Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Biochem Biophys Res Commun ; 546: 59-64, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33571905

RESUMO

Adenylate kinase2 (AK2) catalyzes trans-compartmental nucleotide exchange, but the functional implications of this mitochondrial intermembrane isoform is only partially understood. Here, transgenic AK2-/- null homozygosity was lethal early in embryo, indicating a mandatory role for intact AK2 in utero development. In the adult, conditional organ-specific ablation of AK2 precipitated abrupt heart failure with Krebs cycle and glycolytic metabolite buildup, suggesting a vital contribution to energy demanding cardiac performance. Depressed pump function recovered to pre-deletion levels overtime, suggestive of an adaptive response. Compensatory upregulation of phosphotransferase AK1, AK3, AK4 isozymes, creatine kinase isoforms, and hexokinase, along with remodeling of cell cycle/growth genes and mitochondrial ultrastructure supported organ rescue. Taken together, the requirement of AK2 in early embryonic stages, and the immediate collapse of heart performance in the AK2-deficient postnatal state underscore a primordial function of the AK2 isoform. Unsalvageable in embryo, loss of AK2 in the adult heart was recoverable, underscoring an AK2-integrated bioenergetics system with innate plasticity to maintain homeostasis on demand.


Assuntos
Adenilato Quinase/metabolismo , Desenvolvimento Embrionário , Homeostase , Miocárdio/enzimologia , Miocárdio/metabolismo , Adaptação Fisiológica , Adenilato Quinase/deficiência , Adenilato Quinase/genética , Animais , Ciclo do Ácido Cítrico , Perda do Embrião , Desenvolvimento Embrionário/genética , Metabolismo Energético , Feminino , Deleção de Genes , Genes Essenciais/genética , Glicólise , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Homeostase/genética , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos
2.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34768825

RESUMO

Heart failure pathobiology is permissive to reparative intent. Regenerative therapies exemplify an emerging disruptive innovation aimed at achieving structural and functional organ restitution. However, mixed outcomes, complexity in use, and unsustainable cost have curtailed broader adoption, mandating the development of novel cardio-regenerative approaches. Lineage guidance offers a standardized path to customize stem cell fitness for therapy. A case in point is the molecular induction of the cardiopoiesis program in adult stem cells to yield cardiopoietic cell derivatives designed for heart failure treatment. Tested in early and advanced clinical trials in patients with ischemic heart failure, clinical grade cardiopoietic cells were safe and revealed therapeutic improvement within a window of treatment intensity and pre-treatment disease severity. With the prospect of mass customization, cardiopoietic guidance has been streamlined from the demanding, recombinant protein cocktail-based to a protein-free, messenger RNA-based single gene protocol to engineer affordable cardiac repair competent cells. Clinical trial biobanked stem cells enabled a systems biology deconvolution of the cardiopoietic cell secretome linked to therapeutic benefit, exposing a paracrine mode of action. Collectively, this new knowledge informs next generation regenerative therapeutics manufactured as engineered cellular or secretome mimicking cell-free platforms. Launching biotherapeutics tailored for optimal outcome and offered at mass production cost would contribute to advancing equitable regenerative care that addresses population health needs.


Assuntos
Insuficiência Cardíaca/reabilitação , Insuficiência Cardíaca/terapia , Medicina Regenerativa/métodos , Células-Tronco Adultas/citologia , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Infarto do Miocárdio/terapia , Células-Tronco/citologia
3.
Biol Reprod ; 102(3): 607-619, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31621839

RESUMO

Heritable mitochondrial DNA (mtDNA) mutations are common, yet only a few recurring pathogenic mtDNA variants account for the majority of known familial cases in humans. Purifying selection in the female germline is thought to be responsible for the elimination of most harmful mtDNA mutations during oogenesis. Here we show that deleterious mtDNA mutations are abundant in ovulated mature mouse oocytes and preimplantation embryos recovered from PolG mutator females but not in their live offspring. This implies that purifying selection acts not in the maternal germline per se, but during post-implantation development. We further show that oocyte mtDNA mutations can be captured and stably maintained in embryonic stem cells and then reintroduced into chimeras, thereby allowing examination of the effects of specific mutations on fetal and postnatal development.


Assuntos
Blastocisto/metabolismo , DNA Mitocondrial/genética , Mutação , Oócitos/metabolismo , Animais , DNA Mitocondrial/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Oogênese/genética
4.
Am J Physiol Renal Physiol ; 315(5): F1493-F1499, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30019935

RESUMO

There is no therapy that promotes maturation and functionality of a dialysis arteriovenous fistula (AVF). The search for such therapies largely relies on evaluation of vascular responses and putative therapies in experimental AVFs. We studied an AVF in mice with chronic kidney disease (CKD). We demonstrate numerous stressors in the vein of the AVF-CKD group, including pathological shear, mitogenic, inflammatory, and hypoxia-reoxygenation stress. Because stress promotes premature senescence, we examined whether senescence is induced in the vein of the AVF-CKD model. We demonstrate a senescence phenotype in the AVF-CKD model, as indicated by increased expression of p16Ink4a, p21Cip1, and p53 and expected changes for certain senescence-associated microRNAs. RNA-sequencing analysis demonstrated differential expression of ~10,000 genes, including upregulation of proinflammatory and proliferative genes, in the vein of the AVF-CKD group. The vein in the AVF-CKD group exhibited telomere erosion and increased senescence-associated ß-galactosidase activity and staining. Senescence was induced in the artery of the AVF-CKD group and in the vein of the AVF without CKD. Finally, given the rapidly rising clinical interest in senolytics, we provide proof of concept of senolytics as a therapeutic approach by demonstrating that senolytics decrease p16Ink4a expression in the AVF-CKD model. This study introduces a novel concept underlying the basis for maturational and functional failure in human dialysis AVFs and identifies a new target for senolytic therapy.


Assuntos
Derivação Arteriovenosa Cirúrgica , Senescência Celular , Complicações Pós-Operatórias/patologia , Diálise Renal , Insuficiência Renal Crônica/terapia , Cauda/irrigação sanguínea , Remodelação Vascular , Veias/cirurgia , Animais , Derivação Arteriovenosa Cirúrgica/efeitos adversos , Velocidade do Fluxo Sanguíneo , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Dasatinibe/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Fenótipo , Complicações Pós-Operatórias/tratamento farmacológico , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/fisiopatologia , Quercetina/farmacologia , Fluxo Sanguíneo Regional , Estresse Mecânico , Grau de Desobstrução Vascular , Remodelação Vascular/efeitos dos fármacos , Remodelação Vascular/genética , Veias/metabolismo , Veias/patologia , Veias/fisiopatologia
5.
Arterioscler Thromb Vasc Biol ; 35(6): 1401-12, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25882068

RESUMO

OBJECTIVE: Neuropilin-1 (NRP-1) is a multidomain membrane receptor involved in angiogenesis and development of neuronal circuits, however, the role of NRP-1 in cardiovascular pathophysiology remains elusive. APPROACH AND RESULTS: In this study, we first observed that deletion of NRP-1 induced peroxisome proliferator-activated receptor γ coactivator 1α in cardiomyocytes and vascular smooth muscle cells, which was accompanied by dysregulated cardiac mitochondrial accumulation and induction of cardiac hypertrophy- and stress-related markers. To investigate the role of NRP-1 in vivo, we generated mice lacking Nrp-1 in cardiomyocytes and vascular smooth muscle cells (SM22-α-Nrp-1 KO), which exhibited decreased survival rates, developed cardiomyopathy, and aggravated ischemia-induced heart failure. Mechanistically, we found that NRP-1 specifically controls peroxisome proliferator-activated receptor γ coactivator 1 α and peroxisome proliferator-activated receptor γ in cardiomyocytes through crosstalk with Notch1 and Smad2 signaling pathways, respectively. Moreover, SM22-α-Nrp-1 KO mice exhibited impaired physical activities and altered metabolite levels in serum, liver, and adipose tissues, as demonstrated by global metabolic profiling analysis. CONCLUSIONS: Our findings provide new insights into the cardioprotective role of NRP-1 and its influence on global metabolism.


Assuntos
Cardiomiopatias/metabolismo , Insuficiência Cardíaca/metabolismo , Isquemia Miocárdica/metabolismo , Neuropilina-1/metabolismo , Animais , Homeostase , Camundongos Knockout , Proteínas dos Microfilamentos , Mitocôndrias Cardíacas/metabolismo , Proteínas Musculares , Músculo Liso Vascular/metabolismo , Miócitos Cardíacos/metabolismo , PPAR gama/metabolismo , Receptor Cross-Talk , Receptor Notch1/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Fatores de Transcrição/metabolismo
6.
Stem Cells Transl Med ; 13(2): 116-124, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38006196

RESUMO

Patients with heart failure experience limitations in daily activity and poor quality-of-life. Prospective surveillance of health-related quality-of-life supplemented traditional death and hospitalization outcomes in the multinational, randomized, double-blinded CHART-1 clinical trial that assessed cardiopoiesis-guided cell therapy in ischemic heart failure patients with reduced left ventricular ejection fraction. The Minnesota Living with Heart Failure Questionnaire (MLHFQ), a Food and Drug Administration qualified instrument for evaluating therapeutic effectiveness, was applied through the 1-year follow-up. Cell treated (n = 109) and sham procedure (n = 140) cohorts reported improved MLHFQ scores comparable between the 2 study arms (mean treatment difference with baseline adjustment -3.2 points, P = .107). Superiority of cell treatment over sham in betterment of the MLHFQ score was demonstrated in patients with pre-existing advanced left ventricular enlargement (baseline-adjusted mean treatment difference -6.4 points, P = .009). In this highly responsive subpopulation, benefit on the MLHFQ score paralleled reduction in death and hospitalization post-cell therapy (adjusted Mann-Whitney odds 1.43, 95% CI, 1.01-2.01; P = .039). The potential of cell therapy in addressing the quality-of-life dimension of heart failure requires further evaluation for disease relief.


Assuntos
Insuficiência Cardíaca , Função Ventricular Esquerda , Humanos , Volume Sistólico , Estudos Prospectivos , Insuficiência Cardíaca/terapia , Qualidade de Vida
7.
J Am Heart Assoc ; 13(9): e032172, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38700022

RESUMO

BACKGROUND: The purpose of this study was to investigate a therapeutic approach targeting the inflammatory response and consequent remodeling from ischemic myocardial injury. METHODS AND RESULTS: Coronary thrombus aspirates were collected from patients at the time of ST-segment-elevation myocardial infarction and subjected to array-based proteome analysis. Clinically indistinguishable at myocardial infarction (MI), patients were stratified into vulnerable and resilient on the basis of 1-year left ventricular ejection fraction and death. Network analysis from coronary aspirates revealed prioritization of tumor necrosis factor-α signaling in patients with worse clinical outcomes. Infliximab, a tumor necrosis factor-α inhibitor, was infused intravenously at reperfusion in a porcine MI model to assess whether infliximab-mediated immune modulation impacts post-MI injury. At 3 days after MI (n=7), infliximab infusion increased proregenerative M2 macrophages in the myocardial border zone as quantified by immunofluorescence (24.1%±23.3% in infliximab versus 9.29%±8.7% in sham; P<0.01). Concomitantly, immunoassays of coronary sinus samples quantified lower troponin I levels (41.72±7.34 pg/mL versus 58.11±10.75 pg/mL; P<0.05) and secreted protein analysis revealed upregulation of injury-modifying interleukin-2, -4, -10, -12, and -18 cytokines in the infliximab-treated cohort. At 4 weeks (n=12), infliximab treatment resulted in significant protective influence, improving left ventricular ejection fraction (53.9%±5.4% versus 36.2%±5.3%; P<0.001) and reducing scar size (8.31%±10.9% versus 17.41%±12.5%; P<0.05). CONCLUSIONS: Profiling of coronary thrombus aspirates in patients with ST-segment-elevation MI revealed highest association for tumor necrosis factor-α in injury risk. Infliximab-mediated immune modulation offers an actionable pathway to alter MI-induced inflammatory response, preserving contractility and limiting adverse structural remodeling.


Assuntos
Modelos Animais de Doenças , Infliximab , Remodelação Ventricular , Infliximab/uso terapêutico , Infliximab/farmacologia , Animais , Humanos , Masculino , Pessoa de Meia-Idade , Remodelação Ventricular/efeitos dos fármacos , Feminino , Infarto do Miocárdio com Supradesnível do Segmento ST/tratamento farmacológico , Infarto do Miocárdio com Supradesnível do Segmento ST/imunologia , Função Ventricular Esquerda/efeitos dos fármacos , Suínos , Idoso , Fator de Necrose Tumoral alfa/metabolismo , Volume Sistólico/efeitos dos fármacos , Trombose Coronária/prevenção & controle , Trombose Coronária/tratamento farmacológico , Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/imunologia , Troponina I/sangue , Troponina I/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo
8.
J Physiol ; 591(17): 4335-49, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23568891

RESUMO

Dyssynchronous myocardial motion aggravates cardiac pump function. Cardiac resynchronization using pacing devices is a standard-of-care in the management of heart failure. Post-infarction, however, scar tissue formation impedes the efficacy of device-based therapy. The present study tests a regenerative approach aimed at targeting the origin of abnormal motion to prevent dyssynchronous organ failure. Induced pluripotent stem (iPS) cells harbour a reparative potential, and were here bioengineered from somatic fibroblasts reprogrammed with the stemness factors OCT3/4, SOX2, KLF4, and c-MYC. In a murine infarction model, within 30 min of coronary ligation, iPS cells were delivered to mapped infarcted areas. Focal deformation and dysfunction underlying progressive heart failure was resolved prospectively using speckle-tracking imaging. Tracked at high temporal and spatial resolution, regional iPS cell transplantation restored, within 10 days post-infarction, the contractility of targeted infarcted foci and nullified conduction delay in adjacent non-infarcted regions. Local iPS cell therapy, but not delivery of parental fibroblasts or vehicle, prevented or normalized abnormal strain patterns correcting the decrease in peak strain, disparity of time-to-peak strain, and pathological systolic stretch. Focal benefit of iPS cell intervention translated into improved left ventricular conduction and contractility, reduced scar, and reversal of structural remodelling, protecting from organ decompensation. Thus, in ischaemic cardiomyopathy, targeted iPS cell transplantation synchronized failing ventricles, offering a regenerative strategy to achieve biological resynchronization.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Contração Miocárdica , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Função Ventricular , Animais , Terapia de Ressincronização Cardíaca , Reprogramação Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Ultrassonografia , Septo Interventricular/diagnóstico por imagem
9.
J Exp Med ; 204(2): 405-20, 2007 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-17283208

RESUMO

Embryonic stem cells have the distinct potential for tissue regeneration, including cardiac repair. Their propensity for multilineage differentiation carries, however, the liability of neoplastic growth, impeding therapeutic application. Here, the tumorigenic threat associated with embryonic stem cell transplantation was suppressed by cardiac-restricted transgenic expression of the reprogramming cytokine TNF-alpha, enhancing the cardiogenic competence of recipient heart. The in vivo aptitude of TNF-alpha to promote cardiac differentiation was recapitulated in embryoid bodies in vitro. The procardiogenic action required an intact endoderm and was mediated by secreted cardio-inductive signals. Resolved TNF-alpha-induced endoderm-derived factors, combined in a cocktail, secured guided differentiation of embryonic stem cells in monolayers produce cardiac progenitors termed cardiopoietic cells. Characterized by a down-regulation of oncogenic markers, up-regulation, and nuclear translocation of cardiac transcription factors, this predetermined population yielded functional cardiomyocyte progeny. Recruited cardiopoietic cells delivered in infarcted hearts generated cardiomyocytes that proliferated into scar tissue, integrating with host myocardium for tumor-free repair. Thus, cardiopoietic programming establishes a strategy to hone stem cell pluripotency, offering a tumor-resistant approach for regeneration.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Coração/fisiologia , Miócitos Cardíacos/citologia , Regeneração/fisiologia , Transplante de Células-Tronco/métodos , Fator de Necrose Tumoral alfa/metabolismo , Animais , Primers do DNA , Células-Tronco Embrionárias/transplante , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Neoplasias/prevenção & controle , Fatores de Transcrição/metabolismo
10.
Sci Rep ; 12(1): 7314, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35513538

RESUMO

Plasmalemmal ATP sensitive potassium (KATP) channels are recognized metabolic sensors, yet their cellular reach is less well understood. Here, transgenic Kir6.2 null hearts devoid of the KATP channel pore underwent multiomics surveillance and systems interrogation versus wildtype counterparts. Despite maintained organ performance, the knockout proteome deviated beyond a discrete loss of constitutive KATP channel subunits. Multidimensional nano-flow liquid chromatography tandem mass spectrometry resolved 111 differentially expressed proteins and their expanded network neighborhood, dominated by metabolic process engagement. Independent multimodal chemometric gas and liquid chromatography mass spectrometry unveiled differential expression of over one quarter of measured metabolites discriminating the Kir6.2 deficient heart metabolome. Supervised class analogy ranking and unsupervised enrichment analysis prioritized nicotinamide adenine dinucleotide (NAD+), affirmed by extensive overrepresentation of NAD+ associated circuitry. The remodeled metabolome and proteome revealed functional convergence and an integrated signature of disease susceptibility. Deciphered cardiac patterns were traceable in the corresponding plasma metabolome, with tissue concordant plasma changes offering surrogate metabolite markers of myocardial latent vulnerability. Thus, Kir6.2 deficit precipitates multiome reorganization, mapping a comprehensive atlas of the KATP channel dependent landscape.


Assuntos
NAD , Proteoma , Trifosfato de Adenosina , Coração , Canais KATP/genética , Canais KATP/metabolismo , NAD/metabolismo , Proteoma/metabolismo
11.
Am J Physiol Renal Physiol ; 301(4): F845-51, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21697243

RESUMO

The study of hemodynamic alterations following the creation of an arteriovenous fistula (AVF) is relevant to vascular adaptive responses and hemodialysis access dysfunction. This study examined such alterations in a murine AVF created by anastomosing the carotid artery to the jugular vein. AVF blood flow was markedly increased due to reduced AVF vascular resistance. Despite such markedly increased basal blood flow, AVF blood flow further increased in response to acetylcholine. This AVF model exhibited increased cardiac output and decreased systemic vascular resistance; the kidney, in contrast, exhibited decreased blood flow and increased vascular resistance. Augmentation in AVF blood flow was attended by increased arterial heme oxygenase-1 (HO-1) mRNA and protein expression, the latter localized to smooth muscle cells of the AVF artery; AVF blood flow was substantially reduced in HO-1(-/-) mice compared with HO-1(+/+) mice. Finally, in a murine model of a representative disease known to exhibit impaired hemodynamic responses (sickle cell disease), the creation of an AVF was attended by decreased AVF flow and impaired AVF function. We conclude that this AVF model exhibits markedly increased AVF blood flow, a vasodilatory reserve capacity, increased cardiac output, decreased renal blood flow, and a dependency on intact hemodynamic responses, in general, and HO-1 expression, in particular, in achieving and maintaining AVF blood flow. We suggest that these findings support the utility of this model in investigating the basis for and the consequences of hemodynamic stress, including shear stress, and the pathobiology of hemodialysis AVF dysfunction.


Assuntos
Fístula Arteriovenosa/fisiopatologia , Hemodinâmica/fisiologia , Acetilcolina/farmacologia , Anemia Falciforme/fisiopatologia , Animais , Fístula Arteriovenosa/enzimologia , Débito Cardíaco/efeitos dos fármacos , Débito Cardíaco/fisiologia , Artérias Carótidas , Modelos Animais de Doenças , Heme Oxigenase-1/biossíntese , Hemodinâmica/efeitos dos fármacos , Veias Jugulares , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/fisiopatologia , Fluxo Sanguíneo Regional , Resistência Vascular/efeitos dos fármacos , Resistência Vascular/fisiologia
12.
Stem Cells ; 28(8): 1355-67, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20572010

RESUMO

Transplantation of pluripotent stem cells has proven beneficial in heart failure, yet the proteomic landscape underlying repair remains largely uncharacterized. In a genetic model of dilated cardiomyopathy elicited by pressure overload in the KCNJ11 (potassium inwardly rectifying channel, subfamily J, member 11) null mutant, proteome-wide profiles were here resolved by means of a systems approach prior to and following disease manifestation in the absence or presence of embryonic stem cell treatment. Comparative two-dimensional gel electrophoresis revealed a unique cardiomyopathic proteome in the absence of therapy, remodeled in response to stem cell treatment. Specifically, linear ion trap quadrupole-Orbitrap mass spectrometry determined the identities of 93 and 109 differentially expressed proteins from treated and untreated cardiomyopathic hearts, respectively. Mapped protein-protein relationships and corresponding neighborhoods incorporated the stem cell-dependent subproteome into a nonstochastic network with divergent composition from the stem cell-independent counterpart. Stem cell intervention produced a distinct proteome signature across a spectrum of biological processes ranging from energetic metabolism, oxidoreductases, and stress-related chaperones to processes supporting protein synthesis/degradation, signaling, and transport regulation, cell structure and scaffolding. In the absence of treatment, bioinformatic interrogation of the disease-only proteome network prioritized adverse cardiac outcomes, ablated or ameliorated following stem cell transplantation. Functional and structural measurements validated improved myocardial contractile performance, reduced ventricular size and decreased cardiac damage in the treated cohort. Unbiased systems assessment unmasked "cardiovascular development" as a prioritized biological function in stem cell-reconstructed cardiomyopathic hearts. Thus, embryonic stem cell treatment transformed the cardiomyopathic proteome to demote disease-associated adverse effects and sustain a procardiogenic developmental response, supplying a regenerative substrate for heart failure repair.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/terapia , Células-Tronco Embrionárias/fisiologia , Canais KATP/genética , Animais , Cardiomiopatia Dilatada/genética , Linhagem Celular , Eletroforese em Gel Bidimensional , Células-Tronco Embrionárias/citologia , Feminino , Canais KATP/deficiência , Masculino , Camundongos , Transplante de Células-Tronco , Espectrometria de Massas em Tandem
13.
Circ Res ; 105(7): 648-56, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19696409

RESUMO

RATIONALE: Induced pluripotent stem cells (iPS) allow derivation of pluripotent progenitors from somatic sources. Originally, iPS were induced by a stemness-related gene set that included the c-MYC oncogene. OBJECTIVE: Here, we determined from embryo to adult the cardiogenic proficiency of iPS programmed without c-MYC, a cardiogenicity-associated transcription factor. METHODS AND RESULTS: Transgenic expression of 3 human stemness factors SOX2, OCT4, and KLF4 here reset murine fibroblasts to the pluripotent ground state. Transduction without c-MYC reversed cellular ultrastructure into a primitive archetype and induced stem cell markers generating 3-germ layers, all qualifiers of acquired pluripotency. Three-factor induced iPS (3F-iPS) clones reproducibly demonstrated cardiac differentiation properties characterized by vigorous beating activity of embryoid bodies and robust expression of cardiac Mef2c, alpha-actinin, connexin43, MLC2a, and troponin I. In vitro isolated iPS-derived cardiomyocytes demonstrated functional excitation-contraction coupling. Chimerism with 3F-iPS derived by morula-stage diploid aggregation was sustained during prenatal heart organogenesis and contributed in vivo to normal cardiac structure and overall performance in adult tumor-free offspring. CONCLUSIONS: Thus, 3F-iPS bioengineered without c-MYC achieve highest stringency criteria for bona fide cardiogenesis enabling reprogrammed fibroblasts to yield de novo heart tissue compatible with native counterpart throughout embryological development and into adulthood.


Assuntos
Transdiferenciação Celular , Fibroblastos/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Actinina/metabolismo , Potenciais de Ação , Animais , Sinalização do Cálcio , Linhagem da Célula , Transdiferenciação Celular/genética , Células Cultivadas , Quimerismo , Conexina 43/metabolismo , Técnicas de Cultura Embrionária , Feminino , Fibroblastos/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição MEF2 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Contração Miocárdica/genética , Miócitos Cardíacos/ultraestrutura , Fatores de Regulação Miogênica/metabolismo , Cadeias Leves de Miosina/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Organogênese , Células-Tronco Pluripotentes/ultraestrutura , Gravidez , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Engenharia Tecidual/métodos , Transdução Genética , Troponina I/metabolismo
14.
NPJ Regen Med ; 6(1): 57, 2021 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-34556664

RESUMO

Life expectancy has increased by three decades since the mid-twentieth century. Parallel healthspan expansion has however not followed, largely impeded by the pandemic of chronic diseases afflicting a growing older population. The lag in quality of life is a recognized challenge that calls for prioritization of disease-free longevity. Contemporary communal, clinical and research trends aspiring to extend the health horizon are here outlined in the context of an evolving epidemiology. A shared action integrating public and societal endeavors with emerging interventions that target age-related multimorbidity and frailty is needed. A multidimensional buildout of a curative perspective, boosted by modern anti-senescent and regenerative technology with augmented decision making, would require dedicated resources and cost-effective validation to responsibly bridge the healthspan-lifespan gap for a future of equitable global wellbeing.

15.
Regen Med ; 16(3): 309-322, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33622049

RESUMO

Regenerative medicine, poised to transform 21st century healthcare, has aspired to enrich care options by bringing cures to patients in need. Science-driven responsible and regulated translation of innovative technology has enabled the launch of previously unimaginable care pathways adopted prudently for select serious diseases and disabilities. The collective resolve to advance the design, manufacture and validity of affordable regenerative solutions aims to democratize such health benefits for all. The objective of this Review is to outline the framework and prerequisites that underpin clinical readiness of regenerative care. Integrated research and development, specialized workforce education and accessible evidence-based practice implementation are at the core of realizing an equitable regenerative medicine vision.


Assuntos
Atenção à Saúde , Medicina Regenerativa , Humanos , Recursos Humanos
16.
Biomark Med ; 15(10): 775-783, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34169733

RESUMO

Risk of outcome variability challenges therapeutic innovation. Selection of the most suitable candidates is predicated on reliable response indicators. Especially for emergent regenerative biotherapies, determinants separating success from failure in achieving disease rescue remain largely unknown. Accordingly, (pre)clinical development programs have placed increased emphasis on the multi-dimensional decoding of repair capacity and disease resolution, attributes defining responsiveness. To attain regenerative goals for each individual, phenotype-based patient selection is poised for an upgrade guided by new insights into disease biology, translated into refined surveillance of response regulators and deep learning-amplified clinical decision support.


Assuntos
Insuficiência Cardíaca
17.
Stem Cells Transl Med ; 10(3): 385-397, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33098750

RESUMO

To optimize the regenerative proficiency of stem cells, a cardiopoietic protein-based cocktail consisting of multiple growth factors has been developed and advanced into clinical trials for treatment of ischemic heart failure. Streamlining the inductors of cardiopoiesis would address the resource intensive nature of the current stem cell enhancement protocol. To this end, the microencapsulated-modified-mRNA (M3 RNA) technique was here applied to introduce early cardiogenic genes into human adipose-derived mesenchymal stem cells (AMSCs). A single mesodermal transcription factor, Brachyury, was sufficient to trigger high expression of cardiopoietic markers, Nkx2.5 and Mef2c. Engineered cardiopoietic stem cells (eCP) featured a transcriptome profile distinct from pre-engineered AMSCs. In vitro, eCP demonstrated protective antioxidant capacity with enhanced superoxide dismutase expression and activity; a vasculogenic secretome driving angiogenic tube formation; and macrophage polarizing immunomodulatory properties. In vivo, in a murine model of myocardial infarction, intramyocardial delivery of eCP (600 000 cells per heart) improved cardiac performance and protected against decompensated heart failure. Thus, heart repair competent stem cells, armed with antioxidant, vasculogenic, and immunomodulatory traits, are here engineered through a protein-independent single gene manipulation, expanding the available regenerative toolkit.


Assuntos
Proteínas Fetais , Insuficiência Cardíaca , Transplante de Células-Tronco Mesenquimais , Células-Tronco , Proteínas com Domínio T , Tecido Adiposo/citologia , Animais , Proteínas Fetais/genética , Insuficiência Cardíaca/terapia , Humanos , Camundongos , Infarto do Miocárdio/terapia , Secretoma , Proteínas com Domínio T/genética , Transcriptoma
18.
Stem Cells Transl Med ; 10(9): 1320-1328, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34047493

RESUMO

Stem cell paracrine activity is implicated in cardiac repair. Linkage between secretome functionality and therapeutic outcome was here interrogated by systems analytics of biobanked human cardiopoietic cells, a regenerative biologic in advanced clinical trials. Protein chip array identified 155 proteins differentially secreted by cardiopoietic cells with clinical benefit, expanded into a 520 node network, collectively revealing inherent vasculogenic properties along with cardiac and smooth muscle differentiation and development. Next generation RNA sequencing, refined by pathway analysis, pinpointed miR-146 dependent regulation upstream of the decoded secretome. Intracellular and extracellular integration unmasked commonality across cardio-vasculogenic processes. Mirroring the secretome pattern, infarcted hearts benefiting from cardiopoietic cell therapy restored the disease proteome engaging cardiovascular system functions. The cardiopoietic cell secretome thus confers a therapeutic molecular imprint on recipient hearts, with response informed by predictive systems profiling.


Assuntos
Infarto do Miocárdio , Proteoma , Coração/fisiologia , Humanos , Infarto do Miocárdio/terapia , Secretoma , Células-Tronco
19.
Circulation ; 120(5): 408-16, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19620500

RESUMO

BACKGROUND: Nuclear reprogramming provides an emerging strategy to produce embryo-independent pluripotent stem cells from somatic tissue. Induced pluripotent stem cells (iPS) demonstrate aptitude for de novo cardiac differentiation, yet their potential for heart disease therapy has not been tested. METHODS AND RESULTS: In this study, fibroblasts transduced with human stemness factors OCT3/4, SOX2, KLF4, and c-MYC converted into an embryonic stem cell-like phenotype and demonstrated the ability to spontaneously assimilate into preimplantation host morula via diploid aggregation, unique to bona fide pluripotent cells. In utero, iPS-derived chimera executed differentiation programs to construct normal heart parenchyma patterning. Within infarcted hearts in the adult, intramyocardial delivery of iPS yielded progeny that properly engrafted without disrupting cytoarchitecture in immunocompetent recipients. In contrast to parental nonreparative fibroblasts, iPS treatment restored postischemic contractile performance, ventricular wall thickness, and electric stability while achieving in situ regeneration of cardiac, smooth muscle, and endothelial tissue. CONCLUSIONS: Fibroblasts reprogrammed by human stemness factors thus acquire the potential to repair acute myocardial infarction, establishing iPS in the treatment of heart disease.


Assuntos
Células-Tronco Adultas/citologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco/métodos , Adulto , Células-Tronco Adultas/fisiologia , Animais , Desdiferenciação Celular/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Genes myc , Vetores Genéticos , Coração/embriologia , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco Pluripotentes/fisiologia , Recuperação de Função Fisiológica , Fatores de Transcrição SOXB1/genética , Transdução Genética
20.
Stem Cells ; 27(7): 1697-705, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19544428

RESUMO

Stem cells are an emerging strategy for treatment of myocardial infarction, limited however to postinjury intervention. Preventive stem cell-based therapy to augment stress tolerance has yet to be considered for lifelong protection. Here, pluripotent stem cells were microsurgically introduced at the blastocyst stage of murine embryo development to ensure stochastic integration and sustained organ contribution. Engineered chimera displayed excess in body weight due to increased fat deposits, but were otherwise devoid of obesity-related morbidity. Remarkably, and in sharp contrast to susceptible nonchimeric offspring, chimera was resistant to myocardial infarction induced by permanent coronary occlusion. Infarcted nonchimeric adult hearts demonstrated progressive deterioration in ejection fraction, while age-matched 12-14-months-old chimera recovered from equivalent ischemic insult to regain within one-month preocclusion contractile performance. Electrical remodeling and ventricular enlargement with fibrosis, prominent in failing nonchimera, were averted in the chimeric cohort characterized by an increased stem cell load in adipose tissue and upregulated markers of biogenesis Ki67, c-Kit, and stem cell antigen-1 in the myocardium. Favorable outcome in infarcted chimera translated into an overall benefit in workload capacity and survival. Thus, prenatal stem cell transplant yields a cardioprotective phenotype in adulthood, expanding cell-based indications beyond traditional postinjury applications to include pre-emptive therapy.


Assuntos
Blastocisto/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Infarto do Miocárdio/prevenção & controle , Transplante de Células-Tronco/métodos , Animais , Blastocisto/fisiologia , Blastômeros/citologia , Blastômeros/fisiologia , Embrião de Mamíferos , Masculino , Camundongos , Obesidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA