Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cell Physiol ; 236(12): 8160-8170, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34170016

RESUMEN

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.


Asunto(s)
Hipertrofia/metabolismo , Miocitos Cardíacos/metabolismo , Isoformas de Proteínas/metabolismo , Receptor ErbB-4/metabolismo , Empalme Alternativo/genética , Animales , Proliferación Celular/fisiología , Humanos , Fosforilación/fisiología , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-4/genética , Transducción de Señal/fisiología
2.
J Clin Endocrinol Metab ; 106(4): 1163-1182, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33367756

RESUMEN

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.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Mutación de Línea Germinal , Neoplasia Endocrina Múltiple/genética , Adolescente , Neoplasias de las Glándulas Suprarrenales/diagnóstico , Neoplasias de las Glándulas Suprarrenales/genética , Adulto , Anciano , Australia , Preescolar , Familia , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple/clasificación , Neoplasia Endocrina Múltiple/diagnóstico , Neoplasias Primarias Múltiples/diagnóstico , Neoplasias Primarias Múltiples/genética , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/genética , Linaje , Feocromocitoma/diagnóstico , Feocromocitoma/genética , Adulto Joven
3.
J Transl Med ; 18(1): 431, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33183308

RESUMEN

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.


Asunto(s)
Pruebas Genéticas , Neoplasias , Australia , Genómica , Humanos , Mutación , Neoplasias/diagnóstico , Neoplasias/genética , Médicos
4.
J Neuroimmunol ; 323: 109-114, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30196823

RESUMEN

Guillain-Barré syndrome (GBS) is considered to have an immune-mediated basis, but the genetic contribution to GBS is unclear. We conducted a GWAS involving 215 GBS patients and 1,105 healthy controls. No significant associations of individual SNPs or imputed HLA types were observed. We performed a genome-wide complex trait analysis for evaluation of the heritability of GBS, and found that common SNPs contribute up to 25% of susceptibility to the disease. Genetic risk score analysis showed no evidence of overlap in genetic susceptibility factors of GBS and multiple sclerosis. Given the unexplained heritability of the trait further larger GWAS are indicated.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Síndrome de Guillain-Barré/diagnóstico , Síndrome de Guillain-Barré/genética , Polimorfismo de Nucleótido Simple/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Síndrome de Guillain-Barré/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Exp Eye Res ; 170: 58-66, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29458060

RESUMEN

The development and growth of the vertebrate ocular lens is dependent on the regulated proliferation of an anterior monolayer of epithelial cells, and their subsequent differentiation into elongate fiber cells. The growth factor rich ocular media that bathes the lens mediates these cellular processes, and their respective intracellular signaling pathways are in turn regulated to ensure that the proper lens architecture is maintained. Recent studies have proposed that Cysteine Rich Motor Neuron 1 (Crim1), a transmembrane protein involved in organogenesis of many tissues, might influence cell adhesion, polarity and proliferation in the lens by regulating integrin-signaling. Here, we characterise the lens and eyes of the Crim1KST264 mutant mice, and show that the loss of Crim1 function in the ocular tissues results in inappropriate differentiation of the lens epithelium into fiber cells. Furthermore, restoration of Crim1 levels in just the lens tissue of Crim1KST264 mice is sufficient to ameliorate most of the dysgenesis observed in the mutant animals. Based on our findings, we propose that tight regulation of Crim1 activity is required for maintenance of the lens epithelium, and its depletion leads to ectopic differentiation into fiber cells, dramatically altering lens structure and ultimately leading to microphthalmia and aphakia.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/fisiología , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Cristalino/embriología , Actinas/metabolismo , Animales , Diferenciación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Desarrollo Embrionario , Epitelio/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Cristalino/citología , Cristalino/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta2/metabolismo , beta-Cristalinas/metabolismo
6.
J Mol Histol ; 48(1): 53-61, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27803996

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Vasos Coronarios/citología , Células Endoteliales/metabolismo , Homeostasis , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Supervivencia Celular/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Mutación , Transducción de Señal
8.
Histol Histopathol ; 31(10): 1049-57, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27044529

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana/metabolismo , Organogénesis/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas , Humanos
9.
Sci Rep ; 6: 19832, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26821812

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Cardiopatías Congénitas/genética , Corazón/crecimiento & desarrollo , Organogénesis/genética , Animales , Diferenciación Celular/genética , Desarrollo Embrionario/genética , Transición Epitelial-Mesenquimal/genética , Cardiopatías Congénitas/patología , Humanos , Ratones , Ratones Noqueados , Miocardio/metabolismo , Comunicación Paracrina/genética , Pericardio/embriología , Proteína Smad2/genética , Factor de Crecimiento Transformador beta1/genética
10.
J Pathol ; 229(5): 685-96, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23224993

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Linaje de la Célula , Células Endoteliales/patología , Enfermedades Renales/patología , Riñón/patología , Monocitos/patología , Mutación , Miofibroblastos/patología , Animales , Biomarcadores/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula/genética , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Marcadores Genéticos , Genotipo , Integrasas/genética , Riñón/irrigación sanguínea , Riñón/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Miofibroblastos/metabolismo , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2
11.
Genesis ; 50(9): 711-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22511315

RESUMEN

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.


Asunto(s)
Anomalías Múltiples/embriología , Receptores de Proteínas Morfogenéticas Óseas/genética , Desarrollo Embrionario/genética , Ingeniería Genética/métodos , Anomalías Múltiples/genética , Alelos , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Quimera , Cruzamientos Genéticos , Exones , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Genotipo , Integrasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Especificidad de Órganos , Organogénesis/genética , Fenotipo , Embarazo , Recombinación Genética
12.
Curr Top Dev Biol ; 90: 193-229, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20691850

RESUMEN

The mammalian kidney may well be one of the most complex organs of postnatal life. Each adult human kidney contains on average more than one million functional filtration units, the nephrons, residing within a specialized cellular interstitium. Each kidney also contains over 25 distinct cell types, each of which must be specifically aligned with respect to each other to ensure both normal development and ultimately, normal renal function. Despite this complexity, the development of the kidney can be simplistically described as the coordinate formation of two distinct sets of tubules. These tubules develop cooperatively with each other in time and space, yet represent two distinct but classical types of tubulogenesis. The first of these tubules, the ureteric bud, forms as an outgrowth of another epithelial tube, the nephric duct, and undergoes extensive branching morphogenesis to create the collecting system of the kidney. The second tubules are the nephrons themselves which arise via a mesenchyme-to-epithelial transition induced by the first set of tubules. These tubules never branch, but must elongate to become intricately patterned and functionally segmented tubules. The molecular drivers for these two tales of tubulogenesis include many gene families regulating tubulogenesis and branching morphogenesis in other organs; however, the individual players and codependent interrelationships between a branched and non-branched tubular network make organogenesis in the kidney unique. Here we review both what is known and remains to be understood in kidney tubulogenesis.


Asunto(s)
Riñón/embriología , Morfogénesis/fisiología , Organogénesis/fisiología , Animales , Regulación del Desarrollo de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Riñón/anatomía & histología , Riñón/crecimiento & desarrollo , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Túbulos Renales/anatomía & histología , Túbulos Renales/embriología , Túbulos Renales/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal/fisiología , Células Madre/citología , Células Madre/fisiología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
13.
Kidney Int ; 76(11): 1161-71, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19776720

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Riñón/irrigación sanguínea , Microvasos , Animales , Endotelio Vascular , Ratones , Ratones Transgénicos
14.
Dev Biol ; 328(1): 148-59, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19389363

RESUMEN

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.


Asunto(s)
Movimiento Celular , Vasos Coronarios/fisiología , Endotelio Vascular/citología , Pericardio/citología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Embrión no Mamífero , Endotelio Vascular/metabolismo , Miocardio/citología , Miocardio/metabolismo , Miocitos del Músculo Liso/metabolismo , Pericardio/embriología , Pericardio/metabolismo , Codorniz
15.
J Am Soc Nephrol ; 18(6): 1697-708, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17460146

RESUMEN

Crim1, a transmembrane cysteine-rich repeat-containing protein that is related to chordin, plays a role in the tethering of growth factors at the cell surface. Crim1 is expressed in the developing kidney; in parietal cells, podocytes, and mesangial cells of the glomerulus; and in pericytes that surround the arterial vasculature. A gene-trap mouse line with an insertion in the Crim1 gene (Crim1(KST264/KST264)) displayed perinatal lethality with defects in multiple organ systems. This study further analyzed the defects that are present within the kidneys of these mice. Crim1(KST264/KST264) mice displayed abnormal glomerular development, illustrated by enlarged capillary loops, podocyte effacement, and mesangiolysis. When outbred, homozygotes that reached birth displayed podocyte and glomerular endothelial cell defects and marked albuminuria. The podocytic co-expression of Crim1 with vascular endothelial growth factor-A (VEGF-A) suggested a role for Crim1 in the regulation of VEGF-A action. Crim1 and VEGF-A were shown to interact directly, providing evidence that cysteine-rich repeat-containing proteins can bind to non-TGF-beta superfamily ligands. Crim1(KST264/KST264) mice display a mislocalization of VEGF-A within the developing glomerulus, as assessed by immunogold electron microscopy and increased activation of VEGF receptor 2 (Flk1) in the glomerular endothelial cells, suggesting that Crim1 regulates the delivery of VEGF-A by the podocytes to the endothelial cells. This is the first in vivo demonstration of regulation of VEGF-A delivery and supports the hypothesis that Crim1 functions to regulate the release of growth factors from the cell of synthesis.


Asunto(s)
Albuminuria/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/embriología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Edad , Albuminuria/fisiopatología , Animales , Animales no Consanguíneos , Células COS , Capilares/embriología , Capilares/metabolismo , Chlorocebus aethiops , Cistina/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Tasa de Filtración Glomerular , Glomérulos Renales/ultraestructura , Masculino , Células Mesangiales/metabolismo , Células Mesangiales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Inmunoelectrónica , Podocitos/metabolismo , Podocitos/ultraestructura , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Dev Dyn ; 236(2): 502-11, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17106887

RESUMEN

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.


Asunto(s)
Anomalías Múltiples/embriología , Receptores de Proteínas Morfogenéticas Óseas/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/genética , Organogénesis/genética , Fenotipo , Anomalías Múltiples/genética , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Cartilla de ADN , Técnicas Histológicas , Immunoblotting , Proteínas de la Membrana/metabolismo , Ratones , Mutagénesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transgenes/genética
17.
Dev Biol ; 279(2): 378-90, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15733666

RESUMEN

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.


Asunto(s)
Tipificación del Cuerpo , Capilares/embriología , Circulación Coronaria/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Corazón/embriología , Miocardio/metabolismo , Animales , Capilares/anatomía & histología , Embrión no Mamífero/anatomía & histología , Embrión no Mamífero/fisiología , Factores de Crecimiento de Fibroblastos/genética , Corazón/anatomía & histología , Morfogénesis/fisiología , Comunicación Paracrina , Codorniz/anatomía & histología , Codorniz/embriología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Physiol Genomics ; 18(2): 129-40, 2004 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-15138308

RESUMEN

Transcriptional regulatory cascades during epicardial and coronary vascular development from proepicardial progenitor cells remain to be defined. We have used immunohistochemistry of human embryonic tissues to demonstrate that the TBX5 transcription factor is expressed not only in the myocardium, but also throughout the embryonic epicardium and coronary vasculature. TBX5 is not expressed in other human fetal vascular beds. Furthermore, immunohistochemical analyses of human embryonic tissues reveals that unlike their epicardial counterparts, delaminating epicardial-derived cells do not express TBX5 as they migrate through the subepicardium before undergoing epithelial-mesenchymal transformation required for coronary vasculogenesis. In the chick, Tbx5 is expressed in the embryonic proepicardial organ (PEO), which is composed of the epicardial and coronary vascular progenitor cells. Retrovirus-mediated overexpression of human TBX5 inhibits cell incorporation of infected proepicardial cells into the nascent chick epicardium and coronary vasculature. TBX5 overexpression as well as antisense-mediated knockdown of chick Tbx5 produce a cell-autonomous defect in the PEO that prevents proepicardial cell migration. Thus, both increasing and decreasing Tbx5 dosage impairs development of the proepicardium. Culture of explanted PEOs demonstrates that untreated chick proepicardial cells downregulate Tbx5 expression during cell migration. Therefore, we propose that Tbx5 participates in regulation of proepicardial cell migration, a critical event in the establishment of the epicardium and coronary vasculature.


Asunto(s)
Movimiento Celular/fisiología , Corazón/embriología , Pericardio/embriología , Proteínas de Dominio T Box/fisiología , Animales , Diferenciación Celular/fisiología , División Celular/genética , Línea Celular Tumoral , Embrión de Pollo , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Perros , Dosificación de Gen , Edad Gestacional , Humanos , Miocardio/química , Miocardio/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/virología , Pericardio/citología , Pericardio/metabolismo , Retroviridae/genética , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Transfección , Proteínas de Pez Cebra/genética
19.
Development ; 131(3): 581-92, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14711873

RESUMEN

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.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Diferenciación Celular/fisiología , Corazón/embriología , Ramos Subendocárdicos/metabolismo , Animales , Ácido Aspártico Endopeptidasas/genética , Tipificación del Cuerpo/fisiología , Embrión de Pollo , Regulación hacia Abajo/fisiología , Enzimas Convertidoras de Endotelina , Gadolinio/metabolismo , Corazón/fisiología , Metaloendopeptidasas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Dev Dyn ; 228(2): 161-72, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14517988

RESUMEN

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.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Corazón/embriología , Miocitos Cardíacos/metabolismo , Pericardio/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Apoptosis , División Celular , Embrión de Pollo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Inmunohistoquímica , Ligandos , Microcirugia , Mitógenos/metabolismo , Miocitos Cardíacos/citología , ARN Mensajero/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...