Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(16)2022 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-36012498

RESUMO

Sinoatrial node dysfunction can manifest as bradycardia, leading to symptoms of syncope and sudden cardiac death. Electronic pacemakers are the current standard of care but are limited due to a lack of biological chronotropic control, cost of revision surgeries, and risk of lead- and device-related complications. We therefore aimed to develop a biological alternative to electronic devices by using a clinically relevant gene therapy vector to demonstrate conversion of cardiomyocytes into sinoatrial node-like cells in an in vitro context. Neonatal rat ventricular myocytes were transduced with recombinant adeno-associated virus vector 6 encoding either hTBX18 or green fluorescent protein and maintained for 3 weeks. At the endpoint, qPCR, Western blot analysis and immunocytochemistry were used to assess for reprogramming into pacemaker cells. Cell morphology and Arclight action potentials were imaged via confocal microscopy. Compared to GFP, hTBX18-transduced cells showed that hTBX18, HCN4 and Cx45 were upregulated. Cx43 was significantly downregulated, while sarcomeric α-actinin remained unchanged. Cardiomyocytes transduced with hTBX18 acquired the tapering morphology of native pacemaker cells, as compared to the block-like, striated appearance of ventricular cardiomyocytes. Analysis of the action potentials showed phase 4 depolarization and a significant decrease in the APD50 of the hTBX18-transduced cells. We have demonstrated that rAAV-hTBX18 gene transfer to ventricular myocytes results in morphological, molecular, physiological, and functional changes, recapitulating the pacemaker phenotype in an in vitro setting. The generation of these induced pacemaker-like cells using a clinically relevant vector opens new prospects for biological pacemaker development.


Assuntos
Miócitos Cardíacos , Nó Sinoatrial , Potenciais de Ação , Animais , Relógios Biológicos/fisiologia , Dependovirus , Vetores Genéticos/genética , Miócitos Cardíacos/metabolismo , Ratos
2.
Gene Ther ; 26(9): 399-406, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31467408

RESUMO

Differences between mouse and human hearts pose a significant limitation to the value of small animal models when predicting vector behavior following recombinant adeno-associated viral (rAAV) vector-mediated cardiac gene therapy. Hence, sheep have been adopted as a preclinical animal, as they better model the anatomy and cardiac physiological processes of humans. There is, however, no comprehensive data on the shedding profile of rAAV in sheep following intracoronary delivery, so as to understand biosafety risks in future preclinical and clinical applications. In this study, sheep received intracoronary delivery of rAAV serotypes 2/6 (2 × 1012 vg), 2/8, and 2/9 (1 × 1013 vg) at doses previously administered in preclinical and clinical trials. This was followed by assessment over 96 h to examine vector shedding in urine, feces, nasal mucus, and saliva samples. Vector genomes were detected via real-time quantitative PCR in urine and feces up to 48 and 72 h post vector delivery, respectively. Of these results, functional vector particles were only detected via a highly sensitive infectious replication assay in feces samples up to 48 h following vector delivery. We conclude that rAAV-mediated gene transfer into sheep hearts results in low-grade shedding of non-functional vector particles for all excreta samples, except in the case of feces, where functional vector particles are present up to 48 h following vector delivery. These results may be used to inform containment and decontamination guidelines for large animal dealings, and to understand the biosafety risks associated with future preclinical and clinical uses of rAAV.


Assuntos
Dependovirus/genética , Vetores Genéticos , Eliminação de Partículas Virais , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Cateterismo , Vasos Coronários , Dependovirus/imunologia , Dependovirus/fisiologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Células HeLa , Humanos , Injeções Intra-Arteriais , Masculino , Infecções por Parvoviridae/imunologia , Infecções por Parvoviridae/urina , Infecções por Parvoviridae/virologia , Reação em Cadeia da Polimerase em Tempo Real , Ovinos , Replicação Viral
3.
Heart Lung Circ ; 27(8): 967-975, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29573957

RESUMO

BACKGROUND: Vitamin D has been implicated in the prevention of heart failure. However the underlying mechanism remains unclear. We hypothesised that these effects may be partially mediated by cardiac stem/progenitor cells (CPCs). Therefore, we examined the effects of 1,25-dihydroxyvitamin D3 (1,25D) on cell cycle activity and differentiation of a previously described CPC population called cardiac colony-forming unit fibroblasts (cCFU-Fs). METHODS: cCFU-Fs were isolated from adult male C57Bl/6 mouse hearts using fluorescence-activated cell sorting. The effect of 1,25D on cell proliferation and differentiation were was assessed by colony-forming and fibroblast differentiation assays. Cell cycle was analysed by flow cytometry. Mice with induced myocardial infarction (MI) were treated with 1,25D or vehicle controls and cardiac function assessed by echocardiography. RESULTS: 1,25D dose-dependently increased expression of vitamin D receptor (Vdr) and reduced large colony formation. Addition of 1,25D to cCFU-Fs slowed cell proliferation, promoted cell cycle arrest and decreased expression of pro-fibrotic factors during TGF-ß-induced fibroblast differentiation of cCFU-Fs. After MI, 1,25D-treated mice had less left ventricular wall thinning and significant improvement in left ventricular systolic function compared to vehicle-treated controls. Although no significant changes in myocardial fibrotic area and cardiomyocyte size were noted, treatment with 1,25D significantly inhibited cardiac interstitial cell proliferation after MI. CONCLUSIONS: Vitamin D signalling promotes cardioprotection after myocardial infarction. This may be through modulation of cCFU-F cell cycle. The role of 1,25D and VDR in regulating cardiac stem/progenitor cell function therefore warrants further investigation.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Insuficiência Cardíaca/prevenção & controle , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Vitamina D/uso terapêutico , Animais , Células Cultivadas , Modelos Animais de Doenças , Ecocardiografia , Citometria de Fluxo , Insuficiência Cardíaca/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/diagnóstico , Vitaminas/uso terapêutico
4.
Mater Sci Eng C Mater Biol Appl ; 96: 824-830, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30606596

RESUMO

Polyester-based scaffolds have been employed in tissue engineering due to their biocompatibility, biodegradability, microstructure, and affordability. However, the acidic degradation byproducts of most common polyesters have the potential to cause inflammation and/or necrosis. In this study, we introduce a porous scaffold with benign degradation byproducts fabricated by gas-foaming based on poly(propylene carbonate) (PPC) blended with starch and bioglass particles. The pore sizes ranged from 100 to 500 µm. Manufacturing parameters were tuned from sub-critical to super-critical conditions to optimize porosity, pore size, pore interconnectivity, and mechanical properties. The biological behavior of the constructs was evaluated by in vitro toxicity and proliferation assays and in vivo subcutaneous biocompatibility. Tissue integration was observed in a joint implantation model, supporting the further development of the scaffold for tissue engineering applications.


Assuntos
Implantes Absorvíveis , Fibroblastos/metabolismo , Gases/química , Teste de Materiais , Polipropilenos/química , Alicerces Teciduais/química , Fibroblastos/citologia , Humanos , Porosidade
5.
Adv Healthc Mater ; 8(1): e1801307, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30511808

RESUMO

Considerable progress has been made in the field of microfluidics to develop complex systems for modeling human skin and dermal wound healing processes. While microfluidic models have attempted to integrate multiple cell types and/or 3D culture systems, to date they have lacked some elements needed to fully represent dermal wound healing. This paper describes a cost-effective, multicellular microfluidic system that mimics the paracrine component of early inflammation close to normal wound healing. Collagen and Matrigel are tested as materials for coating and adhesion of dermal fibroblasts and human umbilical vein endothelial cells (HUVECs). The wound-on-chip model consists of three interconnecting channels and is able to simulate wound inflammation by adding tumor necrosis factor alpha (TNF-α) or by triculturing with macrophages. Both the approaches significantly increase IL-1ß, IL-6, IL-8 in the supernatant (p < 0.05), and increases in cytokine levels are attenuated by cotreatment with an anti-inflammatory agent, Dexamethasone. Incorporation of M1 and M2 macrophages cocultured with fibroblasts and HUVECs leads to a stimulation of cytokine production as well as vascular structure formation, particularly with M2 macrophages. In summary, this wound-on-chip system can be used to model the paracrine component of the early inflammatory phase of wound healing and has the potential for the screening of anti-inflammatory compounds.


Assuntos
Microambiente Celular , Derme/patologia , Inflamação/patologia , Dispositivos Lab-On-A-Chip , Modelos Biológicos , Cicatrização , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Citocinas/metabolismo , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Necrose Tumoral alfa
6.
Stem Cells Dev ; 27(3): 184-198, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29205098

RESUMO

Cardiac progenitor cells (CPCs) are being developed as a promising treatment for heart failure. Although clinical trials have predominantly used donor cardiac biopsies to derive CPCs, a better solution could be to use previously cryopreserved human heart tissue. This would enable timely and convenient access to healthy and young heart samples for CPC production. However, few studies have attempted to isolate CPCs from previously cryopreserved heart tissue. In this study, we isolated CPCs from eight nondiseased human heart samples previously cryopreserved as part of the Sydney Heart Bank. Resulting cells were strongly positive for known fibroblast (DDR2, Vimentin), mesenchymal/CPC (PDGFRα, CD90) markers, and for pluripotency genes (SOX2, NANOG, MYC, KLF4), whereas being negative for the pan-hematopoietic marker (CD45). Outgrowth cells from aged hearts had decreased proliferative and self-renewing capacity that correlated with shorter telomere lengths compared with cells from young hearts. No telomerase activity was detected in any cells isolated. Colony-forming assays and fluorescence-activated cell sorting were used to enrich PDGFRα+/CD90+/CD31- CPCs. Multipotent potential was confirmed using in vitro differentiation assays with smooth muscle (MYH11+), endothelial cell (vWF+), and cardiomyocyte-like (cTnT+, α-actinin+) cell formation. Single cell assays demonstrated clonogenicity of PDGFRα+ CPCs with maintenance of prolonged self-renewing capacity (>2 months), and pluripotency gene expression at both early and late culture passages. Our results demonstrate that multipotent PDGFRα+ CPCs can be harvested and expanded from previously banked cryopreserved human heart samples. These data support cardiac tissue banking as a strategy for improved access to CPCs for future clinical therapies.


Assuntos
Antígenos de Diferenciação/biossíntese , Separação Celular , Criopreservação , Regulação da Expressão Gênica , Miocárdio , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/biossíntese , Células Cultivadas , Humanos , Fator 4 Semelhante a Kruppel , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo
7.
Biophys Rev ; 7(1): 127-139, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28509972

RESUMO

The discovery of multiple classes of cardiac progenitor cells in the adult mammalian heart has generated hope for their use as a therapeutic in heart failure. However, successful results from animal models have not always yielded similar findings in human studies. Recent Phase I/II trials of c-Kit (SCIPIO) and cardiosphere-based (CADUCEUS) cardiac progenitor cells have demonstrated safety and some therapeutic efficacy. Gaps remain in our understanding of the origins, function and relationships between the different progenitor cell families, many of which are heterogeneous populations with overlapping definitions. Another challenge lies in the limitations of small animal models in replicating the human heart. Cryopreserved human cardiac tissue provides a readily available source of cardiac progenitor cells and may help address these questions. We review important findings and relative unknowns of the main classes of cardiac progenitor cells, highlighting differences between animal and human studies.

8.
Mol Endocrinol ; 29(8): 1144-55, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26121234

RESUMO

Aldosterone (Aldo) activates both genomic and nongenomic signaling pathways in the cardiovascular system. Activation of genomic signaling pathways contributes to the adverse cardiac actions of Aldo during reperfusion injury; however, the extent nongenomic signaling pathways contribute has been difficult to identify due to lack of a specific ligand that activates only nongenomic signaling pathways. Using a pegylated aldosterone analog, aldosterone-3-carboxymethoxylamine-TFP ester conjugated to methoxypegylated amine (Aldo-PEG), we are able for the first time to distinguish between nongenomic and genomic cardiac actions of Aldo. We confirm Aldo-PEG activates phosphorylation of ERK1/2 in rat cardiomyocyte H9c2 cells similar to Aldo and G protein-coupled receptor 30 (GPR30 or GPER) agonist G1. GPER antagonist, G36, but not mineralocorticoid receptor (MR) antagonist spironolactone, prevented ERK1/2 phosphorylation by Aldo, Aldo-PEG, and G1. The selective nongenomic actions of Aldo-PEG are confirmed, with Aldo-PEG increasing superoxide production in H9c2 cells to similar levels as Aldo but having no effect on subcellular localization of MR. Striatin serves as a scaffold for GPER and MR, with GPER antagonist G36, but not spironolactone, restoring MR-striatin complexes. Aldo-PEG had no effect on MR-dependent transcriptional activation, whereas Aldo increased transcript levels of serum-regulated kinase 1 and plasminogen activator inhibitor-1. Using our ex vivo experimental rat model of myocardial infarction, we found aggravated infarct size and apoptosis by Aldo but not Aldo-PEG. Our studies confirm that in the heart, activation of nongenomic signaling pathways alone are not sufficient to trigger the deleterious effects of aldosterone during myocardial reperfusion injury.


Assuntos
Aldosterona/farmacologia , Traumatismo por Reperfusão/metabolismo , Aminas/química , Animais , Proteínas de Ligação a Calmodulina/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Traumatismo por Reperfusão Miocárdica , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais , Espironolactona/farmacologia , Superóxidos/metabolismo , Ativação Transcricional
9.
Endocrinology ; 155(2): 568-75, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24424037

RESUMO

Age-specific incidence of ischemic heart disease in men is higher than in women, although women die more frequently without previous symptoms; the molecular mechanism(s) are poorly understood. Most studies focus on protection by estrogen, with less attention on androgen receptor-mediated androgen actions. Our aim was to determine the role of androgens in the sex differences in cardiac damage during myocardial infarction. Mature age-matched male and female Sprague Dawley rats, intact or surgically gonadectomized (Gx), received testosterone (T) or 17ß-estradiol (E2) via subdermal SILASTIC (Dow Corning Corp.) implants; a subset of male rats received dihydrotestosterone. After 21 days, animals were anesthetized, and hearts were excised and subjected to ex vivo regional ischemia-reperfusion (I-R). Hearts from intact males had larger infarcts than those from females following I-R; Gx produced the opposite effect, confirming a role for sex steroids. In Gx males, androgens (dihydrotestosterone, T) and E2 aggravated I-R-induced cardiac damage, whereas in Gx females, T had no effect and E2 reduced infarct area. Increased circulating T levels up-regulated androgen receptor and receptor for advanced glycation end products, which resulted in enhanced apoptosis aggravating cardiac damage in both males and females. In conclusion, our study demonstrates, for the first time, that sex steroids regulate autophagy during myocardial infarction and shows that a novel mechanism of action for androgens during I-R is down-regulation of antiapoptotic protein Bcl-xL (B cell lymphoma-extra large), a key controller for cross talk between autophagy and apoptosis, shifting the balance toward apoptosis and leading to aggravated cardiac damage.


Assuntos
Androgênios/farmacologia , Apoptose/efeitos dos fármacos , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Caracteres Sexuais , Animais , Di-Hidrotestosterona/farmacologia , Regulação para Baixo/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Masculino , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Testosterona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA