Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Physiol ; 236(12): 8160-8170, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34170016

RESUMO

Epidermal growth factor (EGF) receptors (ErbB1-ErbB4) promote cardiac development and growth, although the specific EGF ligands and receptor isoforms involved in growth/repair versus pathology remain undefined. We challenged ventricular cardiomyocytes with EGF-like ligands and observed that selective activation of ErbB4 (the receptor for neuregulin 1 [NRG1]), but not ErbB1 (the receptor for EGF, EGFR), stimulated hypertrophy. This lack of direct ErbB1-mediated hypertrophy occurred despite robust activation of extracellular-regulated kinase 1/2 (ERK) and protein kinase B. Hypertrophic responses to NRG1 were unaffected by the tyrosine kinase inhibitor (AG1478) at concentrations that are selective for ErbB1 over ErbB4. NRG1-induced cardiomyocyte enlargement was suppressed by small interfering RNA (siRNA) knockdown of ErbB4 and ErbB2, whereas ERK phosphorylation was only suppressed by ErbB4 siRNA. Four ErbB4 isoforms exist (JM-a/JM-b and CYT-1/CYT-2), generated by alternative splicing, and their expression declines postnatally and following cardiac hypertrophy. Silencing of all four isoforms in cardiomyocytes, using an ErbB4 siRNA, abrogated NRG1-induced hypertrophic promoter/reporter activity, which was rescued by coexpression of knockdown-resistant versions of the ErbB4 isoforms. Thus, ErbB4 confers cardiomyocyte hypertrophy to NRG1, and all four ErbB4 isoforms possess the capacity to mediate this effect.


Assuntos
Hipertrofia/metabolismo , Miócitos Cardíacos/metabolismo , Isoformas de Proteínas/metabolismo , Receptor ErbB-4/metabolismo , Processamento Alternativo/genética , Animais , Proliferação de Células/fisiologia , Humanos , Fosforilação/fisiologia , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-4/genética , Transdução de Sinais/fisiologia
2.
J Clin Endocrinol Metab ; 106(4): 1163-1182, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33367756

RESUMO

CONTEXT: Pathogenic germline MAX variants are associated with pheochromocytoma and paraganglioma (PPGL), pituitary neuroendocrine tumors and, possibly, other endocrine and nonendocrine tumors. OBJECTIVE: To report 2 families with germline MAX variants, pheochromocytomas (PCs) and multiple other tumors. METHODS: Clinical, genetic, immunohistochemical, and functional studies at University hospitals in Australia on 2 families with germline MAX variants undergoing usual clinical care. The main outcome measures were phenotyping; germline and tumor sequencing; immunohistochemistry of PC and other tumors; functional studies of MAX variants. RESULTS: Family A has multiple individuals with PC (including bilateral and metastatic disease) and 2 children (to date, without PC) with neuroendocrine tumors (paravertebral ganglioneuroma and abdominal neuroblastoma, respectively). One individual has acromegaly; immunohistochemistry of PC tissue showed positive growth hormone-releasing hormone staining. Another individual with previously resected PCs has pituitary enlargement and elevated insulin-like growth factor (IGF-1). A germline MAX variant (c.200C>A, p.Ala67Asp) was identified in all individuals with PC and both children, with loss of heterozygosity in PC tissue. Immunohistochemistry showed loss of MAX staining in PCs and other neural crest tumors. In vitro studies confirmed the variant as loss of function. In Family B, the proband has bilateral and metastatic PC, prolactin-producing pituitary tumor, multigland parathyroid adenomas, chondrosarcoma, and multifocal pulmonary adenocarcinomas. A truncating germline MAX variant (c.22G>T, p.Glu8*) was identified. CONCLUSION: Germline MAX mutations are associated with PCs, ganglioneuromas, neuroblastomas, pituitary neuroendocrine tumors, and, possibly, parathyroid adenomas, as well as nonendocrine tumors of chondrosarcoma and lung adenocarcinoma, suggesting MAX is a novel multiple endocrine neoplasia gene.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Mutação em Linhagem Germinativa , Neoplasia Endócrina Múltipla/genética , Adolescente , Neoplasias das Glândulas Suprarrenais/diagnóstico , Neoplasias das Glândulas Suprarrenais/genética , Adulto , Idoso , Austrália , Pré-Escolar , Família , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Neoplasia Endócrina Múltipla/classificação , Neoplasia Endócrina Múltipla/diagnóstico , Neoplasias Primárias Múltiplas/diagnóstico , Neoplasias Primárias Múltiplas/genética , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/genética , Linhagem , Feocromocitoma/diagnóstico , Feocromocitoma/genética , Adulto Jovem
3.
J Transl Med ; 18(1): 431, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33183308

