Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Cardiovasc Res ; 118(11): 2428-2436, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34387303

RESUMO

Exogenous cell-based therapy has emerged as a promising new strategy to facilitate repair of hearts damaged by acute or chronic injury. However, the field of cell-based therapy is handicapped by the lack of standardized definitions and terminology, making comparisons across studies challenging. Even the term 'stem cell therapy' is misleading because only a small percentage of cells derived from adult bone marrow, peripheral blood, or adipose tissue meets the accepted haematopoietic or developmental definition of stem cells. Furthermore, cells (stem or otherwise) are dynamic biological products, meaning that their surface-marker expression, phenotypic and functional characteristics, and the products they secrete in response to their microenvironment can change. It is also important to point out that most surface markers are seldom specific for a cell type. In this article, we discuss the lack of consistency in the descriptive terminology used in cell-based therapies and offer guidelines aimed at standardizing nomenclature and definitions to improve communication among investigators and the general public.


Assuntos
Tecido Adiposo , Terapia Baseada em Transplante de Células e Tecidos , Adulto , Humanos , Pulmão , Transplante de Células-Tronco
2.
Eur J Heart Fail ; 23(4): 661-674, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33811444

RESUMO

AIMS: CONCERT-HF is an NHLBI-sponsored, double-blind, placebo-controlled, Phase II trial designed to determine whether treatment with autologous bone marrow-derived mesenchymal stromal cells (MSCs) and c-kit positive cardiac cells (CPCs), given alone or in combination, is feasible, safe, and beneficial in patients with heart failure (HF) caused by ischaemic cardiomyopathy. METHODS AND RESULTS: Patients were randomized (1:1:1:1) to transendocardial injection of MSCs combined with CPCs, MSCs alone, CPCs alone, or placebo, and followed for 12 months. Seven centres enrolled 125 participants with left ventricular ejection fraction of 28.6 ± 6.1% and scar size 19.4 ± 5.8%, in New York Heart Association class II or III. The proportion of major adverse cardiac events (MACE) was significantly decreased by CPCs alone (-22% vs. placebo, P = 0.043). Quality of life (Minnesota Living with Heart Failure Questionnaire score) was significantly improved by MSCs alone (P = 0.050) and MSCs + CPCs (P = 0.023) vs. placebo. Left ventricular ejection fraction, left ventricular volumes, scar size, 6-min walking distance, and peak oxygen consumption did not differ significantly among groups. CONCLUSIONS: This is the first multicentre trial assessing CPCs and a combination of two cell types from different tissues in HF patients. The results show that treatment is safe and feasible. Even with maximal guideline-directed therapy, both CPCs and MSCs were associated with improved clinical outcomes (MACE and quality of life, respectively) in ischaemic HF without affecting left ventricular function or structure, suggesting possible systemic or paracrine cellular mechanisms. Combining MSCs with CPCs was associated with improvement in both these outcomes. These results suggest potential important beneficial effects of CPCs and MSCs and support further investigation in HF patients.


Assuntos
Insuficiência Cardíaca , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Minnesota , Qualidade de Vida , Volume Sistólico , Resultado do Tratamento , Função Ventricular Esquerda
3.
ESC Heart Fail ; 8(3): 2306-2309, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33652498

RESUMO

The concept that cell-based repair of myocardial injury might be possible was introduced almost two decades ago; however, the field of cardiovascular reparative medicine has been criticized as translation to clinically effective approaches has been slow. The recent retraction of a series of papers has further impacted perception of this area of research. As researchers, clinicians, and teachers in this field, we felt it incumbent to critically appraise the current state of cardiac cell repair, determine what can be learned from past mistakes, and formulate best practices for future work. This special communication summarizes an introspective assessment of what has fallen short, how to prevent similar issues, and how the field might best move forward in the service of science and patients.


Assuntos
Regeneração , Transplante de Células-Tronco , Terapia Baseada em Transplante de Células e Tecidos , Coração , Humanos
4.
PLoS One ; 15(8): e0237401, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32841277

RESUMO

Implantation of bone marrow-derived cells (BMCs) into mouse hearts post-myocardial infarction (MI) limits cardiac functional decline. However, clinical trials of post-MI BMC therapy have yielded conflicting results. While most laboratory experiments use healthy BMC donor mice, clinical trials use post-MI autologous BMCs. Post-MI mouse BMCs are therapeutically impaired, due to inflammatory changes in BMC composition. Thus, therapeutic efficacy of the BMCs progressively worsens after MI but recovers as donor inflammatory response resolves. The availability of post-MI patient BM mononuclear cells (MNCs) from the TIME and LateTIME clinical trials enabled us to test if human post-MI MNCs undergo a similar period of impaired efficacy. We hypothesized that MNCs from TIME trial patients would be less therapeutic than healthy human donor MNCs when implanted into post-MI mouse hearts, and that therapeutic properties would be restored in MNCs from LateTIME trial patients. Post-MI SCID mice received MNCs from healthy donors, TIME patients, or LateTIME patients. Cardiac function improved considerably in the healthy donor group, but neither the TIME nor LateTIME group showed therapeutic effect. Conclusion: post-MI human MNCs lack therapeutic benefits possessed by healthy MNCs, which may partially explain why BMC clinical trials have been less successful than mouse studies.


Assuntos
Transplante de Medula Óssea , Ensaios Clínicos como Assunto , Infarto do Miocárdio/terapia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Infarto do Miocárdio/genética , Resultado do Tratamento
6.
JACC CardioOncol ; 2(4): 581-595, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33403362

RESUMO

BACKGROUND: Anthracycline-induced cardiomyopathy (AIC) may be irreversible with a poor prognosis, disproportionately affecting women and young adults. Administration of allogeneic bone marrow-derived mesenchymal stromal cells (allo-MSCs) is a promising approach to heart failure (HF) treatment. OBJECTIVES: SENECA (Stem Cell Injection in Cancer Survivors) was a phase 1 study of allo-MSCs in AIC. METHODS: Cancer survivors with chronic AIC (mean age 56.6 years; 68% women; NT-proBNP 1,426 pg/ml; 6 enrolled in an open-label, lead-in phase and 31 subjects randomized 1:1) received 1 × 108 allo-MSCs or vehicle transendocardially. Primary objectives were safety and feasibility. Secondary efficacy measures included cardiac function and structure measured by cardiac magnetic resonance imaging (CMR), functional capacity, quality of life (Minnesota Living with Heart Failure Questionnaire), and biomarkers. RESULTS: A total of 97% of subjects underwent successful study product injections; all allo-MSC-assigned subjects received the target dose of cells. Follow-up visits were well-attended (92%) with successful collection of endpoints in 94% at the 1-year visit. Although 58% of subjects had non-CMR compatible devices, CMR endpoints were successfully collected in 84% of subjects imaged at 1 year. No new tumors were reported. There were no significant differences between allo-MSC and vehicle groups with regard to clinical outcomes. Secondary measures included 6-min walk test (p = 0.056) and Minnesota Living with Heart Failure Questionnaire score (p = 0.048), which tended to favor the allo-MSC group. CONCLUSIONS: In this first-in-human study of cell therapy in patients with AIC, transendocardial administration of allo-MSCs appears safe and feasible, and CMR was successfully performed in the majority of the HF patients with devices. This study lays the groundwork for phase 2 trials aimed at assessing efficacy of cell therapy in patients with AIC.

8.
J Am Heart Assoc ; 8(2): e011001, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30661439

RESUMO

Background The effects of race on response to medical therapy in people with peripheral artery disease ( PAD ) are unknown. Methods and Results In the PROPEL (Progenitor Cell Release Plus Exercise to Improve Functional Performance in PAD) Trial, PAD participants were randomized to 1 of 4 groups for 6 months: supervised treadmill exercise+granulocyte-macrophage colony-stimulating factor ( GM - CSF ) (Group 1), exercise+placebo (Group 2), attention control+ GM - CSF (Group 3), or attention control+placebo (Group 4). Change in 6-minute walk distance was measured at 12- and 26-week follow-up. In these exploratory analyses, groups receiving GM - CSF (Groups 1 and 3), placebo (Groups 2 and 4), exercise (Groups 1 and 2), and attention control (Groups 2 and 4) were combined, maximizing statistical power for studying the effects of race on response to interventions. Of 210 PAD participants, 141 (67%) were black and 64 (30%) were white. Among whites, GM - CSF improved 6-minute walk distance by +22.0 m (95% CI : -4.5, +48.5, P=0.103) at 12 weeks and +44.4 m (95% CI : +6.9, +82.0, P=0.020) at 26 weeks, compared with placebo. Among black participants, there was no effect of GM - CSF on 6-minute walk distance at 12-week ( P=0.26) or 26-week (-5.0 m [-27.5, +17.5, P=0.66]) follow-up, compared with placebo. There was an interaction of race on the effect of GM - CSF on 6-minute walk change at 26-week follow-up ( P=0.018). Exercise improved 6-minute walk distance in black ( P=0.006) and white ( P=0.034) participants without interaction. Conclusions GM - CSF improved 6-minute walk distance in whites with PAD but had no effect in black participants. Further study is needed to confirm racial differences in GM - CSF efficacy in PAD . Clinical Trial Registration URL : http://www.clinicaltrials.gov . Unique identifier: NCT 01408901.


Assuntos
Terapia por Exercício/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Extremidade Inferior/irrigação sanguínea , Doença Arterial Periférica/etnologia , Grupos Raciais , Caminhada/fisiologia , Idoso , Relação Dose-Resposta a Droga , Método Duplo-Cego , Teste de Esforço , Feminino , Seguimentos , Humanos , Injeções Subcutâneas , Masculino , Doença Arterial Periférica/fisiopatologia , Doença Arterial Periférica/terapia , Estudos Retrospectivos , Distribuição por Sexo , Fatores Sexuais , Resultado do Tratamento , Estados Unidos/epidemiologia
9.
Circ Res ; 123(4): 495-505, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-30355250

RESUMO

Although clinical trials of cell-based approaches to cardiovascular disease have yielded some promising results, no cell-based therapy has achieved regulatory approval for a cardiovascular indication. To broadly assess the challenges to regulatory approval and identify strategies to facilitate this goal, the Cardiac Safety Research Consortium sponsored a session during the Texas Heart Institute International Symposium on Cardiovascular Regenerative Medicine in September 2017. This session convened leaders in cardiovascular regenerative medicine, including participants from academia, the pharmaceutical industry, the US Food and Drug Administration, and the Cardiac Safety Research Consortium, with particular focus on treatments closest to regulatory approval. A goal of the session was to identify barriers to regulatory approval and potential pathways to overcome them. Barriers identified include manufacturing and therapeutic complexity, difficulties identifying an optimal comparator group, limited industry capacity for funding pivotal clinical trials, and challenges to demonstrating efficacy on clinical end points required for regulatory decisions. Strategies to overcome these barriers include precompetitive development of a cell therapy registry network to enable dual-purposing of clinical data as part of pragmatic clinical trial design, development of standardized terminology for product activity and end points to facilitate this registry, use of innovative statistical methods and quality of life or functional end points to supplement outcomes such as death or heart failure hospitalization and reduce sample size, involvement of patients in determining the research agenda, and use of the Food and Drug Administration's new Regenerative Medicine Advanced Therapy designation to facilitate early discussion with regulatory authorities when planning development pathways.


Assuntos
Cardiologia/métodos , Congressos como Assunto , Cardiopatias/terapia , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos , Animais , Humanos
10.
Curr Opin Organ Transplant ; 23(6): 664-672, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30247170

RESUMO

PURPOSE OF REVIEW: Heart transplantation is the only curative treatment option for end-stage heart failure. However, a shortage of donor organs is a major limitation of this approach. Regenerative medicine targets the goal of increasing the number of available hearts for transplantation. In this review, we highlight the state of the art of building a bioartificial heart. We summarize the components needed, the hurdles, and likely translational steps to make the dream of transplanting a totally functional bioartificial heart a possibility. RECENT FINDINGS: The therapies being developed in regenerative medicine aim not only to repair, but also to regenerate or replace failing tissues and organs. The engineering of bioartificial hearts utilizing patient-derived cells could theoretically solve the two main complications of heart transplantations: graft rejection and lifelong immunosuppression. Although many hurdles remain, scientists have reached a point in which some of these hurdles have been overcome. Decellularized heart scaffolds have emerged over the past decade as one of the most promising biofabrications. Two possible options for organ scaffolds exist: nontransplantable human hearts and porcine hearts. The use of these scaffolds could lead to the availability of an unlimited number of transplantable organs. The current challenge remains improving processes required for recellularization - including those for cells, bioreactors, and physiologic conditioning. Researchers should focus to solve these hurdles and pave the way toward the dream of in-vivo bioengineered heart maturation. SUMMARY: Regenerative medicine has emerged as one of the most promising fields of translational research and has the potential to both minimize the need for donor organs and increase their availability. Meeting the challenge of implanting a totally functional bioengineered heart lies in solving multiple issues simultaneously. Dwarfing the technical hurdles, cost is the largest barrier to success. The scientific hurdles mainly involve scaling up and scaling out of laboratory cell processes, building bioreactors, and delivering cells into every needed region of an organ scaffold. Maintaining sterility and quantifying readiness of the nascent organs are also critical for success.


Assuntos
Transplante de Coração/métodos , Coração/fisiopatologia , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos , Animais , Humanos , Suínos
11.
Artif Organs ; 42(10): 970-982, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30044011

RESUMO

Engineering a bioartificial heart has become a possibility in part because of the regenerative medicine approaches to repairing or replacing damaged organs that have evolved over the past two decades. With the advent of inducible pluripotent stem cell technology, it is now possible to generate personalized cells that make the concept of autologous tissue engineering imaginable. Scaffolds that provide form, function, and biological cues to cells likewise potentially enable the engineering of biocompatible vascularized solid organs. Decellularized organs or tissue matrices retain organ complexity and structure at the macro and micro scales, contain biologically active molecules that support cell phenotype and function, and are vascularized allowing full thickness tissue generation. There is also dynamic reciprocity between the extracellular matrix and cells, which does not occur with synthetic scaffolds and allows both to evolve as functional need changes, making it a unique scaffold. Yet, building a whole heart from decellularized scaffolds and cells requires delivering hundreds of billions of multiple types of cardiac cells appropriately and providing a milieu where they can survive and mature. We propose a novel type of in vivo organ engineering utilizing pre-clinical models where decellularized hearts are heterotopically transplanted with the intent to harness the capability of the body to at least in part repopulate the scaffold. By adding load and electrical input, possibly via temporary mechanical assistance, we posit that vascular and parenchymal cell maturation can occur. In this study, we implanted porcine decellularized hearts acutely and chronically in living recipients in a heterotopic position. We demonstrated that the surgical procedure is critical to prevent coagulation and to increase graft patency. We also demonstrated that short-term implantation promotes endothelial cell adhesion to the vessel lumens and that long-term implantation also promotes tissue formation with evidence of cardiomyocytes and endothelial cells present within the graft. Utilizing endogenous repair capabilities of the recipient in response to a naked ECM, we allowed the transplanted scaffold to direct host cells-both organizationally and functionally. Thus, the scaffold provided necessary cues for cell organization and remodeling within the transplanted organ. Future work would involve culturing partially recellularized engineered organs in bioreactors where mechanical and electrical stimulation can be controlled to promote organ development and then transplanting these after a minimal level of maturation has been achieved.


Assuntos
Órgãos Bioartificiais , Matriz Extracelular/química , Matriz Extracelular/transplante , Transplante de Coração/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Bovinos , Adesão Celular , Células Endoteliais/citologia , Feminino , Masculino , Miocárdio/química , Miocárdio/citologia , Miócitos Cardíacos/citologia , Suínos
13.
Circ Res ; 122(12): 1703-1715, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29703749

RESUMO

RATIONALE: Autologous bone marrow mesenchymal stem cells (MSCs) and c-kit+ cardiac progenitor cells (CPCs) are 2 promising cell types being evaluated for patients with heart failure (HF) secondary to ischemic cardiomyopathy. No information is available in humans about the relative efficacy of MSCs and CPCs and whether their combination is more efficacious than either cell type alone. OBJECTIVE: CONCERT-HF (Combination of Mesenchymal and c-kit+ Cardiac Stem Cells As Regenerative Therapy for Heart Failure) is a phase II trial aimed at elucidating these issues by assessing the feasibility, safety, and efficacy of transendocardial administration of autologous MSCs and CPCs, alone and in combination, in patients with HF caused by chronic ischemic cardiomyopathy (coronary artery disease and old myocardial infarction). METHODS AND RESULTS: Using a randomized, double-blinded, placebo-controlled, multicenter, multitreatment, and adaptive design, CONCERT-HF examines whether administration of MSCs alone, CPCs alone, or MSCs+CPCs in this population alleviates left ventricular remodeling and dysfunction, reduces scar size, improves quality of life, or augments functional capacity. The 4-arm design enables comparisons of MSCs alone with CPCs alone and with their combination. CONCERT-HF consists of 162 patients, 18 in a safety lead-in phase (stage 1) and 144 in the main trial (stage 2). Stage 1 is complete, and stage 2 is currently randomizing patients from 7 centers across the United States. CONCLUSIONS: CONCERT-HF will provide important insights into the potential therapeutic utility of MSCs and CPCs, given alone and in combination, for patients with HF secondary to ischemic cardiomyopathy. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02501811.


Assuntos
Insuficiência Cardíaca/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Miócitos Cardíacos/citologia , Transplante de Células-Tronco/métodos , Terapia Combinada/métodos , Método Duplo-Cego , Estudos de Viabilidade , Insuficiência Cardíaca/etiologia , Humanos , Isquemia Miocárdica/complicações , Miócitos Cardíacos/química , Proteínas Proto-Oncogênicas c-kit , Projetos de Pesquisa , Transplante Autólogo , Resultado do Tratamento , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/terapia , Remodelação Ventricular
14.
Circ Res ; 122(3): 479-488, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29208679

RESUMO

RATIONALE: The TIME trial (Timing in Myocardial Infarction Evaluation) was the first cell therapy trial sufficiently powered to determine if timing of cell delivery after ST-segment-elevation myocardial infarction affects recovery of left ventricular (LV) function. OBJECTIVE: To report the 2-year clinical and cardiac magnetic resonance imaging results and their modification by microvascular obstruction. METHODS AND RESULTS: TIME was a randomized, double-blind, placebo-controlled trial comparing 150 million bone marrow mononuclear cells versus placebo in 120 patients with anterior ST-segment-elevation myocardial infarctions resulting in LV dysfunction. Primary end points included changes in global (LV ejection fraction) and regional (infarct and border zone) function. Secondary end points included changes in LV volumes, infarct size, and major adverse cardiac events. Here, we analyzed the continued trajectory of these measures out to 2 years and the influence of microvascular obstruction present at baseline on these long-term outcomes. At 2 years (n=85), LV ejection fraction was similar in the bone marrow mononuclear cells (48.7%) and placebo groups (51.6%) with no difference in regional LV function. Infarct size and LV mass decreased ≥30% in each group at 6 months and declined gradually to 2 years. LV volumes increased ≈10% at 6 months and remained stable to 2 years. Microvascular obstruction was present in 48 patients at baseline and was associated with significantly larger infarct size (56.5 versus 36.2 g), greater adverse LV remodeling, and marked reduction in LV ejection fraction recovery (0.2% versus 6.2%). CONCLUSIONS: In one of the longest serial cardiac magnetic resonance imaging analyses of patients with large anterior ST-segment-elevation myocardial infarctions, bone marrow mononuclear cells administration did not improve recovery of LV function over 2 years. Microvascular obstruction was associated with reduced recovery of LV function, greater adverse LV remodeling, and more device implantations. The use of cardiac magnetic resonance imaging leads to greater dropout of patients over time because of device implantation in patients with more severe LV dysfunction resulting in overestimation of clinical stability of the cohort. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00684021.


Assuntos
Transplante de Medula Óssea/métodos , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Disfunção Ventricular Esquerda/terapia , Adulto , Idoso , Método Duplo-Cego , Feminino , Seguimentos , Ventrículos do Coração/patologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Microcirculação , Pessoa de Meia-Idade , Tamanho do Órgão , Infarto do Miocárdio com Supradesnível do Segmento ST/complicações , Infarto do Miocárdio com Supradesnível do Segmento ST/patologia , Volume Sistólico , Fatores de Tempo , Disfunção Ventricular Esquerda/etiologia
15.
Med Hypotheses ; 109: 38-41, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29150290

RESUMO

Contributory risk factors to premature coronary artery disease (CAD) in premenopausal women are poorly understood and data on this subset of women is lacking. There is growing evidence that the process of inflammation is a part of the atherosclerotic process. Mechanistic insights from animal work suggest that the profile of circulating cytokines reflects both endothelial integrity and the presence of immune and progenitor cells. Significant differences in pro- and anti-inflammatory cytokine concentrations between patients with and without CAD exist. Young women with obstructive CAD may experience differences in pro-inflammatory cytokines and the recruitment of reparative cells that secrete T-Helper (Th2 cytokines compared to women without CAD. Thus, cytokine balance may play a role in obstructive CAD in young women. In this pilot study we set out to identify an array of circulating inflammatory marker profiles which could be useful for the development of risk assessment and preventive strategies. We tested the hypothesis that an increase in serologic Th1 cytokines relative to Th2)/hematopoietic regulatory (HR) cytokines is related to premature coronary atherosclerosis in premenopausal women.


Assuntos
Aterosclerose/diagnóstico , Aterosclerose/metabolismo , Citocinas/metabolismo , Pré-Menopausa , Adulto , Biomarcadores/metabolismo , Angiografia Coronária , Células Endoteliais/metabolismo , Feminino , Humanos , Sistema Imunitário , Inflamação , Pessoa de Meia-Idade , Modelos Biológicos , Projetos Piloto , Medição de Risco , Fatores de Risco , Células Th1/imunologia , Células Th2/imunologia
16.
Circ Res ; 120(12): 1947-1957, 2017 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-28490433

RESUMO

RATIONALE: Intracoronary infusion of bone marrow (BM) mononuclear cells after acute myocardial infarction (AMI) has led to limited improvement in left ventricular function. Although experimental AMI models have implicated cytokine-related impairment of progenitor cell function, this response has not been investigated in humans. OBJECTIVE: To test the hypothesis that peripheral blood (PB) cytokines predict BM endothelial progenitor cell colony outgrowth and cardiac function after AMI. METHODS AND RESULTS: BM and PB samples were collected from 87 participants 14 to 21 days after AMI and BM from healthy donors was used as a reference. Correlations between cytokine concentrations and cell phenotypes, cell functions, and post-AMI cardiac function were determined. PB interleukin-6 (IL-6) negatively correlated with endothelial colony-forming cell colony maximum in the BM of patients with AMI (estimate±SE, -0.13±0.05; P=0.007). BM from healthy individuals showed a dose-dependent decrease in endothelial colony-forming cell colony outgrowth in the presence of exogenous IL-1ß or IL-6 (P<0.05). Blocking the IL-1R or IL-6R reversed cytokine impairment. In AMI study participants, the angiogenic cytokine platelet-derived growth factor BB glycoprotein correlated positively with BM-derived colony-forming unit-endothelial colony maximum (estimate±SE, 0.01±0.002; P<0.001), multipotent mesenchymal stromal cell colony maximum (estimate±SE, 0.01±0.002; P=0.002) in BM, and mesenchymal stromal cell colony maximum in PB (estimate±SE, 0.02±0.005; P<0.001). CONCLUSIONS: Two weeks after AMI, increased PB platelet-derived growth factor BB glycoprotein was associated with increased BM function, whereas increased IL-6 was associated with BM impairment. Validation studies confirmed inflammatory cytokine impairment of BM that could be reversed by blocking IL-1R or IL-6R. Together, these studies suggest that blocking IL-1 or IL-6 receptors may improve the regenerative capacity of BM cells after AMI. CLINICAL TRIAL REGISTRATIONS: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00684060.


Assuntos
Células da Medula Óssea/fisiologia , Citocinas/sangue , Interleucina-1beta/sangue , Interleucina-6/sangue , Infarto do Miocárdio/sangue , Medula Óssea/fisiologia , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/diagnóstico
17.
Curr Stem Cell Rep ; 3(1): 35-44, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28261549

RESUMO

PURPOSE OF REVIEW: In this review, we focus on the multiple advancements in the field of cardiovascular regenerative medicine and the state-of-the art of building a heart. An organ is comprised of cells, but cells alone do not comprise an organ. We summarize the components needed, the hurdles, and likely translational steps defining the opportunities for discovery. RECENT FINDINGS: The therapies being developed in regenerative medicine aim not only to repair, but also to regenerate or replace ailing tissues and organs. The first generation of cardiac regenerative medicine was gene therapy. The past decade has focused primarily on cell therapy, particularly for repair after ischemic injury with mixed results. Although cell therapy is promising, it will likely never reverse end-stage heart failure; and thus, the unmet need is, and will remain, for organs. Scientists have now tissue engineering and regenerative medicine concepts to invent alternative therapies for a wide spectrum of diseases encompassing cardiovascular, respiratory, gastrointestinal, hepatic, renal, musculoskeletal, ocular, and neurodegenerative disorders. Current studies focus on potential scaffolds and applying concepts and techniques learned with testbeds to building human sized organs. Special focus has been given to scaffold sources, cells types and sources, and cell integration with scaffolds. The complexity arises in combining them to yield an organ. SUMMARY: Regenerative medicine has emerged as one of the most promising fields of translational research and has the potential to minimize both the need for, and increase the availability of, donor organs. The field is characterized by its integration of biology, physical sciences, and engineering. The proper integration of these fields could lead to off-the-shelf bioartificial organs that are suitable for transplantation. Building a heart will necessarily require a scaffold that can provide cardiac function. We believe that the advent of decellularization methods provides complex, unique, and natural scaffold sources. Ultimately, cell biology and tissue engineering will need to synergize with scaffold biology, finding cell sources and reproducible ways to expand their numbers is an unmet need. But tissue engineering is moving toward whole organ synthesis at an unparalleled pace.

18.
Circulation ; 135(15): 1417-1428, 2017 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-28209728

RESUMO

BACKGROUND: Atherosclerotic peripheral artery disease affects 8% to 12% of Americans >65 years of age and is associated with a major decline in functional status, increased myocardial infarction and stroke rates, and increased risk of ischemic amputation. Current treatment strategies for claudication have limitations. PACE (Patients With Intermittent Claudication Injected With ALDH Bright Cells) is a National Heart, Lung, and Blood Institute-sponsored, randomized, double-blind, placebo-controlled, phase 2 exploratory clinical trial designed to assess the safety and efficacy of autologous bone marrow-derived aldehyde dehydrogenase bright (ALDHbr) cells in patients with peripheral artery disease and to explore associated claudication physiological mechanisms. METHODS: All participants, randomized 1:1 to receive ALDHbr cells or placebo, underwent bone marrow aspiration and isolation of ALDHbr cells, followed by 10 injections into the thigh and calf of the index leg. The coprimary end points were change from baseline to 6 months in peak walking time (PWT), collateral count, peak hyperemic popliteal flow, and capillary perfusion measured by magnetic resonance imaging, as well as safety. RESULTS: A total of 82 patients with claudication and infrainguinal peripheral artery disease were randomized at 9 sites, of whom 78 had analyzable data (57 male, 21 female patients; mean age, 66±9 years). The mean±SEM differences in the change over 6 months between study groups for PWT (0.9±0.8 minutes; 95% confidence interval [CI] -0.6 to 2.5; P=0.238), collateral count (0.9±0.6 arteries; 95% CI, -0.2 to 2.1; P=0.116), peak hyperemic popliteal flow (0.0±0.4 mL/s; 95% CI, -0.8 to 0.8; P=0.978), and capillary perfusion (-0.2±0.6%; 95% CI, -1.3 to 0.9; P=0.752) were not significant. In addition, there were no significant differences for the secondary end points, including quality-of-life measures. There were no adverse safety outcomes. Correlative relationships between magnetic resonance imaging measures and PWT were not significant. A post hoc exploratory analysis suggested that ALDHbr cell administration might be associated with an increase in the number of collateral arteries (1.5±0.7; 95% CI, 0.1-2.9; P=0.047) in participants with completely occluded femoral arteries. CONCLUSIONS: ALDHbr cell administration did not improve PWT or magnetic resonance outcomes, and the changes in PWT were not associated with the anatomic or physiological magnetic resonance imaging end points. Future peripheral artery disease cell therapy investigational trial design may be informed by new anatomic and perfusion insights. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01774097.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Doença Arterial Periférica/terapia , Idoso , Aldeído Desidrogenase/metabolismo , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Comorbidade , Exercício Físico , Extremidades/irrigação sanguínea , Feminino , Seguimentos , Humanos , Claudicação Intermitente/terapia , Masculino , Pessoa de Meia-Idade , Perfusão , Doença Arterial Periférica/diagnóstico , Doença Arterial Periférica/metabolismo , Qualidade de Vida , Fatores de Risco , Resultado do Tratamento
20.
J Cell Physiol ; 232(11): 3139-3145, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28059438

RESUMO

Numerous protocols exist for isolating aortic endothelial and smooth muscle cells from small animals. However, establishing a protocol for isolating pure cell populations from large animal vessels that are more elastic has been challenging. We developed a simple sequential enzymatic approach to isolate highly purified populations of porcine aortic endothelial and smooth muscle cells. The lumen of a porcine aorta was filled with 25 U/ml dispase solution and incubated at 37°C to dissociate the endothelial cells. The smooth muscle cells were isolated by mincing the tunica media of the treated aorta and incubating the pieces in 0.2% and then 0.1% collagenase type I solution. The isolated endothelial cells stained positive for von Willebrand factor, and 97.2% of them expressed CD31. Early and late passage endothelial cells had a population doubling time of 38 hr and maintained a capacity to take up DiI-Ac-LDL and form tubes in Matrigel®. The isolated smooth muscle cells stained highly positive for alpha-smooth muscle actin, and an impurities assessment showed that only 1.8% were endothelial cells. Population doubling time for the smooth muscle cells was ∼70 hr at passages 3 and 7; and the cells positively responded to endothelin-1, as shown by a 66% increase in the intracellular calcium level. This simple protocol allows for the isolation of highly pure populations of endothelial and smooth muscle cells from porcine aorta that can survive continued passage in culture without losing functionality or becoming overgrown by fibroblasts.


Assuntos
Separação Celular/métodos , Células Endoteliais/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/patologia , Animais , Aorta/citologia , Transporte Biológico , Biomarcadores/sangue , Cálcio/metabolismo , Proliferação de Células , Colagenases/metabolismo , Endopeptidases/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotelina-1/farmacologia , Citometria de Fluxo , Lipoproteínas LDL/metabolismo , Microscopia de Fluorescência , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica , Fenótipo , Sus scrofa , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA