Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Tissue Res ; 384(3): 757-769, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33830297

RESUMEN

The adult mammalian heart is non-regenerative because cardiomyocytes withdraw from the cell cycle shortly after birth. Embryonic mammalian hearts, in which cardiomyocytes are genetically ablated in a salt-and-pepper-like pattern, regenerate due to compensation by residual cardiomyocytes. To date, it remains unknown whether or how transmural ventricular defects at the looped heart stage regenerate after cryoinjury. We established a cryoablation model in stage 16 chick embryonic hearts. In hearts at 5 h post cryoinjury (hpc), cryoinjury-induced defects were approximately 200 µm in width in the primitive ventricle; thereafter, the defect was filled with mesenchymal cells accumulating between the epicardium and endocardium. The defect began to regress at 4 days post cryoinjury (dpc) and disappeared around 9 dpc. Immunohistochemistry showed that there were no isl1-positive cells in either the scar tissue or residual cardiomyocytes. BrdU incorporation into residual cardiomyocytes was transiently downregulated in association with upregulation of p27 (Kip1), suggesting that cell cycle arrest occurred at G1-to-S transition immediately after cryoinjury. Estimated cell cycle length was examined, and the results showed that the shortest cell cycle length was 18 h at stages 19-23; it increased with development due to elongation of the G2-M-G1 phase and 30 h at stages 27-29. The S phase length was constant at 6-8 h. The cell cycle length was elongated immediately after cryoinjury, and it reversed at 1-2 dpc. Cryoablated transmural defects in the early embryonic heart were restored by compensation by residual myocytes.


Asunto(s)
Miocardio , Miocitos Cardíacos , Regeneración , Animales , Ciclo Celular , Proliferación Celular , Embrión de Pollo , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo
2.
J Mol Cell Cardiol ; 147: 62-73, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32777295

RESUMEN

BACKGROUND: During the formation of the coronary artery stem, endothelial strands from the endothelial progenitor pool surrounding the conotruncus penetrate into the aortic wall. Vascular endothelial growth factors (VEGFs) as well as CXCL12/CXCR4 signaling are thought to play a role in the formation of the coronary stem. However, the mechanisms regulating how endothelial strands exclusively invade into the aorta remain unknown. METHODS AND RESULTS: Immunohistochemistry showed that before the formation of endothelial strands, Sema3a was highly expressed in endothelial progenitors surrounding the great arteries. At the onset of/during invasion of endothelial strands into the aorta, Sema3a was downregulated and CXCR4 was upregulated in the endothelial strands. In situ hybridization showed that Cxcl12 was highly expressed in the aortic wall compared with in the pulmonary artery. Using avian embryonic hearts, we established two types of endothelial penetration assay, in which coronary endothelial strands preferentially invaded into the aorta in culture. Sema3a blocking peptide induced an excess number of endothelial strands penetrating into the pulmonary artery, whereas recombinant Sema3a inhibited the formation of endothelial strands. In cultured coronary endothelial progenitors, recombinant VEGF protein induced CXCR4-positive endothelial strands, which were capable of being attracted by CXCL12-impregnated beads. Monoazo rhodamine detected that hypoxia was predominant in aortic/subaortic region in ovo and hypoxic condition downregulated the expression of Sema3a in culture. CONCLUSION: Results suggested that hypoxia in the aortic region downregulates the expression of Sema3a, thereby enhancing VEGF activity to induce the formation of CXCR4-positive endothelial strands, which are subsequently attracted into the Cxcl12-positive aortic wall to connect the aortic lumen.


Asunto(s)
Quimiocina CXCL12/metabolismo , Vasos Coronarios/metabolismo , Regulación hacia Abajo/genética , Hipoxia/genética , Receptores CXCR4/metabolismo , Animales , Aorta/embriología , Aorta/metabolismo , Células Cultivadas , Pollos , Vasos Coronarios/embriología , Células Endoteliales/metabolismo , Codorniz/embriología , Semaforina-3A/metabolismo , Regulación hacia Arriba
3.
Genesis ; 57(7-8): e23300, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31021052

RESUMEN

Retinoic acid (RA) is a vitamin A metabolite that acts as a morphogen and teratogen. Excess or defective RA signaling causes developmental defects including in the heart. The heart develops from the anterior lateral plate mesoderm. Cardiogenesis involves successive steps, including formation of the primitive heart tube, cardiac looping, septation, chamber development, coronary vascularization, and completion of the four-chambered heart. RA is dispensable for primitive heart tube formation. Before looping, RA is required to define the anterior/posterior boundaries of the heart-forming mesoderm as well as to form the atrium and sinus venosus. In outflow tract elongation and septation, RA signaling is required to maintain/differentiate cardiogenic progenitors in the second heart field at the posterior pharyngeal arches level. Epicardium-secreted insulin-like growth factor, the expression of which is regulated by hepatic mesoderm-derived erythropoietin under the control of RA, promotes myocardial proliferation of the ventricular wall. Epicardium-derived RA induces the expression of angiogenic factors in the myocardium to form the coronary vasculature. In cardiogenic events at different stages, properly controlled RA signaling is required to establish the functional heart.


Asunto(s)
Corazón/embriología , Miocardio/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Animales , Evolución Molecular , Humanos
4.
Dev Growth Differ ; 60(2): 97-111, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29392712

RESUMEN

The origin of coronary endothelial cells (ECs) has been investigated in avian species, and the results showed that the coronary ECs originate from the proepicardial organ (PEO) and developing epicardium. Genetic approaches in mouse models showed that the major source of coronary ECs is the sinus venosus endothelium or ventricular endocardium. To clarify and reconcile the differences between avian and mouse species, we examined the source of coronary ECs in avian embryonic hearts. Using an enhanced green fluorescent protein-Tol2 system and fluorescent dye labeling, four types of quail-chick chimeras were made and quail-specific endothelial marker (QH1) immunohistochemistry was performed. The developing PEO consisted of at least two cellular populations in origin, one was sinus venosus endothelium-derived inner cells and the other was surface mesothelium-derived cells. The majority of ECs in the coronary stems, ventricular free wall, and dorsal ventricular septum originated from the sinus venosus endothelium. The ventricular endocardium contributed mainly to the septal artery and a few cells to the coronary stems. Surface mesothelial cells of the PEO differentiated mainly into a smooth muscle phenotype, but a few differentiated into ECs. In avian species, the coronary endothelium had a heterogeneous origin in a region-specific manner, and the sources of ECs were basically the same as those observed in mice.


Asunto(s)
Vasos Coronarios/embriología , Células Endoteliales/citología , Endotelio Vascular/embriología , Células Epiteliales/citología , Corazón/embriología , Animales , Diferenciación Celular , Embrión de Pollo , Pollos , Quimera/embriología , Endotelio Vascular/citología , Células Epiteliales/fisiología , Epitelio/fisiología , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Miocardio/citología , Técnicas de Cultivo de Órganos , Pericardio/citología , Pericardio/embriología , Codorniz/embriología
5.
Development ; 139(2): 385-95, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22186728

RESUMEN

The progress of molecular genetics has enabled us to identify the genes responsible for congenital heart malformations. However, recent studies suggest that congenital heart diseases are induced not only by mutations in certain genes, but also by abnormal maternal factors. A high concentration of maternal retinoic acid (RA), the active derivative of vitamin A, is well known as a teratogenic agent that can cause developmental defects. Our previous studies have shown that the maternal administration of RA to mice within a narrow developmental window induces outflow tract (OFT) septum defects, a condition that closely resembles human transposition of the great arteries (TGA), although the responsible factors and pathogenic mechanisms of the TGA induced by RA remain unknown. We herein demonstrate that the expression of Tbx2 in the OFT myocardium is responsive to RA, and its downregulation is associated with abnormal OFT development. We found that RA could directly downregulate the Tbx2 expression through a functional retinoic acid response element (RARE) in the Tbx2 promoter region, which is also required for the initiation of Tbx2 transcription during OFT development. Tgfb2 expression was also downregulated in the RA-treated OFT region and was upregulated by Tbx2 in a culture system. Moreover, defective epithelial-mesenchymal transition caused by the excess RA was rescued by the addition of Tgfß2 in an organ culture system. These data suggest that RA signaling participates in the Tbx2 transcriptional mechanism during OFT development and that the Tbx2-Tgfß2 cascade is one of the key pathways involved in inducing the TGA phenotype.


Asunto(s)
Defectos de la Almohadilla Endocárdica/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Intercambio Materno-Fetal/fisiología , Miocardio/metabolismo , Transducción de Señal/fisiología , Proteínas de Dominio T Box/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Tretinoina/metabolismo , Animales , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Defectos de la Almohadilla Endocárdica/etiología , Femenino , Galactósidos , Inmunohistoquímica , Hibridación in Situ , Indoles , Luciferasas , Ratones , Análisis por Micromatrices , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tretinoina/fisiología
6.
Dev Dyn ; 241(2): 284-93, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22184055

RESUMEN

BACKGROUND: The cardiac progenitor cells for the outflow tract (OFT) reside in the visceral mesoderm and mesodermal core of the pharyngeal region, which are defined as the secondary and anterior heart fields (SHF and AHF), respectively. RESULTS: Using chick embryos, we injected fluorescent-dye into the SHF or AHF at stage 14, and the destinations of the labeled cells were examined at stage 31. Labeled cells from the right SHF were found in the myocardium on the left dorsal side of the OFT, and cells from the left SHF were detected on the right ventral side of the OFT. Labeled cells from the right and left AHF migrated to regions of the ventral wall of the OFT close to the aortic and pulmonary valves, respectively. CONCLUSION: These observations indicate that myocardial progenitors from the SHF and AHF contribute to distinct conotruncal regions and that cells from the SHF migrate rotationally while cells from the AHF migrate in a non-rotational manner.


Asunto(s)
Movimiento Celular , Mioblastos Cardíacos/fisiología , Miocardio , Faringe/embriología , Animales , Embrión de Pollo , Cardiopatías Congénitas/embriología , Faringe/citología
7.
Differentiation ; 81(5): 299-306, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21497984

RESUMEN

Tenascin C (TNC) is an extracellular glycoprotein that is thought to be involved in tissue remodeling during organogenesis and regeneration. Using avian embryonic hearts, we investigated the spatiotemporal expression patterns of TNC during the formation of the proximal coronary artery. Immunohistochemistry showed that TNC was deposited around the developing coronary stem and that TNC colocalized with vascular smooth muscle α-actin. A quail-chick chimera, in which a quail proepicardial organ (PEO) had been transplanted, showed that quail tissue-derived cells contributed to the establishment of the endothelial and mural cells of the proximal coronary artery, and the quail tissue-derived mural cells displayed TNC. Proepicardial cells cultured in TNC showed the myofibroblast/smooth muscle cell phenotype and neutralizing anti-TNC antibody suppressed the expression of smooth muscle markers. These observations suggest that TNC plays a role in the mural smooth muscle development of the nascent proximal coronary artery.


Asunto(s)
Actinas/metabolismo , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Músculo Liso Vascular/metabolismo , Pericardio/citología , Tenascina/genética , Tenascina/metabolismo , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Embrión de Pollo , Corazón/embriología , Inmunohistoquímica/métodos , Desarrollo de Músculos , Músculo Liso Vascular/citología , Organogénesis , Pericardio/embriología , Pericardio/metabolismo , Codorniz , Tenascina/biosíntesis
8.
Dev Growth Differ ; 53(3): 366-77, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21492150

RESUMEN

During axis formation in amniotes, posterior and lateral epiblast cells in the area pellucida undergo a counter-rotating movement along the midline to form primitive streak (Polonaise movements). Using chick blastoderms, we investigated the signaling involved in this cellular movement in epithelial-epiblast. In cultured posterior blastoderm explants from stage X to XI embryos, either Lefty1 or Cerberus-S inhibited initial migration of the explants on chamber slides. In vivo analysis showed that inhibition of Nodal signaling by Lefty1 affected the movement of DiI-marked epiblast cells prior to the formation of primitive streak. In Lefty1-treated embryos without a primitive streak, Brachyury expression showed a patchy distribution. However, SU5402 did not affect the movement of DiI-marked epiblast cells. Multi-cellular rosette, which is thought to be involved in epithelial morphogenesis, was found predominantly in the posterior half of the epiblast, and Lefty1 inhibited the formation of rosettes. Three-dimensional reconstruction showed two types of rosette, one with a protruding cell, the other with a ventral hollow. Our results suggest that Nodal signaling may have a pivotal role in the morphogenetic movements of epithelial epiblast including Polonaise movements and formation of multi-cellular rosette.


Asunto(s)
Blastodermo/metabolismo , Movimiento Celular/fisiología , Pollos , Morfogénesis/fisiología , Proteína Nodal/metabolismo , Transducción de Señal/fisiología , Animales , Blastodermo/citología , Movimiento Celular/efectos de los fármacos , Embrión de Pollo , Epitelio/embriología , Proteínas Fetales/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Morfogénesis/efectos de los fármacos , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas de Dominio T Box/metabolismo
9.
Anat Rec (Hoboken) ; 304(8): 1732-1744, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33191650

RESUMEN

Cushion tissues, the primordia of valves and septa of the adult heart, are formed in the atrioventricular (AV) and outflow tract (OFT) regions of the embryonic heart. The cushion tissues are generated by the endothelial-mesenchymal transition (EMT), involving many soluble factors, extracellular matrix, and transcription factors. Moreover, neural crest-derived mesenchymal cells also migrate into the OFT cushion. The transcription factor Msx1 is known to be expressed in the endothelial and mesenchymal cells during cushion tissue formation. However, its exact role in EMT during cushion tissue formation is still unknown. In this study, we investigated the expression patterns of Msx1 mRNA and protein during chick heart development. Msx1 mRNA was localized in endothelial cells of the AV region at Stage 14, and its protein was first detected at Stage 15. Thereafter, Msx1 mRNA and protein were observed in the endothelial and mesenchymal cells of the OFT and AV regions. in vitro assays showed that ectopic Msx1 expression in endothelial cells induced p27, a cell-cycle inhibitor, expression and inhibited fibroblast growth factor 4 (FGF4)-induced cell proliferation. Although the FGF signal reduced the EMT-inducing activities of transforming growth factor ß (TGFß), ectopic Msx1 expression in endothelial cells enhanced TGFß signaling-induced αSMA, an EMT marker, expression. These results suggest that Msx1 may support the transformation of endothelial cells due to a TGFß signal in EMT during cushion tissue formation.


Asunto(s)
Proliferación Celular/fisiología , Cojinetes Endocárdicos/embriología , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Factor de Transcripción MSX1/metabolismo , Miocardio/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Embrión de Pollo , Cojinetes Endocárdicos/metabolismo , Factor de Transcripción MSX1/genética , Antígeno Nuclear de Célula en Proliferación/genética , Transducción de Señal/fisiología
10.
Anat Sci Int ; 95(4): 559-563, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32333262

RESUMEN

The brachial plexus is an important nervous structure from which all major nerves to the upper limb arise. It typically originates from the anterior rami of the C5-T1 spinal nerves. As it passes laterally, the roots are successively organized into three trunks, six divisions, and three cords. The BP is susceptible to injury during the perinatal and postnatal periods, as well as in adulthood. Its structure can show considerable variation, and there is a wealth of literature describing its variations, providing indispensable information to neurosurgeons. Here, we report a novel unilateral variant of the brachial plexus found in an adult Japanese male cadaver. In this case, the middle trunk arose from the C7 and C8 spinal nerves, and the inferior trunk continued from the T1 alone. At the interscalene triangle, the subclavian artery was situated between the C8 and T1 nerves. The posterior cord arose from the posterior divisions of the superior and middle trunks, while the root from the T1 nerve/inferior trunk was absent. The anterior division of the middle trunk gave independent roots to the musculocutaneous and median nerves, without completely establishing the lateral cord. A communicating branch arose from the musculocutaneous nerve to join the median nerve. Some branches from the roots and cords also deviated from typical configurations. This case represents a rare combination of variations in the trunks, divisions, cords, and the median nerve and offers a valuable addition to the literature regarding variations in the brachial plexus.


Asunto(s)
Variación Anatómica , Plexo Braquial/anatomía & histología , Nervios Espinales/anatomía & histología , Extremidad Superior/inervación , Anciano de 80 o más Años , Cadáver , Humanos , Masculino , Nervio Mediano/anatomía & histología , Arteria Subclavia/anatomía & histología
11.
J Vet Med Sci ; 81(1): 22-25, 2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30404952

RESUMEN

We investigated the effects of an injectable pimobendan solution (0.15 mg/kg) on cardiac function in healthy dogs. Fifteen dogs were divided into placebo, intravenous pimobendan injection, and subcutaneous pimobendan injection groups. In the placebo, the heart rate, systolic and end-diastolic left ventricular pressure (LVPs and LVEDP), and peak positive (max dP/dt) and negative (min dP/dt) first derivatives of the left ventricular pressure did not change for 60 min. After the intravenous pimobendan injection, LVEDP decreased significantly within 5 min, while the max dP/dt increased, and the effects continued until 60 min. In comparison, there were no hemodynamic changes after the subcutaneous pimobendan injection. This study demonstrates that injectable pimobendan induced a rapid inotropic effect and decreased the LVEDP in dogs.


Asunto(s)
Cardiotónicos/farmacología , Perros , Corazón/efectos de los fármacos , Piridazinas/farmacología , Vasodilatadores/farmacología , Administración Intravenosa/veterinaria , Animales , Relación Dosis-Respuesta a Droga , Femenino , Hemodinámica/efectos de los fármacos , Masculino , Inhibidores de Fosfodiesterasa 3/farmacología , Piridazinas/administración & dosificación , Distribución Aleatoria
12.
Dev Growth Differ ; 50(3): 143-57, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18312428

RESUMEN

During heart development at the gastrula stage, inhibition of bone morphogenetic protein (BMP) activity affects the heart specification but does not impair the expression of smooth muscle alpha-actin (SMA), which is first expressed in the heart mesoderm and recruited into initial heart myofibrils. Interaction of tissues between posterior epiblast and hypoblast at the early blastula stage is necessary to induce the expression of SMA, in which Nodal and Chordin are thought to be involved. Here we investigated the role of fibroblast growth factor-8 (FGF8) in the expression of SMA. In situ hybridization and reverse transcription-polymerase chain reaction showed that Fgf8b is expressed predominantly in the nascent hypoblast. Anti-FGF8b antibody inhibited the expression of SMA, cTNT, and Tbx5, which are BMP-independent heart mesoderm/early cardiomyocyte genes, but not Brachyury in cultured posterior blastoderm, and combined FGF8b and Nodal, but neither factor alone induced the expression of SMA in association with heart specific markers in cultured epiblast. Although FGF8b did not induce the upregulation of phospho-Smad2, anti-FGF8b properties suppressed phospho-Smad2 in cultured blastoderm. FGF8b was able to reverse the BMP-induced inhibition of cardiomyogenesis. The results suggest that FGF8b acts on the epiblast synergistically with Nodal at the pregastrula stage and may play a role in the expression of SMA during early cardiogenesis.


Asunto(s)
Actinas/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Estratos Germinativos/metabolismo , Corazón/embriología , Actinas/genética , Animales , Biomarcadores , Embrión de Pollo , Proteínas Fetales/biosíntesis , Proteínas Fetales/genética , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Mesodermo/metabolismo , Proteína Nodal , Técnicas de Cultivo de Órganos , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Proteína Smad2/biosíntesis , Proteína Smad2/genética , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Factor de Crecimiento Transformador beta/fisiología , Troponina T/biosíntesis , Troponina T/genética
13.
Anat Sci Int ; 93(4): 533-539, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29948975

RESUMEN

Coronary vessel development has been investigated in avian and mouse embryonic hearts. Quail embryos are a useful tool to examine vascular development, particularly because the QH1 antibody and transgenic quail line, Tg (tie1:H2B-eYFP), are useful to trace endothelial cells. However, there are only a few descriptions of the quail coronary vessels. Using ink injection coronary angiography, we examined the course of coronary vessels in the fetal quail heart. The major coronary arteries were the right and left septal arteries, which, respectively, branched off from the right and left coronary stems. The right septal artery ran posteriorly (dorsally) and penetrated the ventricular free wall to distribute to the posterior surface of the ventricles. The left septal artery ran anteriorly (ventrally) and penetrated the ventricular free wall to distribute to the anterior surface of the ventricles. The right and left circumflex arteries were directed posteriorly along the atrioventricular sulci. The cardiac veins consisted of three major tributaries: the middle, great, and anterior cardiac veins. The middle cardiac vein ascended along the posterior interventricular sulcus and emptied into the right atrium. The great cardiac vein ran along the anterior interventricular sulcus, entered the space between the left atrium and conus arteriosus and emptied into the right atrium behind the aortic bulb. The anterior cardiac vein drained the anterior surface of the right ventricle and connected to the anterior base of the right atrium. The course of coronary vessels in the quail heart was basically the same as that observed in chick but was different from those of mouse and human.


Asunto(s)
Vasos Coronarios/anatomía & histología , Embrión no Mamífero/anatomía & histología , Tabiques Cardíacos/anatomía & histología , Ventrículos Cardíacos/anatomía & histología , Codorniz/anatomía & histología , Anatomía Comparada , Animales , Vasos Coronarios/diagnóstico por imagen , Embrión no Mamífero/diagnóstico por imagen , Atrios Cardíacos/anatomía & histología , Atrios Cardíacos/diagnóstico por imagen , Tabiques Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/diagnóstico por imagen , Microscopía , Fotograbar
14.
Invest Ophthalmol Vis Sci ; 48(7): 3051-5, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17591872

RESUMEN

PURPOSE: To examine the role of tumor necrosis factor alpha (TNFalpha) in stromal neovascularization in injured cornea in vivo and in cytokine-enhanced vessel-like endothelial cell tube formation in vitro. METHODS: An in vitro model of angiogenesis was used to examine the roles of TNFalpha on tube formation by human umbilical vein endothelial cells (HUVECs) cocultured with fibroblasts on induction by transforming growth factor beta1 (TGFbeta1) and vascular endothelial growth factor (VEGF). Central cauterization was used to induce stromal neovascularization in corneas of wild-type (WT) and TNFalpha-null (Tnfalpha(-/-)) mice. At 7, 14, or 21 days of injury, experimental mice were killed, and the eyes were enucleated and subjected to histologic and immunohistochemical examination and real-time reverse transcription-polymerase chain reaction. RESULTS: HUVECs formed a vessel-like tube structure on the fibroblast feeder layer. Adding TGFbeta1, VEGF, or both augmented vessel-like tube formation by HUVECs cocultured with fibroblasts. Adding TNFalpha (5 ng/mL) completely abolished the formation of tube-like structures despite the presence or absence of TGFbeta1 or VEGF in coculture. In vivo, cauterization of the central cornea induced the formation of CD31(+) new vessels surrounding the limbus in WT mice. More prominent central stromal neovascularization accompanied by increased expression of TGFbeta1 and VEGF was found in Tnfalpha(-/-) mice compared with WT mice. CONCLUSIONS: In addition to inhibiting TGFbeta1 and VEGF expression by fibroblasts, endogenous TNFalpha may counter the induction effects of TGFbeta1 and VEGF on vascular endothelial cells and may block neovascularization.


Asunto(s)
Neovascularización de la Córnea/prevención & control , Sustancia Propia/irrigación sanguínea , Factor de Necrosis Tumoral alfa/fisiología , Animales , Técnicas de Cocultivo , Neovascularización de la Córnea/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética , Factor de Necrosis Tumoral alfa/farmacología , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
15.
Arch Ophthalmol ; 125(5): 647-54, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17502504

RESUMEN

OBJECTIVE: To determine the effects of Smad7 gene transfer in the prevention of fibrogenic responses by the retinal pigment epithelium, a major cause of proliferative vitreoretinopathy after retinal detachment, in mice. METHODS: Retinal detachment-induced proliferative vitreoretinopathy in a mouse model. Forty-eight eyes received either an adenoviral gene transfer of Smad7 or Cre recombinase gene only. The eyes were histologically analyzed. A retinal pigment epithelial cell line, ARPE-19, was used to determine whether Smad7 gene transfection suppresses the fibrogenic response to transforming growth factor (TGF) beta2 exposure. RESULTS: The Smad7 gene transfer inhibited TGF-beta2/Smad signaling in ARPE-19 cells and expression of collagen type I and TGF-beta1 but had no effect on their basal levels. In vivo Smad7 overexpression resulted in suppression of Smad2/3 signals and of the fibrogenic response to epithelial-mesenchymal transition by the retinal pigment epithelium. CONCLUSION: Smad7 gene transfer suppresses fibrogenic responses to TGF-beta2 by retinal pigment epithelial cells in vitro and in vivo. Clinical Relevance Smad7 gene transfer might be a new strategy to prevent and treat proliferative vitreoretinopathy.


Asunto(s)
Modelos Animales de Enfermedad , Expresión Génica/fisiología , Epitelio Pigmentado Ocular/metabolismo , Proteína smad7/genética , Factor de Crecimiento Transformador beta/farmacología , Vitreorretinopatía Proliferativa/prevención & control , Adenoviridae/genética , Animales , Western Blotting , Línea Celular , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Fibrosis , Técnica del Anticuerpo Fluorescente Indirecta , Vectores Genéticos , Ratones , Epitelio Pigmentado Ocular/efectos de los fármacos , Epitelio Pigmentado Ocular/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transfección , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba , Vitreorretinopatía Proliferativa/metabolismo , Vitreorretinopatía Proliferativa/patología
16.
Circ Res ; 94(3): 346-52, 2004 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-14684625

RESUMEN

Studies have shown that the proximal coronary artery (PCA) develops via endothelial ingrowth from the peritruncal ring (PR) of the coronary vasculature. However, the details of PCA formation remain unclear. We examined the development of PCAs in quail embryonic hearts from 5 to 9 days of incubation (embryonic day [ED]) using double-immunostaining for QH1 (quail endothelial marker) and smooth muscle alpha-actin. At 6 to 7 ED, several QH1-positive endothelial strands from the PR penetrated the facing sinuses, and in some embryos, several endothelial strands penetrated the posterior (noncoronary) sinus. At 7 to 8 ED, the endothelial strands penetrating the facing sinuses seemed to fuse, forming a proximal coronary stem that was demarcated from the aortic wall by the nascent smooth muscle layer of the coronary artery. By 9 ED, two coronary stems were completely formed, and the endothelial strands previously penetrating the noncoronary sinus had disappeared. Confocal microscopy at 6 ED revealed discontinuous QH1-positive endothelial progenitors in the aortic wall at sites where the endothelial strands would later develop. Observations demonstrate that during the formation of the PCA, endothelial strands from the PR penetrate the facing sinuses and then fuse, whereas those strands penetrating the noncoronary sinus disappear. Thereafter, the coronary artery tunica media demarcates the definitive PCA from the aortic media.


Asunto(s)
Vasos Coronarios/embriología , Coturnix/embriología , Embrión no Mamífero/irrigación sanguínea , Corazón/embriología , Actinas/análisis , Animales , Biomarcadores/análisis , Capilares/embriología , Embrión no Mamífero/química , Desarrollo Embrionario , Endotelio Vascular/embriología , Inmunohistoquímica , Microscopía Confocal , Microscopía Fluorescente , Músculo Liso/química , Seno Aórtico/embriología , Factores de Tiempo
17.
Kaibogaku Zasshi ; 81(4): 117-24, 2006 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-17191690

RESUMEN

In order to understand the three-dimensional gross anatomy of the heart, it is important to observe the inner structures of the chambers and the spatial relation between the valves and the ventricles. In our dissecting laboratory, we designed a guide to dissection of the heart according to the following procedures. First, we observe the surface anatomy of the heart in the pericardial cavity, remove the heart and then identify the coronary vessels, open the four chambers and observe the intra-cardiac structures. Next, we remove the atria from the ventricles and examine the relation between the valves and the chambers. Our guide is useful for learning the three-dimensional gross anatomy which is fundamental to understanding the normal function and disease of the heart.


Asunto(s)
Disección/métodos , Corazón/anatomía & histología , Humanos
18.
Congenit Anom (Kyoto) ; 56(5): 196-202, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27329052

RESUMEN

D-transposition of the great arteries (TGA) is one of the most common conotruncal heart defects at birth and is characterized by a discordant ventriculoarterial connection with a concordant atrioventricular connection. The morphological etiology of TGA is an inverted or arrested rotation of the heart outflow tract (OFT, conotruncus), by which the aorta is transposed in the right ventral direction to the pulmonary trunk. The rotational defect of the OFT is thought to be attributed to hypoplasia of the subpulmonic conus, which originates from the left anterior heart field (AHF) residing in the mesodermal core of the first and second pharyngeal arches. AHF, especially on the left, at the early looped heart stage (corresponding to Carnegie stage 10-11 in the human embryo) is one of the regions responsible for the impediment that causes TGA morphology. In human or experimentally produced right isomerism, malposition of the great arteries including D-TGA is frequently associated. Mutations in genes involving left-right (L-R) asymmetry, such as NODAL, ACTRIIB and downstream target FOXH1, have been found in patients with right isomerism as well as in isolated TGA. The downstream pathways of Nodal-Foxh1 play a critical role not only in L-R determination in the lateral plate mesoderm but also in myocardial specification and differentiation in the AHF, suggesting that TGA is a phenotype in heterotaxia as well as the primary developmental defect of the AHF.


Asunto(s)
Transposición de los Grandes Vasos/etiología , Animales , Deleción Cromosómica , Cromosomas Humanos Par 22 , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Corazón/embriología , Hemodinámica , Humanos , Ratones , Mutación , Organogénesis/genética , Fenotipo , Transposición de los Grandes Vasos/diagnóstico , Transposición de los Grandes Vasos/fisiopatología
19.
Virchows Arch ; 446(6): 663-73, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15906084

RESUMEN

Natural killer-T (NKT) cells are rich in the liver. However, their involvement in liver injury is not fully understood. We developed here a new murine model of NKT-cell-activation-associated liver injury, and investigated a role of tumor necrosis factor alpha (TNF-alpha) and Fas in pathogenesis. We injected intraperitoneally alpha-galactosylceramide (alpha-GalCer), an NKT-cell stimulant, into D-galactosamine (GalN)-sensitized mice. Survival rate, pathological changes of the liver, and plasma concentrations of cytokines were studied. Alpha-GalCer/GalN administration gave a lethal effect within 7 h, making pathological changes such as massive parenchymal hemorrhage, hepatocyte apoptosis, sinusoidal endothelial cell injury, and close apposition of lymphocytes to apoptotic hepatocytes. Anti-NK1.1 mAb-pretreated mice and Valpha14NKT knock out (KO) mice did not develop liver injury. Tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) were elevated at 4 h in the plasma. These cytokines were produced by hepatic lymphocytes as demonstrated by in vitro stimulation with alpha-GalCer. The lethal effect was suppressed in TNF-alpha KO mice, TNF receptor-1 KO mice, and lpr/lpr (Fas deficient) mice, whereas it was not in IFN-gamma KO mice. These results indicate that the present liver injury is characterized by parenchymal hemorrhage and hepatocyte apoptosis, and mediated by TNF-alpha secretion and direct cytotoxicity of alpha-GalCer-activated NKT cells.


Asunto(s)
Galactosilceramidas/toxicidad , Células Asesinas Naturales/inmunología , Hepatopatías/inmunología , Hígado/inmunología , Hígado/lesiones , Animales , Apoptosis/fisiología , Enfermedad Hepática Inducida por Sustancias y Drogas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Galactosamina/farmacología , Etiquetado Corte-Fin in Situ , Interferón gamma/sangre , Interferón gamma/deficiencia , Interferón gamma/genética , Hígado/patología , Hepatopatías/patología , Ratones , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo
20.
Congenit Anom (Kyoto) ; 45(4): 107-18, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16359490

RESUMEN

ABSTRACT The heart is the first organ to form and function during development. In the pregastrula chick embryo, cells contributing to the heart are found in the postero-lateral epiblast. During the pregastrula stages, interaction between the posterior epiblast and hypoblast is required for the anterior lateral plate mesoderm (ALM) to form, from which the heart will later develop. This tissue interaction is replaced by an Activin-like signal in culture. During gastrulation, the ALM is committed to the heart lineage by endoderm-secreted BMP and subsequently differentiates into cardiomyocyte. The right and left precardiac mesoderms migrate toward the ventral midline to form the beating primitive heart tube. Then, the heart tube generates a right-side bend, and the d-loop and presumptive heart segments begin to appear segmentally: outflow tract (OT), right ventricle, left ventricle, atrioventricular (AV) canal, atrium and sinus venosus. T-box transcription factors are involved in the formation of the heart segments: Tbx5 identifies the left ventricle and Tbx20 the right ventricle. After the formation of the heart segments, endothelial cells in the OT and AV regions transform into mesenchyme and generate valvuloseptal endocardial cushion tissue. This phenomenon is called endocardial EMT (epithelial-mesenchymal transformation) and is regulated mainly by BMP and TGFbeta. Finally, heart septa that have developed in the OT, ventricle, AV canal and atrium come into alignment and fuse, resulting in the completion of the four-chambered heart. Altered development seen in the cardiogenetic process is involved in the pathogenesis of congenital heart defects. Therefore, understanding the molecular nature regulating the 'nodal point' during heart development is important in order to understand the etiology of congenital heart defects, as well as normal heart development.


Asunto(s)
Corazón/embriología , Animales , Proteínas Morfogenéticas Óseas/fisiología , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/embriología , Mesodermo/fisiología , Organogénesis , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA