression in palatal shelves is a key step toward understanding upstream regulation of Tgfb3 expression during palatogenesis and should enable the development of improved tools to investigate palatal epithelial fusion.

19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...