RESUMO

BACKGROUND: Clinical whole exome sequencing was introduced in an Australian centre in 2017, as an alternative to Sanger sequencing. We aimed to identify predictors of cancer physicians' somatic mutation test ordering behaviour. METHODS: A validated instrument assessed somatic mutation test ordering, genomic confidence, perceived utility of tumour molecular profiling, and percent of patients eligible for targeted therapy. A cash incentive was included in 189/244 questionnaires which were mailed to all Queensland cancer specialists in November 2018. RESULTS: 110 participated (response rate 45%); 54.7% oncologists, and the remainder were surgeons, haematologists and pulmonologists. Oncologists were more likely to respond (p = 0.008), and cash incentive improved the response rate (p < 0.001). 67/102 (65.7%) of physicians ordered ≥ 5 somatic mutation tests annually. Oncologists saw 86.75 unique patients monthly and ordered 2.33 somatic mutation tests (2.2%). An average of 51/110 (46.1%) reported having little/no genomic confidence. Logistic regression identified two significant predictors of somatic mutation test ordering: being an oncologist (OR 3.557, CI 1.338-9.456; p = 0.011) and having greater confidence in interpreting somatic results (OR 5.926, CI 2.230-15.74; p < 0.0001). CONCLUSIONS: Consistent with previous studies, the majority of cancer physicians ordered somatic mutation tests. However, the percentage of patients on whom tests were ordered was low. Almost half respondents reported low genomic confidence. Somatic mutation test ordering was higher amongst oncologists and those with increased confidence in interpreting somatic variants. It is unclear whether genomically confident individuals ordered more tests or whether ordering more tests increased genomic confidence. Educational interventions could improve confidence and enhance test ordering behaviour.


Assuntos
Testes Genéticos , Neoplasias , Austrália , Genômica , Humanos , Mutação , Neoplasias/diagnóstico , Neoplasias/genética , Médicos
4.
J Mol Histol ; 48(1): 53-61, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27803996

RESUMO

Endothelial cells form a critical component of the coronary vasculature, yet the factors regulating their development remain poorly defined. Here we reveal a novel role for the transmembrane protein CRIM1 in mediating cardiac endothelial cell development. In the absence of Crim1 in vivo, the coronary vasculature is malformed, the number of endothelial cells reduced, and the canonical BMP pathway dysregulated. Moreover, we reveal that CRIM1 can bind IGFs, and regulate IGF signalling within endothelial cells. Finally, loss of CRIM1 from human cardiac endothelial cells results in misregulation of endothelial genes, predicted by pathway analysis to be involved in an increased inflammatory response and cytolysis, reminiscent of endothelial cell dysfunction in cardiovascular disease pathogenesis. Collectively, these findings implicate CRIM1 in endothelial cell development and homeostasis in the coronary vasculature.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Vasos Coronários/citologia , Células Endoteliais/metabolismo , Homeostase , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Sobrevivência Celular/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Knockout , Mutação , Transdução de Sinais
6.
Histol Histopathol ; 31(10): 1049-57, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27044529

RESUMO

The regulation of growth factor localization, availability and activity is critical during embryogenesis to ensure appropriate organogenesis. This process is regulated through the coordinated expression of growth factors and their cognate receptors, as well as via proteins that can bind, sequester or localize growth factors to distinct locations. One such protein is the transmembrane protein Crim1. This protein has been shown to be expressed broadly within the developing embryo, and to regulate organogenesis within the eye, kidney and placenta. Mechanistically, Crim1 has been revealed to mediate organogenesis via its interaction with growth factors including TGFßs, BMPs, VEGFs and PDFGs. More recently, Crim1 has been shown to influence cardiac development, providing further insights into the function of this protein. This review will provide an overview of the role of Crim1 in organogenesis, largely focusing on how this protein regulates growth factor signaling in the nascent heart. Moreover, we will address the challenges ahead relating to further elucidating how Crim1 functions during development.


Assuntos
Proteínas de Membrana/metabolismo , Organogênese/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas , Humanos
7.
Sci Rep ; 6: 19832, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26821812

RESUMO

The epicardium has a critical role during embryonic development, contributing epicardium-derived lineages to the heart, as well as providing regulatory and trophic signals necessary for myocardial development. Crim1 is a unique trans-membrane protein expressed by epicardial and epicardially-derived cells but its role in cardiogenesis is unknown. Using knockout mouse models, we observe that loss of Crim1 leads to congenital heart defects including epicardial defects and hypoplastic ventricular compact myocardium. Epicardium-restricted deletion of Crim1 results in increased epithelial-to-mesenchymal transition and invasion of the myocardium in vivo, and an increased migration of primary epicardial cells. Furthermore, Crim1 appears to be necessary for the proliferation of epicardium-derived cells (EPDCs) and for their subsequent differentiation into cardiac fibroblasts. It is also required for normal levels of cardiomyocyte proliferation and apoptosis, consistent with a role in regulating epicardium-derived trophic factors that act on the myocardium. Mechanistically, Crim1 may also modulate key developmentally expressed growth factors such as TGFßs, as changes in the downstream effectors phospho-SMAD2 and phospho-ERK1/2 are observed in the absence of Crim1. Collectively, our data demonstrates that Crim1 is essential for cell-autonomous and paracrine aspects of heart development.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/genética , Cardiopatias Congênitas/genética , Coração/crescimento & desenvolvimento , Organogênese/genética , Animais , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética , Transição Epitelial-Mesenquimal/genética , Cardiopatias Congênitas/patologia , Humanos , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Comunicação Parácrina/genética , Pericárdio/embriologia , Proteína Smad2/genética , Fator de Crescimento Transformador beta1/genética
8.
J Pathol ; 229(5): 685-96, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23224993

RESUMO

Crim1 is a transmembrane protein that regulates the bioavailability of growth factors such as VEGFA. Crim1(KST264)(/)(KST264) hypomorphic mice develop renal disease characterized by glomerular cysts and loss of endothelial integrity, progressing to peritubular and pericystic fibrosis. Peritubular capillary endothelial cells display morphological changes as well as detachment from the basement membrane. In this study, gene expression profiling of CD31(+) endothelial cells isolated from Crim1(KST264)(/)(KST264) kidneys showed up-regulation of transcripts associated with fibrosis (Col3a1, Loxl1), endothelial dysfunction (Abp1, Dcn, Lcn2), biomarkers of renal damage (Lcn2, Havcr1/Kim1) as well as evidence for a TGFß1/TNF-associated inflammatory process. To determine whether the aberrant endothelium may in part contribute to the fibrogenic process, Tie2Cre-DsRed lineage tracing was undertaken in Crim1(KST264/KST264) mice. Approximately 31% of de novo αSMA(+) myofibroblasts detected within the tubulointerstitium were Tie2(+) DsRed(+) . However, 5.3% were F4/80(+) DsRed(+) , indicating a small population of myofibroblasts of monocytic rather than endothelial origin. In contrast, only 12% of myofibroblasts located around glomerular cysts were Tie2(+) DsRed(+) , with 7.7% being monocyte-derived (F4/80(+) DsRed(+) ). Collectively, this model supports the involvement of endothelial cells/monocytes in fibrosis within the tubulointerstitium, but also the heterogeneity of the fibrotic process even within distinct regions of the same kidney.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/genética , Linhagem da Célula , Células Endoteliais/patologia , Nefropatias/patologia , Rim/patologia , Monócitos/patologia , Mutação , Miofibroblastos/patologia , Animais , Biomarcadores/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem da Célula/genética , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal , Fibrose , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Marcadores Genéticos , Genótipo , Integrases/genética , Rim/irrigação sanguínea , Rim/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Miofibroblastos/metabolismo , Fenótipo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptor TIE-2
9.
Genesis ; 50(9): 711-6, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22511315

RESUMO

Crim1 is a developmentally expressed, transmembrane protein essential for normal embryonic development. We generated mice engineered to contain a Crim1 conditional null allele by flanking exons three and four of Crim1 with unidirectional LoxP sites. After crossing Crim1+/FLOX mice with a CMV-Cre line, a Crim1+/Δflox colony was established after germline transmission of the deleted allele. We then analyzed genomic DNA, mRNA transcripts, and protein expression from Crim1Δflox/Δflox null mice to confirm the nature of the genomic lesion. Crim1Δflox/Δflox mice displayed phenotypes similar to those previously described for a Crim1 gene-trap mutant, Crim1KST264/KST264, including perinatal lethality, digit syndactyly, eye, and kidney abnormalities, with varying penetrance and severity. The production of a conditional mutant allele represents a valuable resource for the study of the tissue-specific roles for Crim1, and for understanding the pleimorphic phenotypes associated with Crim1 mutation.


Assuntos
Anormalidades Múltiplas/embriologia , Receptores de Proteínas Morfogenéticas Ósseas/genética , Desenvolvimento Embrionário/genética , Engenharia Genética/métodos , Anormalidades Múltiplas/genética , Alelos , Animais , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Quimera , Cruzamentos Genéticos , Éxons , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Integrases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Especificidade de Órgãos , Organogênese/genética , Fenótipo , Gravidez , Recombinação Genética
10.
Kidney Int ; 76(11): 1161-71, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19776720

RESUMO

Crim1 is a cell-surface, transmembrane protein that binds to a variety of cystine knot-containing growth factors, including vascular endothelial growth factor A. In the developing renal glomerulus, Crim1 acts to tether vascular endothelial growth factor A to the podocyte cell surface, thus regulating its release to glomerular endothelial cells. The hypomorphic transgenic mouse (Crim1(KST264/KST264)) has glomerular cysts and severe glomerular vascular defects because of the lack of functional Crim1 in the glomerulus. Adult transgenic mice have a reduced glomerular filtration rate and glomerular capillary defects. We now show that, in these adult transgenic mice, renal vascular defects are not confined to the glomerulus but also extend to the peritubular microvasculature, as live imaging revealed leakiness of both glomerular and peritubular capillaries. An ultrastructural analysis of the microvasculature showed an abnormal endothelium and collagen deposition between the endothelium and the tubular basement membrane, present even in juvenile mice. Overt renal disease, including fibrosis and renin recruitment, was not evident until adulthood. Our study suggests that Crim1 is involved in endothelial maintenance and integrity and its loss contributes to a primary defect in the extraglomerular vasculature.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/genética , Rim/irrigação sanguínea , Microvasos , Animais , Endotélio Vascular , Camundongos , Camundongos Transgênicos
11.
Dev Biol ; 328(1): 148-59, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19389363

RESUMO

Critical steps in coronary vascular formation include the epithelial-mesenchyme transition (EMT) that epicardial cells undergo to become sub-epicardial; the invasion of the myocardium; and the differentiation of coronary lineages. However, the factors controlling these processes are not completely understood. Epicardial and coronary vascular precursors migrate to the avascular heart tube during embryogenesis via the proepicardium (PE). Here, we show that in the quail embryo fibroblast growth factor receptor (FGFR)-1 is expressed in a spatially and temporally restricted manner in the PE and epicardium-derived cells, including vascular endothelial precursors, and is up-regulated in epicardial cells after EMT. We used replication-defective retroviral vectors to over-express or knock-down FGFR-1 in the PE. FGFR-1 over-expression resulted in increased epicardial EMT. Knock-down of FGFR-1, however, did not inhibit epicardial EMT but greatly compromised the ability of PE progeny to invade the myocardium. The latter could, however, contribute to endothelia and smooth muscle of sub-epicardial vessels. Correct FGFR-1 levels were also important for correct coronary lineage differentiation with, at E12, an increase in the proportion of endothelial cells amongst FGFR-1 over-expressing PE progeny and a decrease in the proportion of smooth muscle cells in antisense FGFR-1 virus-infected PE progeny. Finally, in a heart explant system, constitutive activation of FGFR-1 signaling in epicardial cells resulted in increased delamination from the epicardium, invasion of the sub-epicardium, and invasion of the myocardium. These data reveal novel roles for FGFR-1 signaling in epicardial biology and coronary vascular lineage differentiation, and point to potential new therapeutic avenues.


Assuntos
Movimento Celular , Vasos Coronários/fisiologia , Endotélio Vascular/citologia , Pericárdio/citologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Embrião não Mamífero , Endotélio Vascular/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos de Músculo Liso/metabolismo , Pericárdio/embriologia , Pericárdio/metabolismo , Codorniz
12.
Dev Dyn ; 236(2): 502-11, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17106887

RESUMO

Crim1 is a transmembrane protein, containing six vWF-C type cysteine-rich repeats, that tethers growth factors to the cell surface. A mouse line, KST264, generated in a LacZ insertion mutagenesis gene-trap screen, was examined to elucidate Crim1 function in development. We showed that Crim1(KST264/KST264) mice were not null for Crim1 due to the production of a shortened protein isoform. These mice are likely to represent an effective hypomorph or a dominant-negative for Crim1. Transgene expression recapitulated known Crim1 expression in lens, brain, and limb, but also revealed expression in the smooth muscle cells of the developing heart and renal vasculature, developing cartilage, mature ovary and detrusor of the bladder. Transgene expression was also observed in glomerular epithelial cells, podocytes, mesangial cells, and urothelium in the kidney. Crim1(KST264/KST264) mice displayed perinatal lethality, syndactyly, eye, and kidney abnormalities. The severe and complex phenotype observed in Crim1(KST264/KST264) mice highlights the importance of Crim1 in numerous aspects of organogenesis.


Assuntos
Anormalidades Múltiplas/embriologia , Receptores de Proteínas Morfogenéticas Ósseas/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Membrana/genética , Organogênese/genética , Fenótipo , Anormalidades Múltiplas/genética , Animais , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Primers do DNA , Técnicas Histológicas , Immunoblotting , Proteínas de Membrana/metabolismo , Camundongos , Mutagênese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transgenes/genética
13.
Dev Biol ; 279(2): 378-90, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15733666

RESUMO

The formation of the coronary vessel system is vital for heart development, an essential step of which is the establishment of a capillary plexus that displays a density gradient across the myocardial wall, being higher on the epicardial than the endocardial side. This gradient in capillary plexus formation develops concurrently with transmural gradients of myocardium-derived growth factors, including FGFs. To test the role of the FGF expression gradient in patterning the nascent capillary plexus, an ectopic FGF-over-expressing site was created in the ventricular myocardial wall in the quail embryo via retroviral infection from E2-2.5, thus abolishing the transmural gradient of FGFs. In FGF virus-infected regions of the ventricular myocardium, the capillary density across the transmural axis shifted away from that in control hearts at E7. This FGF-induced change in vessel patterning was more profound at E12, with the middle zone becoming the most vascularized. An up-regulation of FGFR-1 and VEGFR-2 in epicardial and subepicardial cells adjacent to FGF virus-infected myocardium was also detected, indicating a paracrine effect on induction of vascular signaling components in coronary precursors. These results suggest that correct transmural patterning of coronary vessels requires the correct transmural expression of FGF and, therefore, FGF may act as a template for coronary vessel patterning.


Assuntos
Padronização Corporal , Capilares/embriologia , Circulação Coronária/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Animais , Capilares/anatomia & histologia , Embrião não Mamífero/anatomia & histologia , Embrião não Mamífero/fisiologia , Fatores de Crescimento de Fibroblastos/genética , Coração/anatomia & histologia , Morfogênese/fisiologia , Comunicação Parácrina , Codorniz/anatomia & histologia , Codorniz/embriologia , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
Development ; 131(3): 581-92, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14711873

RESUMO

Impulse-conducting Purkinje fibers differentiate from myocytes during embryogenesis. The conversion of contractile myocytes into conduction cells is induced by the stretch/pressure-induced factor, endothelin (ET). Active ET is produced via proteolytic processing from its precursor by ET-converting enzyme 1 (ECE1) and triggers signaling by binding to its receptors. In the embryonic chick heart, ET receptors are expressed by all myocytes, but ECE1 is predominantly expressed in endothelial cells of coronary arteries and endocardium along which Purkinje fiber recruitment from myocytes takes place. Furthermore, co-expression of exogenous ECE1 and ET-precursor in the embryonic heart is sufficient to ectopically convert cardiomyocytes into Purkinje fibers. Thus, localized expression of ECE1 defines the site of Purkinje fiber recruitment in embryonic myocardium. However, it is not known how ECE1 expression is regulated in the embryonic heart. The unique expression pattern of ECE1 in the embryonic heart suggests that blood flow-induced stress/stretch may play a role in patterning ECE1 expression and subsequent induction of Purkinje fiber differentiation. We show that gadolinium, an antagonist for stretch-activated cation channels, downregulates the expression of ECE1 and a conduction cell marker, Cx40, in ventricular chambers, concurrently with delayed maturation of a ventricular conduction pathway. Conversely, pressure-overload in the ventricle by conotruncal banding results in a significant expansion of endocardial ECE1 expression and Cx40-positive putative Purkinje fibers. Coincident with this, an excitation pattern typical of the mature heart is precociously established. These in vivo data suggest that biomechanical forces acting on, and created by, the cardiovascular system during embryogenesis play a crucial role in Purkinje fiber induction and patterning.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Diferenciação Celular/fisiologia , Coração/embriologia , Ramos Subendocárdicos/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Padronização Corporal/fisiologia , Embrião de Galinha , Regulação para Baixo/fisiologia , Enzimas Conversoras de Endotelina , Gadolínio/metabolismo , Coração/fisiologia , Metaloendopeptidases , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Dev Dyn ; 228(2): 161-72, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14517988

RESUMO

Proper heart development requires patterning across the myocardial wall. Early myocardial patterning is characterized by a transmural subdivision of the myocardium into an outer, highly mitotic, compact zone and an inner, trabecular zone with lower mitotic activity. We have shown previously that fibroblast growth factor receptor (FGFR) -mediated signaling is central to myocyte proliferation in the developing heart. Consistent with this, FGFR-1 and FGF2 are more highly expressed in myocytes of the compact zone. However, the mechanism that regulates the transmural pattern of myocyte proliferation and expression of these mitogenic factors is unknown. The present study examined whether this transmural patterning occurs in a myocardium-autonomous manner or by signals from the epicardium. Microsurgical inhibition of epicardium formation in the embryonic chick gives rise to a decrease in myocyte proliferation, accounting for a thinner compact myocardium. We show that the transmural pattern of myocyte mitotic activity is maintained in these hearts. Consistent with this, the expression patterns of FGF1, FGF2, and FGFR-1 across the myocardium persist in the absence of the epicardium. However, FGF2 and FGFR-1 mRNA levels are reduced in proportion to the depletion of epicardium. The results suggest that epicardium-derived signals are essential for maintenance of the correct amount of myocyte proliferation in the compact myocardium, by means of levels of mitogen expression in the myocardium. However, initiation and maintenance of transmural patterning of the myocardium occurs largely independently of the epicardium.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Coração/embriologia , Miócitos Cardíacos/metabolismo , Pericárdio/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Apoptose , Divisão Celular , Embrião de Galinha , Fator 1 de Crescimento de Fibroblastos/metabolismo , Imuno-Histoquímica , Ligantes , Microcirurgia , Mitógenos/metabolismo , Miócitos Cardíacos/citologia , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
16.
Novartis Found Symp ; 250: 142-53; discussion 153-6, 276-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12956328

RESUMO

Impulse-conducting Purkinje cells differentiate from myocytes during embryogenesis. In the embryonic chicken heart, this conversion of contractile myocytes into conduction cells occurs subendocardially and periarterially. The unique sites of Purkinje fibre differentiation suggest that a shear stress-induced paracrine signal from the endocardium and arterial beds may induce adjacent myocytes to differentiate into conduction cells. Consistent with this model, Purkinje fibre marker genes can be induced in cultured embryonic myocytes by endothelin (ET), an endothelial cell-derived signalling peptide. This inductive response is, however, gradually lost from myocytes as embryos develop, and mature myocytes express only genes characteristic of hypertrophy in response to ET. In vivo, active ET is produced, through proteolytic processing, from its precursor by ET-converting enzyme 1 (ECE1) and triggers signalling by binding to its receptors, ETA and ETB. In the embryonic heart, the expression of these ET signalling components changes dynamically, defining the site and timing of Purkinje fibre differentiation within the ventricular myocardium during chick embryogenesis.


Assuntos
Diferenciação Celular/fisiologia , Indução Embrionária , Coração/crescimento & desenvolvimento , Ramos Subendocárdicos/embriologia , Animais , Ácido Aspártico Endopeptidases/metabolismo , Enzimas Conversoras de Endotelina , Endotélio/citologia , Endotélio/metabolismo , Coração/anatomia & histologia , Coração/fisiologia , Metaloendopeptidases , Morfogênese , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ramos Subendocárdicos/anatomia & histologia , Ramos Subendocárdicos/fisiologia , Receptores de Endotelina/metabolismo
17.
Int J Dev Biol ; 46(6): 765-75, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12382942

RESUMO

The cardiac conduction system (CCS) is the component of the heart that initiates and maintains a rhythmic heartbeat. As the embryonic heart forms, the CCS must continue to develop and mature in a coordinated manner to ensure that proper pace making potential and distribution of action potential is maintained at all stages. This requires not only the formation of distinct and disparate components of the CCS, but the integration of these components into a functioning whole as the heart matures. Though research in this area of development may have lagged behind other areas of heart development, in recent years there has been much progress in understanding the ontogeny of the CCS and the developmental cues that drive its formation. This is largely due to studies on the avian heart as well as the use of molecular biology approaches. This review gives a perspective on advances in understanding the development of the vertebrate CCS, and reports new data illuminating the mechanism of conduction cell determination and maintenance in the mammalian heart. As much of our knowledge about the development of the CCS has been derived from the chick embryo, one important area facing the field is the relationship and similarities between the structure and development of avian and mammalian conduction systems. Specifically, the morphology of the distal elements of the mammalian CCS and the manner in which its components are recruited from working cardiomyocytes are areas of research that will, hopefully, receive more attention in the near future. A more general and outstanding question is how the disparate components of all vertebrate conduction systems integrate into a functional entity during embryogenesis. There is mounting evidence linking the patterning and formation of the CCS to instructive cues derived from the cardiac vasculature and, more specifically, to hemodynamic-responsive factors produced by cardiac endothelia. This highlights the need for a greater understanding of the biophysical forces acting on, and created by, the cardiovascular system during embryonic development. A better understanding of these processes will be necessary if therapeutics are to be developed that allow the regeneration of damaged cardiac tissues or the construction of biologically engineered heart tissues.


Assuntos
Indução Embrionária/fisiologia , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/fisiologia , Coração/embriologia , Coração/fisiologia , Animais , Nó Atrioventricular/embriologia , Nó Atrioventricular/fisiologia , Relógios Biológicos/fisiologia , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/fisiologia , Embrião de Galinha , Endotélio/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Miócitos Cardíacos/fisiologia , Ramos Subendocárdicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...