Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Med ; 10(14)2021 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-34300226

RESUMO

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a progressive heart condition which causes fibro-fatty myocardial scarring, ventricular arrhythmias, and sudden cardiac death. Most cases of ARVC can be linked to pathogenic mutations in the cardiac desmosome, but the pathophysiology is not well understood, particularly in early phases when arrhythmias can develop prior to structural changes. Here, we created a novel human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) model of ARVC from a patient with a c.2358delA variant in desmoglein-2 (DSG2). These DSG2-mutant (DSG2Mut) hiPSC-CMs were compared against two wildtype hiPSC-CM lines via immunostaining, RT-qPCR, Western blot, RNA-Seq, cytokine expression and optical mapping. Mutant cells expressed reduced DSG2 mRNA and had altered localization of desmoglein-2 protein alongside thinner, more disorganized myofibrils. No major changes in other desmosomal proteins were noted. There was increased pro-inflammatory cytokine expression that may be linked to canonical and non-canonical NFκB signaling. Action potentials in DSG2Mut CMs were shorter with increased upstroke heterogeneity, while time-to-peak calcium and calcium decay rate were reduced. These were accompanied by changes in ion channel and calcium handling gene expression. Lastly, suppressing DSG2 in control lines via siRNA allowed partial recapitulation of electrical anomalies noted in DSG2Mut cells. In conclusion, the aberrant cytoskeletal organization, cytokine expression, and electrophysiology found DSG2Mut hiPSC-CMs could underlie early mechanisms of disease manifestation in ARVC patients.

4.
Tissue Eng Part C Methods ; 26(2): 80-90, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31830863

RESUMO

Introduction: A key obstacle in the creation of engineered cardiac tissues of clinically relevant sizes is limited diffusion of oxygen and nutrients. Thus, there is a need for organized vascularization within a three-dimensional (3D) tissue environment. Human induced pluripotent stem cell (hiPSC)-derived early vascular cells (EVCs) have shown to improve organization of vascular networks within hydrogels. We hypothesize that introduction of EVCs into 3D microtissue spheroids will lead to increased microvascular formation and improve spheroid formation. Methods: HiPSC-derived cardiomyocytes (CMs) were cocultured with human adult ventricular cardiac fibroblasts (FB) and either human umbilical vein endothelial cells (HUVECs) or hiPSC-derived EVCs for 72 h to form mixed cell spheroids. Three different groups of cell ratios were tested: Group 1 (control) consisted of CM:FB:HUVEC 70:15:15, Group 2 consisted of CM:FB:EVC 70:15:15, and Group 3 consisted of CM:FB:EVC 40:15:45. Vascularization, cell distribution, and cardiac function were investigated. Results: Improved microvasculature was found in EVC spheroids with new morphologies of endothelial organization not found in Group 1 spheroids. CMs were found in a core-shell type distribution in Group 1 spheroids, but more uniformly distributed in EVC spheroids. Contraction rate increased into Group 2 spheroids compared to Group 1 spheroids. Conclusion: The triculture of CM, FB, and EVC within a multicellular cardiac spheroid promotes microvascular formation and cardiac spheroid contraction.


Assuntos
Fibroblastos/citologia , Hidrogéis/química , Células-Tronco Pluripotentes Induzidas/citologia , Contração Miocárdica , Miócitos Cardíacos/citologia , Neovascularização Fisiológica , Técnicas de Cocultura , Humanos , Esferoides Celulares
5.
Circulation ; 140(18): 1491-1505, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31533459

RESUMO

BACKGROUND: Inflammation is a prominent feature of arrhythmogenic cardiomyopathy (ACM), but whether it contributes to the disease phenotype is not known. METHODS: To define the role of inflammation in the pathogenesis of ACM, we characterized nuclear factor-κB signaling in ACM models in vitro and in vivo and in cardiac myocytes from patient induced pluripotent stem cells. RESULTS: Activation of nuclear factor-κB signaling, indicated by increased expression and nuclear accumulation of phospho-RelA/p65, occurred in both an in vitro model of ACM (expression of JUP2157del2 in neonatal rat ventricular myocytes) and a robust murine model of ACM (homozygous knock-in of mutant desmoglein-2 [Dsg2mut/mut]) that recapitulates the cardiac manifestations seen in patients with ACM. Bay 11-7082, a small-molecule inhibitor of nuclear factor-κB signaling, prevented the development of ACM disease features in vitro (abnormal redistribution of intercalated disk proteins, myocyte apoptosis, release of inflammatory cytokines) and in vivo (myocardial necrosis and fibrosis, left ventricular contractile dysfunction, electrocardiographic abnormalities). Hearts of Dsg2mut/mut mice expressed markedly increased levels of inflammatory cytokines and chemotactic molecules that were attenuated by Bay 11-7082. Salutary effects of Bay 11-7082 correlated with the extent to which production of selected cytokines had been blocked. Nuclear factor-κB signaling was also activated in cardiac myocytes derived from a patient with ACM. These cells produced and secreted abundant inflammatory cytokines under basal conditions, and this was also greatly reduced by Bay 11-7082. CONCLUSIONS: Inflammatory signaling is activated in ACM and drives key features of the disease. Targeting inflammatory pathways may be an effective new mechanism-based therapy for ACM.


Assuntos
Arritmias Cardíacas/metabolismo , Cardiomiopatias/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Animais , Arritmias Cardíacas/patologia , Cardiomiopatias/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Ratos Transgênicos , Ratos Wistar , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia
6.
Stem Cell Reports ; 12(5): 982-995, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31056480

RESUMO

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) hold great promise for cardiac studies, but their structural and functional immaturity precludes their use as faithful models of adult myocardium. Here we describe engineered heart slices (EHS), preparations of decellularized porcine myocardium repopulated with hiPSC-CMs that exhibit structural and functional improvements over standard culture. EHS exhibited multicellular, aligned bundles of elongated CMs with organized sarcomeres, positive inotropic responses to isoproterenol, anisotropic conduction of action potentials, and electrophysiological functionality for more than 200 days. We developed a new drug assay, GRIDS, that serves as a "fingerprint" of cardiac drug sensitivity for a range of pacing rates and drug concentrations. GRIDS maps characterized differences in drug sensitivity between EHS and monolayers more clearly than changes in action potential durations or conduction velocities. EHS represent a tissue-like model for long-term culture, structural, and functional improvement, and higher fidelity drug response of hiPSC-CMs.


Assuntos
Coração/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Cardiotônicos/farmacologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células Cultivadas , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Isoproterenol/farmacologia , Suínos
8.
Tissue Eng Part A ; 25(9-10): 725-735, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30520705

RESUMO

IMPACT STATEMENT: Genetic heart diseases such as arrhythmogenic cardiomyopathy (AC), a common genetic cause of sudden cardiac death, can be modeled using patient-specific induced pluripotent stem cell-derived cardiac myocytes (CMs). However, it is important to culture these cells in a multicellular syncytium with exposure to surrounding matrix cues to create more accurate and robust models of the disease due to the importance of cell-cell and cell-matrix interactions. The engineered heart slice, constructed by seeding CMs on intact decellularized matrix slices, allows molecular and functional studies on an aligned multilayered syncytium of CMs. This study reveals the potential for an improved disease-in-a-dish model of AC.


Assuntos
Arritmias Cardíacas , Cardiomiopatias , Células-Tronco Pluripotentes Induzidas , Modelos Cardiovasculares , Mutação , Miocárdio , Placofilinas , Engenharia Tecidual , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Placofilinas/genética , Placofilinas/metabolismo , Suínos
9.
Ann Emerg Med ; 72(2): 156-165, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29887191

RESUMO

STUDY OBJECTIVE: We analyzed the effect of insurance expansion on emergency department (ED) utilization among the uninsured in Maryland, which expanded Medicaid eligibility and created health insurance exchanges in 2014. METHODS: This was a retrospective analysis of statewide administrative claims for July 2012 to December 2015. We used coarsened exact matching to pair uninsured and insured (Medicaid, Medicare, commercial, and other) adult Maryland residents who visited an ED or were hospitalized at baseline (July 2012 to December 2013). We compared ED utilization between these groups after insurance expansion (January 2014 to December 2015), using a difference-in-differences quasi-experimental design. Nonreturning patients from the baseline period were included in the post-insurance expansion rates as having zero visits. RESULTS: Matching yielded 178,381 pairs. In the 12 months before insurance expansion, the baseline uninsured group visited the ED at a rate of 26.1 per 100 patient-quarters versus 28.2 among the insured group (relative rate=0.93). In the 24 months after insurance expansion, 45% of the baseline uninsured returned to an ED, of whom 33% returned uninsured, 40% returned with Medicaid, and 21% returned with commercial insurance. After insurance expansion, with 55% of patients in each group not returning, the ED visit rate for both the baseline uninsured and insured groups was 15.9 per 100 patient-quarters (relative rate=1.00). This 8% relative increase from baseline in ED visits among the uninsured group was driven primarily by increases in higher-acuity visits. Uninsured patients from high-poverty zip codes (N=34,964 pairs) increased their ED utilization by 15% after insurance expansion, whereas baseline uninsured patients with no comorbidities (N=94,330 pairs) showed a 3% decrease. CONCLUSION: Insurance expansion in Maryland was associated with a modest relative increase in ED visits among the uninsured, driven by increases in higher-acuity visits. It remains unclear whether insurance coverage helped the uninsured address their unmet medical needs.


Assuntos
Cuidados Críticos/estatística & dados numéricos , Hospitalização/estatística & dados numéricos , Pessoas sem Cobertura de Seguro de Saúde , Adulto , Idoso , Serviço Hospitalar de Emergência , Feminino , Humanos , Masculino , Maryland/epidemiologia , Medicare , Pessoa de Meia-Idade , Patient Protection and Affordable Care Act , Áreas de Pobreza , Estudos Retrospectivos , Estados Unidos , Adulto Jovem
10.
Biochem Biophys Res Commun ; 473(3): 733-42, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26427871

RESUMO

Proper blood vessel networks are necessary for constructing and re-constructing tissues, promoting wound healing, and delivering metabolic necessities throughout the body. Conversely, an understanding of vascular dysfunction has provided insight into the pathogenesis and progression of diseases both common and rare. Recent advances in stem cell-based regenerative medicine - including advances in stem cell technologies and related progress in bioscaffold design and complex tissue engineering - have allowed rapid advances in the field of vascular biology, leading in turn to more advanced modeling of vascular pathophysiology and improved engineering of vascularized tissue constructs. In this review we examine recent advances in the field of stem cell-derived vasculature, providing an overview of stem cell technologies as a source for vascular cell types and then focusing on their use in three primary areas: studies of vascular development and angiogenesis, improved disease modeling, and the engineering of vascularized constructs for tissue-level modeling and cell-based therapies.


Assuntos
Materiais Biocompatíveis/química , Vasos Sanguíneos/citologia , Imageamento Tridimensional/métodos , Células-Tronco/citologia , Engenharia Tecidual/métodos , Animais , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Microcirculação , Músculo Liso Vascular/citologia , Alicerces Teciduais , Cicatrização/fisiologia
11.
Chest ; 147(3): 824-834, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25732448

RESUMO

Stem cell research and related initiatives in regenerative medicine, cell-based therapy, and tissue engineering have generated considerable scientific and public interest. Researchers are applying stem cell technologies to chest medicine in a variety of ways: using stem cells as models for drug discovery, testing stem cell-based therapies for conditions as diverse as COPD and cystic fibrosis, and producing functional lung and tracheal tissue for physiologic modeling and potential transplantation. Although significant scientific obstacles remain, it is likely that stem cell-based regenerative medicine will have a significant clinical impact in chest medicine. However, stem cell research has also generated substantial controversy, posing a variety of ethical and regulatory challenges for research and clinical practice. Some of the most prominent ethical questions related to the use of stem cell technologies in chest medicine include (1) implications for donors, (2) scientific prerequisites for clinical testing and use, (3) stem cell tourism, (4) innovation and clinical use of emerging stem cell-based interventions, (5) responsible translation of stem cell-based therapies to clinical use, and (6) appropriate and equitable access to emerging therapies. Having a sense of these issues should help to put emerging scientific advances into appropriate context and to ensure the responsible clinical translation of promising therapeutics.


Assuntos
Pneumopatias/terapia , Formulação de Políticas , Pesquisa com Células-Tronco/ética , Humanos , Medicina Regenerativa/ética , Transplante de Células-Tronco/ética , Engenharia Tecidual/ética
12.
Nanomedicine ; 11(4): 1019-28, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25652896

RESUMO

Polymeric nanoparticles (nano-paAPCs) modified with T-cell antigens and encapsulating immunostimulatory or immunoinhibitory factors may act as artificial antigen-presenting cells to circulating immune cells, improving the selective delivery of encapsulated drug or cytokine to antigen-specific T-cells. Paracrine delivery of encapsulated agents from these nanoparticles to adjacent cells facilitate sustained delivery lowering the overall administered dose, thus enhancing the overall drug efficacy while reducing toxicity of pleiotropic factors. Little is known mathematically regarding the local concentration of released agent that accumulates around a nanoparticle that is near or embeds in a cell. These concentration fields are calculated here in an attempt to understand paracrine efficacy of these nano-paAPC systems. The significant factor accumulation that can occur if the particles were to embed in the cell membrane may explain observed experimental data regarding enhanced T-cell activation and nanoparticle-mediated improvement in the drug delivery process to non-internalizing cellular targets. FROM THE CLINICAL EDITOR: In this interesting article, the authors utilized nanosized polymeric artificial presenting cells (nano-paAPC) that released cytokine to study the effects after interaction with T cells. It was found that nano-paAPC were able to embed into cell membrane, with subsequent enhanced T-cell activation. The findings provide further understanding of immune cell interaction and are considered to be important for designing nanoparticles engineered to deliver cytokines or immumodulatory factors to specific immune cells.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Modelos Imunológicos , Nanopartículas , Linfócitos T/imunologia , Animais , Humanos
13.
Stem Cells Transl Med ; 2(10): 727-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23990574

RESUMO

Human somatic cell reprogramming is a leading technology for accelerating disease modeling and drug discovery. Research organizations are sponsoring initiatives to create libraries of induced pluripotent stem cell (iPSC) lines for broad distribution and application. Donor informed consent plays a critical role in supporting the ethical conduct of iPSC research. To date, our organizations have focused on informed consent considerations for somatic cell collection intended specifically for iPSC derivation and distribution. This article considers how somatic cells obtained under general (biomedical) research protocols can be used for iPSC derivation. We present draft Points to Consider regarding the use of human somatic cells for iPSC research. Our goal is to initiate a process designed to develop consensus for the use of previously collected specimens for iPSC research. We anticipate publishing final considerations in early 2014.


Assuntos
Bancos de Espécimes Biológicos/ética , Células-Tronco Pluripotentes Induzidas , Consentimento Livre e Esclarecido , Linhagem Celular , Humanos
14.
Stem Cells Transl Med ; 1(5): 409-21, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-23197820

RESUMO

Induced pluripotent stem cells (iPSCs) have elicited excitement in both the scientific and ethics communities for their potential to advance basic and translational research. They have been hailed as an alternative to derivation from embryos that provides a virtually unlimited source of pluripotent stem cells for research and therapeutic applications. However, research with iPSCs is ethically complex, uniquely encompassing the concerns associated with genomics, immortalized cell lines, transplantation, human reproduction, and biobanking. Prospective donation of tissue specimens for iPSC research thus requires an approach to informed consent that is constructed for this context. Even in the nascent stages of this field, approaches to informed consent have been variable in ways that threaten the simultaneous goals of protecting donors and safeguarding future research and translation, and investigators are seeking guidance. We address this need by providing concrete recommendations for informed consent that balance the perspectives of a variety of stakeholders. Our work combines analysis of consent form language collected from investigators worldwide with a conceptual balancing of normative ethical concerns, policy precedents, and scientific realities. Our framework asks people to consent prospectively to a broad umbrella of foreseeable research, including future therapeutic applications, with recontact possible in limited circumstances. We argue that the long-term goals of regenerative medicine, interest in sharing iPSC lines, and uncertain landscape of future research all would be served by a framework of ongoing communication with donors. Our approach balances the goals of iPSC and regenerative medicine researchers with the interests of individual research participants.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Consentimento Livre e Esclarecido , Medicina Regenerativa/ética , Manejo de Espécimes/normas , Pesquisa com Células-Tronco/ética , Humanos , Guias de Prática Clínica como Assunto , Medicina Regenerativa/legislação & jurisprudência , Pesquisa com Células-Tronco/legislação & jurisprudência
16.
Biomacromolecules ; 12(10): 3469-77, 2011 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-21854075

RESUMO

Amino acid-based core cross-linked star (CCS) polymers (poly(L-lysine)(arm)poly(L-cystine)(core)) with peripheral allyl functionalities were synthesized by sequential ring-opening polymerization (ROP) of amino acid N-carboxyanhydrides (NCAs) via the arm-first approach, using N-(trimethylsilyl)allylamine as the initiator. Subsequent functionalization with a poly(ethylene glycol) (PEG)-folic acid conjugate via thiol-ene click chemistry afforded poly(PEG-b-L-lysine)(arm)poly(L-cystine)(core) stars with outer PEG coronas decorated with folic acid targeting moieties. Similarly, a control was prepared without folic acid, using just PEG. A fluorophore was used to track both star polymers incubated with breast cancer cells (MDA-MB-231) in vitro. Confocal microscopy and flow cytometry revealed that the stars could be internalized into the cells, and higher cell internalization was observed when folic acid moieties were present. Cytotoxicity studies indicate that both stars are nontoxic to MDA-MB-231 cells at concentrations of up to 50 µg/mL. These results make this amino acid-based star polymer an attractive candidate in targeted drug delivery applications including chemotherapy.


Assuntos
Materiais Biocompatíveis/síntese química , Química Click/métodos , Cistina/química , Portadores de Fármacos/síntese química , Ácido Fólico/química , Polilisina/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Materiais Biocompatíveis/análise , Materiais Biocompatíveis/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/análise , Portadores de Fármacos/farmacologia , Feminino , Citometria de Fluxo , Corantes Fluorescentes/análise , Humanos , Micelas , Microscopia Confocal , Polietilenoglicóis/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...