Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 31(13): 1325-1338, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28794185

RESUMO

Deciphering the fundamental mechanisms controlling cardiac specification is critical for our understanding of how heart formation is initiated during embryonic development and for applying stem cell biology to regenerative medicine and disease modeling. Using systematic and unbiased functional screening approaches, we discovered that the Id family of helix-loop-helix proteins is both necessary and sufficient to direct cardiac mesoderm formation in frog embryos and human embryonic stem cells. Mechanistically, Id proteins specify cardiac cell fate by repressing two inhibitors of cardiogenic mesoderm formation-Tcf3 and Foxa2-and activating inducers Evx1, Grrp1, and Mesp1. Most importantly, CRISPR/Cas9-mediated ablation of the entire Id (Id1-4) family in mouse embryos leads to failure of anterior cardiac progenitor specification and the development of heartless embryos. Thus, Id proteins play a central and evolutionarily conserved role during heart formation and provide a novel means to efficiently produce cardiovascular progenitors for regenerative medicine and drug discovery applications.


Assuntos
Linhagem da Célula/genética , Coração/embriologia , Proteínas Inibidoras de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/metabolismo , Organogênese/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Linhagem Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento/genética , Cardiopatias Congênitas/genética , Humanos , Mesoderma/citologia , Mesoderma/fisiologia , Camundongos , Mutação , Sementes , Xenopus laevis/embriologia
2.
Nature ; 525(7570): 479-85, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26375005

RESUMO

The elucidation of factors that activate the regeneration of the adult mammalian heart is of major scientific and therapeutic importance. Here we found that epicardial cells contain a potent cardiogenic activity identified as follistatin-like 1 (Fstl1). Epicardial Fstl1 declines following myocardial infarction and is replaced by myocardial expression. Myocardial Fstl1 does not promote regeneration, either basally or upon transgenic overexpression. Application of the human Fstl1 protein (FSTL1) via an epicardial patch stimulates cell cycle entry and division of pre-existing cardiomyocytes, improving cardiac function and survival in mouse and swine models of myocardial infarction. The data suggest that the loss of epicardial FSTL1 is a maladaptive response to injury, and that its restoration would be an effective way to reverse myocardial death and remodelling following myocardial infarction in humans.


Assuntos
Proteínas Relacionadas à Folistatina/metabolismo , Miocárdio/metabolismo , Pericárdio/crescimento & desenvolvimento , Pericárdio/metabolismo , Regeneração , Animais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Feminino , Proteínas Relacionadas à Folistatina/genética , Humanos , Masculino , Camundongos , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/efeitos dos fármacos , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Pericárdio/citologia , Pericárdio/efeitos dos fármacos , Ratos , Regeneração/efeitos dos fármacos , Transdução de Sinais , Suínos , Transgenes/genética
3.
Genes Dev ; 25(4): 299-309, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21325131

RESUMO

The adult human heart is an ideal target for regenerative intervention since it does not functionally restore itself after injury yet has a modest regenerative capacity that could be enhanced by innovative therapies. Adult cardiac cells with regenerative potential share gene expression signatures with early fetal progenitors that give rise to multiple cardiac cell types, suggesting that the evolutionarily conserved regulatory networks that drive embryonic heart development might also control aspects of regeneration. Here we discuss commonalities of development and regeneration, and the application of the rich developmental biology heritage to achieve therapeutic regeneration of the human heart.


Assuntos
Coração/embriologia , Miócitos Cardíacos/fisiologia , Regeneração/fisiologia , Adulto , Animais , Células-Tronco Embrionárias/fisiologia , Coração/fisiologia , Humanos , Modelos Biológicos , Miocárdio/citologia , Medicina Regenerativa/métodos
4.
Am J Physiol Heart Circ Physiol ; 315(2): H348-H356, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29775410

RESUMO

The G protein-coupled receptor APJ is a promising therapeutic target for heart failure. Constitutive deletion of APJ in the mouse is protective against the hypertrophy-heart failure transition via elimination of ligand-independent, ß-arrestin-dependent stretch transduction. However, the cellular origin of this stretch transduction and the details of its interaction with apelin signaling remain unknown. We generated mice with conditional elimination of APJ in the endothelium (APJendo-/-) and myocardium (APJmyo-/-). No baseline difference was observed in left ventricular function in APJendo-/-, APJmyo-/-, or control (APJendo+/+, APJmyo+/+) mice. After exposure to transaortic constriction, APJendo-/- mice displayed decreased left ventricular systolic function and increased wall thickness, whereas APJmyo-/- mice were protected. At the cellular level, carbon fiber stretch of freshly isolated single cardiomyocytes demonstrated decreased contractile responses to stretch in APJ-/- cardiomyocytes compared with APJ+/+ cardiomyocytes. Ca2+ transients did not change with stretch in either APJ-/- or APJ+/+ cardiomyocytes. Application of apelin to APJ+/+ cardiomyocytes resulted in decreased Ca2+ transients. Furthermore, hearts of mice treated with apelin exhibited decreased phosphorylation in cardiac troponin I NH2-terminal residues (Ser22 and Ser23) consistent with increased Ca2+ sensitivity. These data establish that APJ stretch transduction is mediated specifically by myocardial APJ, that APJ is necessary for stretch-induced increases in contractility, and that apelin opposes APJ's stretch-mediated hypertrophy signaling by lowering Ca2+ transients while maintaining contractility through myofilament Ca2+ sensitization. These findings underscore apelin's unique potential as a therapeutic agent that can simultaneously support cardiac function and protect against the hypertrophy-heart failure transition. NEW & NOTEWORTHY These data address fundamental gaps in our understanding of apelin-APJ signaling in heart failure by localizing APJ's ligand-independent stretch sensing to the myocardium, identifying a novel mechanism of apelin-APJ inotropy via myofilament Ca2+ sensitization, and identifying potential mitigating effects of apelin in APJ stretch-induced hypertrophic signaling.


Assuntos
Receptores de Apelina/metabolismo , Apelina/farmacologia , Insuficiência Cardíaca/metabolismo , Hipertrofia Ventricular Esquerda/metabolismo , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Animais , Receptores de Apelina/genética , Sinalização do Cálcio , Células Cultivadas , Insuficiência Cardíaca/etiologia , Hipertrofia Ventricular Esquerda/complicações , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Troponina I/metabolismo
5.
Nature ; 488(7411): 394-8, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22810587

RESUMO

Cardiac hypertrophy is initiated as an adaptive response to sustained overload but progresses pathologically as heart failure ensues. Here we report that genetic loss of APJ, a G-protein-coupled receptor, confers resistance to chronic pressure overload by markedly reducing myocardial hypertrophy and heart failure. In contrast, mice lacking apelin (the endogenous APJ ligand) remain sensitive, suggesting an apelin-independent function of APJ. Freshly isolated APJ-null cardiomyocytes exhibit an attenuated response to stretch, indicating that APJ is a mechanosensor. Activation of APJ by stretch increases cardiomyocyte cell size and induces molecular markers of hypertrophy. Whereas apelin stimulates APJ to activate Gαi and elicits a protective response, stretch signals in an APJ-dependent, G-protein-independent fashion to induce hypertrophy. Stretch-mediated hypertrophy is prevented by knockdown of ß-arrestins or by pharmacological doses of apelin acting through Gαi. Taken together, our data indicate that APJ is a bifunctional receptor for both mechanical stretch and the endogenous peptide apelin. By sensing the balance between these stimuli, APJ occupies a pivotal point linking sustained overload to cardiomyocyte hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adipocinas , Animais , Aorta/patologia , Apelina , Receptores de Apelina , Arrestinas/deficiência , Arrestinas/genética , Arrestinas/metabolismo , Pressão Sanguínea , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiomegalia/prevenção & controle , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Mecanorreceptores/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Mecanotransdução Celular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , beta-Arrestinas
6.
Development ; 140(7): 1475-85, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23462468

RESUMO

Anatomical congruence of peripheral nerves and blood vessels is well recognized in a variety of tissues. Their physical proximity and similar branching patterns suggest that the development of these networks might be a coordinated process. Here we show that large diameter coronary veins serve as an intermediate template for distal sympathetic axon extension in the subepicardial layer of the dorsal ventricular wall of the developing mouse heart. Vascular smooth muscle cells (VSMCs) associate with large diameter veins during angiogenesis. In vivo and in vitro experiments demonstrate that these cells mediate extension of sympathetic axons via nerve growth factor (NGF). This association enables topological targeting of axons to final targets such as large diameter coronary arteries in the deeper myocardial layer. As axons extend along veins, arterial VSMCs begin to secrete NGF, which allows axons to reach target cells. We propose a sequential mechanism in which initial axon extension in the subepicardium is governed by transient NGF expression by VSMCs as they are recruited to coronary veins; subsequently, VSMCs in the myocardium begin to express NGF as they are recruited by remodeling arteries, attracting axons toward their final targets. The proposed mechanism underlies a distinct, stereotypical pattern of autonomic innervation that is adapted to the complex tissue structure and physiology of the heart.


Assuntos
Vasos Coronários/fisiologia , Coração/embriologia , Coração/inervação , Sistema Nervoso Simpático/embriologia , Animais , Axônios/fisiologia , Células Cultivadas , Embrião de Galinha , Vasos Coronários/embriologia , Vasos Coronários/inervação , Técnicas de Cultura Embrionária , Embrião de Mamíferos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Músculo Liso Vascular/embriologia , Músculo Liso Vascular/inervação , Músculo Liso Vascular/metabolismo , Pericárdio/embriologia , Pericárdio/inervação , Sistema Nervoso Simpático/fisiologia
7.
Adv Funct Mater ; 25(28): 4379-4389, 2015 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-27516731

RESUMO

Biomaterials are extensively used to restore damaged tissues, in the forms of implants (e.g. tissue engineered scaffolds) or biomedical devices (e.g. pacemakers). Once in contact with the physiological environment, nanostructured biomaterials undergo modifications as a result of endogenous proteins binding to their surface. The formation of this macromolecular coating complex, known as 'protein corona', onto the surface of nanoparticles and its effect on cell-particle interactions are currently under intense investigation. In striking contrast, protein corona constructs within nanostructured porous tissue engineering scaffolds remain poorly characterized. As organismal systems are highly dynamic, it is conceivable that the formation of distinct protein corona on implanted scaffolds might itself modulate cell-extracellular matrix interactions. Here, we report that corona complexes formed onto the fibrils of engineered collagen scaffolds display specific, distinct, and reproducible compositions that are a signature of the tissue microenvironment as well as being indicative of the subject's health condition. Protein corona formed on collagen matrices modulated cellular secretome in a context-specific manner ex-vivo, demonstrating their role in regulating scaffold-cellular interactions. Together, these findings underscore the importance of custom-designing personalized nanostructured biomaterials, according to the biological milieu and disease state. We propose the use of protein corona as in situ biosensor of temporal and local biomarkers.

8.
Development ; 138(1): 139-48, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21138976

RESUMO

Epicardial signaling and Rxra are required for expansion of the ventricular myocardial compact zone. Here, we examine Raldh2(-/-) and Rxra(-/-) mouse embryos to investigate the role of retinoic acid (RA) signaling in this developmental process. The heart phenotypes of Raldh2 and Rxra mutants are very similar and are characterized by a prominent defect in ventricular compact zone growth. Although RA activity is completely lost in Raldh2(-/-) epicardium and the adjacent myocardium, RA activity is not lost in Rxra(-/-) hearts, suggesting that RA signaling in the epicardium/myocardium is not required for myocardial compact zone formation. We explored the possibility that RA-mediated target gene transcription in non-cardiac tissues is required for this process. We found that hepatic expression of erythropoietin (EPO), a secreted factor implicated in myocardial expansion, is dependent on both Raldh2 and Rxra. Chromatin immunoprecipitation studies support Epo as a direct target of RA signaling in embryonic liver. Treatment of an epicardial cell line with EPO, but not RA, upregulates Igf2. Furthermore, both Raldh2(-/-) and Rxra(-/-) hearts exhibit downregulation of Igf2 mRNA in the epicardium. EPO treatment of cultured Raldh2(-/-) hearts restores epicardial Igf2 expression and rescues ventricular cardiomyocyte proliferation. We propose a new model for the mechanism of RA-mediated myocardial expansion in which RA directly induces hepatic Epo resulting in activation of epicardial Igf2 that stimulates compact zone growth. This RA-EPO-IGF2 signaling axis coordinates liver hematopoiesis with heart development.


Assuntos
Eritropoetina/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Miocárdio/metabolismo , Pericárdio/metabolismo , Tretinoína/farmacologia , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Eritropoetina/genética , Coração/efeitos dos fármacos , Coração/embriologia , Imuno-Histoquímica , Hibridização In Situ , Fator de Crescimento Insulin-Like II/genética , Ceratolíticos/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Pericárdio/efeitos dos fármacos , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Nat Med ; 13(5): 613-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17468766

RESUMO

Growing evidence indicates that microRNAs (miRNAs or miRs) are involved in basic cell functions and oncogenesis. Here we report that miR-133 has a critical role in determining cardiomyocyte hypertrophy. We observed decreased expression of both miR-133 and miR-1, which belong to the same transcriptional unit, in mouse and human models of cardiac hypertrophy. In vitro overexpression of miR-133 or miR-1 inhibited cardiac hypertrophy. In contrast, suppression of miR-133 by 'decoy' sequences induced hypertrophy, which was more pronounced than that after stimulation with conventional inducers of hypertrophy. In vivo inhibition of miR-133 by a single infusion of an antagomir caused marked and sustained cardiac hypertrophy. We identified specific targets of miR-133: RhoA, a GDP-GTP exchange protein regulating cardiac hypertrophy; Cdc42, a signal transduction kinase implicated in hypertrophy; and Nelf-A/WHSC2, a nuclear factor involved in cardiogenesis. Our data show that miR-133, and possibly miR-1, are key regulators of cardiac hypertrophy, suggesting their therapeutic application in heart disease.


Assuntos
Cardiomegalia/genética , MicroRNAs/genética , Animais , Aorta Torácica/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Oncogênica v-akt/genética , Ratos
10.
Cell Regen ; 13(1): 2, 2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38291287

RESUMO

The regenerative capacity of the adult mammalian heart remains a formidable challenge in biological research. Despite extensive investigations into the loss of regenerative potential during evolution and development, unlocking the mechanisms governing cardiomyocyte proliferation remains elusive. Two recent groundbreaking studies have provided fresh perspectives on mitochondrial-to-nuclear communication, shedding light on novel factors that regulate cardiomyocyte proliferation. The studies identified two mitochondrial processes, fatty acid oxidation and protein translation, as key players in restricting cardiomyocyte proliferation. Inhibition of these processes led to increased cell cycle activity in cardiomyocytes, mediated by reduction in H3k4me3 levels through accumulated α-ketoglutarate (αKG), and activation of the mitochondrial unfolded protein response (UPRmt), respectively. In this research highlight, we discuss the novel insights into mitochondrial-to-nuclear communication presented in these studies, the broad implications in cardiomyocyte biology and cardiovascular diseases, as well as the intriguing scientific questions inspired by the studies that may facilitate future investigations into the detailed molecular mechanisms of cardiomyocyte metabolism, proliferation, and mitochondrial-to-nuclear communications.

12.
Am J Hum Genet ; 86(2): 254-61, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20137777

RESUMO

Frank-Ter Haar syndrome (FTHS), also known as Ter Haar syndrome, is an autosomal-recessive disorder characterized by skeletal, cardiovascular, and eye abnormalities, such as increased intraocular pressure, prominent eyes, and hypertelorism. We have conducted homozygosity mapping on patients representing 12 FTHS families. A locus on chromosome 5q35.1 was identified for which patients from nine families shared homozygosity. For one family, a homozygous deletion mapped exactly to the smallest region of overlapping homozygosity, which contains a single gene, SH3PXD2B. This gene encodes the TKS4 protein, a phox homology (PX) and Src homology 3 (SH3) domain-containing adaptor protein and Src substrate. This protein was recently shown to be involved in the formation of actin-rich membrane protrusions called podosomes or invadopodia, which coordinate pericellular proteolysis with cell migration. Mice lacking Tks4 also showed pronounced skeletal, eye, and cardiac abnormalities and phenocopied the majority of the defects associated with FTHS. These findings establish a role for TKS4 in FTHS and embryonic development. Mutation analysis revealed five different homozygous mutations in SH3PXD2B in seven FTHS families. No SH3PXD2B mutations were detected in six other FTHS families, demonstrating the genetic heterogeneity of this condition. Interestingly however, dermal fibroblasts from one of the individuals without an SH3PXD2B mutation nevertheless expressed lower levels of the TKS4 protein, suggesting a common mechanism underlying disease causation.


Assuntos
Anormalidades Múltiplas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Anormalidades do Olho/complicações , Cardiopatias Congênitas/complicações , Anormalidades Musculoesqueléticas/complicações , Mutação/genética , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Sequência de Aminoácidos , Animais , Pré-Escolar , Mapeamento Cromossômico , Anormalidades do Olho/genética , Feminino , Inativação Gênica , Cardiopatias Congênitas/genética , Homozigoto , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Anormalidades Musculoesqueléticas/genética , Proteínas de Transferência de Fosfolipídeos/química , Síndrome
13.
Muscle Nerve ; 45(1): 128-30, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22190319

RESUMO

The protein kinase product of the gene mutated in myotonic dystrophy 1 (DMPK) is reported to play a role in cardiac pathophysiology. To gain insight into the molecular mechanisms modulated by DMPK, we characterize the impact of DMPK ablation in the context of cardiac ß-adrenergic function. Our data demonstrate that DMPK knockout mice present altered ß-agonist-induced responses and suggest that this is due, at least in part, to a reduced density of ß(1)-adrenergic receptors in cardiac plasma membranes.


Assuntos
Proteínas Serina-Treonina Quinases/deficiência , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ecocardiografia , Isoproterenol/farmacologia , Camundongos , Camundongos Knockout , Miocárdio/citologia , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Distrofia Miotônica/fisiopatologia , Miotonina Proteína Quinase , Fosforilação/efeitos dos fármacos , Receptores Adrenérgicos beta/sangue , Serina/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteínas de Transporte Vesicular/metabolismo
14.
Biol Cell ; 103(10): 467-81, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21732911

RESUMO

BACKGROUND: Blood vessel formation is important for many physiological and pathological processes and is therefore a critical target for drug development. Inhibiting angiogenesis to starve a tumour or promoting 'normalization' of tumour vasculature in order to facilitate delivery of anticancer drugs are both areas of active research. Recapitulation of vessel formation by human cells in vitro allows the investigation of cell-cell and cell-matrix interactions in a controlled environment and is therefore a crucial step in developing HCS (high content screening) and HTS (high throughput screening) assays to search for modulators of blood vessel formation. HUVECs (human umbilical-vein endothelial cells) exemplify primary cells used in angiogenesis assays. However, primary cells have significant limitations that include phenotypic decay and/or senescence by six to eight passages in culture, making stable integration of fluorescent markers and large-scale expansion for HTS problematic. To overcome these limitations for HTS, we developed a novel angiogenic model system that employs stable fluorescent endothelial cell lines based on immortalized HMECs (human microvascular endothelial cell). We then evaluated HMEC cultures, both alone and co-cultured with an EMC (epicardial mesothelial cell) line that contributes vascular smooth muscle cells, to determine the suitability for HTS or HCS. RESULTS: The endothelial and epicardial lines were engineered to express a panel of nuclear- and cytoplasm-localized fluorescent proteins to be mixed and matched to suit particular experimental goals. HMECs retained their angiogenic potential and stably expressed fluorescent proteins for at least 13 passages after transduction. Within 8 h after plating on Matrigel, the cells migrated and coalesced into networks of vessel-like structures. If co-cultured with EMCs, the branches formed cylindrical-shaped structures of HMECs surrounded by EMC derivatives reminiscent of vessels. Network formation measurements revealed responsiveness to media composition and control compounds. CONCLUSIONS: HMEC-based lines retain most of the angiogenic features of primary endothelial cells and yet possess long-term stability and ease of culture, making them intriguing candidates for large-scale primary HCS and HTS (of ~10000-1000000 molecules). Furthermore, inclusion of EMCs demonstrates the feasibility of using epicardial-derived cells, which normally contribute to smooth muscle, to model large vessel formation. In summary, the immortalized fluorescent HMEC and EMC lines and straightforward culture conditions will enable assay development for HCS of angiogenesis.


Assuntos
Linhagem Celular , Células Endoteliais da Veia Umbilical Humana , Neovascularização Patológica , Neovascularização Fisiológica , Bioensaio , Comunicação Celular , Microambiente Celular , Técnicas de Cocultura , Corantes Fluorescentes , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Modelos Cardiovasculares , Miócitos de Músculo Liso/citologia , Pericárdio/citologia , Sensibilidade e Especificidade
15.
Front Med Technol ; 4: 788264, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35252962

RESUMO

Left ventricular (LV) catheterization provides LV pressure-volume (P-V) loops and it represents the gold standard for cardiac function monitoring. This technique, however, is invasive and this limits its applicability in clinical and in-home settings. Ballistocardiography (BCG) is a good candidate for non-invasive cardiac monitoring, as it is based on capturing non-invasively the body motion that results from the blood flowing through the cardiovascular system. This work aims at building a mechanistic connection between changes in the BCG signal, changes in the P-V loops and changes in cardiac function. A mechanism-driven model based on cardiovascular physiology has been used as a virtual laboratory to predict how changes in cardiac function will manifest in the BCG waveform. Specifically, model simulations indicate that a decline in LV contractility results in an increase of the relative timing between the ECG and BCG signal and a decrease in BCG amplitude. The predicted changes have subsequently been observed in measurements on three swine serving as pre-clinical models for pre- and post-myocardial infarction conditions. The reproducibility of BCG measurements has been assessed on repeated, consecutive sessions of data acquisitions on three additional swine. Overall, this study provides experimental evidence supporting the utilization of mechanism-driven mathematical modeling as a guide to interpret changes in the BCG signal on the basis of cardiovascular physiology, thereby advancing the BCG technique as an effective method for non-invasive monitoring of cardiac function.

16.
Dev Biol ; 328(2): 483-92, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19233157

RESUMO

The outermost layer of the vertebrate heart originates from migratory mesothelial cells (epicardium) that give rise to coronary vascular smooth muscles and fibroblasts. The role of the epicardium in myocardial morphogenesis and establishment of normal heart function is still largely unknown. Here, we use Drosophila to investigate non-autonomous influences of epicardial-like tissue surrounding the heart tube on the structural and functional integrity of the myocardium. It has previously been shown that during Drosophila heart formation, mesodermal expression of the homeobox transcription factor even-skipped (eve) is required for specification of a subset of non-myocardial progenitors in the precardiac mesoderm. These progenitors may share some similarities with the vertebrate epicardium. To investigate a non-autonomous epicardial-like influence on myocardial physiology, we studied the consequences of reduced mesodermal Eve expression and epi/pericardial cell numbers on the maturation of the myocardial heart tube, its contractility, and acquisition of a normal heart rhythm in the Drosophila model. Targeting the eve repressor ladybird early (lbe) with the minimal eve mesodermal enhancer efficiently eliminates the mesodermal Eve lineages. These flies exhibit defects in heart structure, including a reduction in systolic and diastolic diameter (akin to 'restrictive cardiomyopathy'). They also exhibit an elevated incidence of arrhythmias and intermittent asystoles, as well as compromised performance under stress. These abnormalities are restored by eve reexpression or by lbe-RNAi co-overexpression. The data suggest that adult heart function in Drosophila is likely to be modulated non-autonomously, possibly by paracrine influences from neighboring cells, such as the epi/pericardium. Thus, Drosophila may serve as a model for finding genetic effectors of epicardial-myocardial interactions relevant to higher organisms.


Assuntos
Diferenciação Celular/fisiologia , Drosophila/fisiologia , Animais , Padronização Corporal/fisiologia , Drosophila/anatomia & histologia , Drosophila/embriologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Epitélio/fisiologia , Coração/anatomia & histologia , Coração/embriologia , Coração/fisiologia , Frequência Cardíaca , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Contração Miocárdica/fisiologia , Pericárdio/citologia , Pericárdio/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
17.
Proc Natl Acad Sci U S A ; 104(46): 18109-14, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17989236

RESUMO

We have previously identified several members of the Wnt/beta-catenin pathway that are differentially expressed in a mouse model with deficient coronary vessel formation. Systemic ablation of beta-catenin expression affects mouse development at gastrulation with failure of both mesoderm development and axis formation. To circumvent this early embryonic lethality and study the specific role of beta-catenin in coronary arteriogenesis, we have generated conditional beta-catenin-deletion mutant animals in the proepicardium by interbreeding with a Cre-expressing mouse that targets coronary progenitor cells in the proepicardium and its derivatives. Ablation of beta-catenin in the proepicardium results in lethality between embryonic day 15 and birth. Mutant mice display impaired coronary artery formation, whereas the venous system and microvasculature are normal. Analysis of proepicardial beta-catenin mutant cells in the context of an epicardial tracer mouse reveals that the formation of the proepicardium, the migration of proepicardial cells to the heart, and the formation of the primitive epicardium are unaffected. However, subsequent processes of epicardial development are dramatically impaired in epicardial-beta-catenin mutant mice, including failed expansion of the subepicardial space, blunted invasion of the myocardium, and impaired differentiation of epicardium-derived mesenchymal cells into coronary smooth muscle cells. Our data demonstrate a functional role of the epicardial beta-catenin pathway in coronary arteriogenesis.


Assuntos
Pericárdio/citologia , Células-Tronco/citologia , beta Catenina/metabolismo , Animais , Sequência de Bases , Linhagem da Célula , Primers do DNA , Imuno-Histoquímica , Camundongos , Camundongos Knockout , beta Catenina/genética
18.
Dev Biol ; 322(1): 208-18, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18718461

RESUMO

Trophic factors secreted both from the endocardium and epicardium regulate appropriate growth of the myocardium during cardiac development. Epicardially-derived cells play also a key role in development of the coronary vasculature. This process involves transformation of epithelial (epicardial) cells to mesenchymal cells (EMT). Similarly, a subset of endocardial cells undergoes EMT to form the mesenchyme of endocardial cushions, which function as primordia for developing valves and septa. While it has been suggested that transforming growth factor-betas (Tgf-beta) play an important role in induction of EMT in the avian epi- and endocardium, the function of Tgf-betas in corresponding mammalian tissues is still poorly understood. In this study, we have ablated the Tgf-beta type I receptor Alk5 in endo-, myo- and epicardial lineages using the Tie2-Cre, Nkx2.5-Cre, and Gata5-Cre driver lines, respectively. We show that while Alk5-mediated signaling does not play a major role in the myocardium during mouse cardiac development, it is critically important in the endocardium for induction of EMT both in vitro and in vivo. Moreover, loss of epicardial Alk5-mediated signaling leads to disruption of cell-cell interactions between the epicardium and myocardium resulting in a thinned myocardium. Furthermore, epicardial cells lacking Alk5 fail to undergo Tgf-beta-induced EMT in vitro. Late term mutant embryos lacking epicardial Alk5 display defective formation of a smooth muscle cell layer around coronary arteries, and aberrant formation of capillary vessels in the myocardium suggesting that Alk5 is controlling vascular homeostasis during cardiogenesis. To conclude, Tgf-beta signaling via Alk5 is not required in myocardial cells during mammalian cardiac development, but plays an irreplaceable cell-autonomous role regulating cellular communication, differentiation and proliferation in endocardial and epicardial cells.


Assuntos
Coração/embriologia , Miocárdio/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Animais , Apoptose , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem da Célula , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/citologia , Vasos Coronários/embriologia , Coxins Endocárdicos/citologia , Coxins Endocárdicos/embriologia , Endocárdio/citologia , Endocárdio/embriologia , Marcação de Genes , Coração/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Técnicas de Cultura de Órgãos , Pericárdio/citologia , Pericárdio/efeitos dos fármacos , Pericárdio/embriologia , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais/genética , Fator de Crescimento Transformador beta1/farmacologia
19.
Dev Biol ; 319(2): 258-66, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18508041

RESUMO

Dissecting the molecular mechanisms that guide the proper development of epicardial cell lineages is critical for understanding the etiology of both congenital and adult forms of human cardiovascular disease. In this study, we describe the function of BAF180, a polybromo protein in ATP-dependent SWI/SNF chromatin remodeling complexes, in coronary development. Ablation of BAF180 leads to impaired epithelial-to-mesenchymal-transition (EMT) and arrested maturation of epicardium around E11.5. Three-dimensional collagen gel assays revealed that the BAF180 mutant epicardial cells indeed possess significantly compromised migrating and EMT potentials. Consequently, the mutant hearts form abnormal surface nodules and fail to develop the fine and continuous plexus of coronary vessels that cover the entire ventricle around E14. PECAM and *-SMA staining assays indicate that these nodules are defective structures resulting from the failure of endothelial and smooth muscle cells within them to form coronary vessels. PECAM staining also reveal that there are very few coronary vessels inside the myocardium of mutant hearts. Consistent with this, quantitative RT-PCR analysis indicate that the expression of genes involved in FGF, TGF, and VEGF pathways essential for coronary development are down-regulated in mutant hearts. Together, these data reveal for the first time that BAF180 is critical for coronary vessel formation.


Assuntos
Vasos Coronários/embriologia , Coração Fetal/fisiologia , Coração/crescimento & desenvolvimento , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Trifosfato de Adenosina/metabolismo , Animais , Vasos Coronários/fisiologia , Proteínas de Ligação a DNA , Embrião de Mamíferos/fisiologia , Proteínas HMGB , Camundongos , Mutação , Pericárdio/embriologia , Pericárdio/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Biotechniques ; 46(2): 101-13, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19317655

RESUMO

The genetic basis of heart development is remarkably conserved from Drosophila to mammals, and insights from flies have greatly informed our understanding of vertebrate heart development. Recent evidence suggests that many aspects of heart function are also conserved and the genes involved in heart development also play roles in adult heart function. We have developed a Drosophila heart preparation and movement analysis algorithm that allows quantification of functional parameters. Our methodology combines high-speed optical recording of beating hearts with a robust, semi-automated analysis to accurately detect and quantify, on a beat-to-beat basis, not only heart rate but also diastolic and systolic intervals, systolic and diastolic diameters, percent fractional shortening, contraction wave velocity, and cardiac arrhythmicity. Here, we present a detailed analysis of hearts from adult Drosophila, 2-3-day-old zebrafish larva, and 8-day-old mouse embryos, indicating that our methodology is potentially applicable to an array of biological models. We detect progressive age-related changes in fly hearts as well as subtle but distinct cardiac deficits in Tbx5 heterozygote mutant zebrafish. Our methodology for quantifying cardiac function in these genetically tractable model systems should provide valuable insights into the genetics of heart function.


Assuntos
Algoritmos , Pressão Sanguínea/fisiologia , Frequência Cardíaca/fisiologia , Coração/embriologia , Coração/fisiologia , Interpretação de Imagem Assistida por Computador/métodos , Contração Miocárdica/fisiologia , Função Ventricular Esquerda/fisiologia , Animais , Drosophila , Coração/anatomia & histologia , Imageamento Tridimensional/métodos , Camundongos , Movimento/fisiologia , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA