Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 190
Filtrar
1.
J Mol Cell Cardiol ; 183: 14-21, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37595498

RESUMO

Despite the dramatic improvements in the management of patients with chronic heart failure which have occurred over the last decades, some of them still exhaust conventional drug-based therapies without being eligible for more aggressive options like heart transplantation or implantation of a left ventricular assist device. Cell therapy has thus emerged as a possible means of filling this niche. Multiple cell types have now been tested both in the laboratory but also in the clinics and it is fair to acknowledge that none of the clinical trials have yet conclusively proven the efficacy of cell-based approaches. These clinical studies, however, have entailed the use of cells from various sources but of non-cardiac lineage origins. Although this might not be the main reason for their failures, the discovery of pluripotent stem cells capable of generating cardiomyocytes now raises the hope that such cardiac-committed cells could be therapeutically more effective. In this review, we will first describe where we currently are with regard to the clinical trials using PSC-differentiated cells and discuss the main issues which remain to be addressed. In parallel, because the capacity of cells to stably engraft in the recipient heart has increasingly been questioned, it has been hypothesized that a major mechanism of action could be the cell-triggered release of biomolecules that foster host-associated reparative pathways. Thus, in the second part of this review, we will discuss the rationale, clinically relevant advantages and pitfalls associated with the use of these PSC "products".


Assuntos
Procedimentos Cirúrgicos Cardíacos , Insuficiência Cardíaca , Células-Tronco Pluripotentes , Humanos , Células-Tronco Embrionárias , Insuficiência Cardíaca/terapia , Miócitos Cardíacos/transplante , Transplante de Células-Tronco
2.
Crit Care ; 26(1): 48, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35189925

RESUMO

BACKGROUND: Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-induced acute respiratory distress syndrome (ARDS) causes high mortality. Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) have potentially relevant immune-modulatory properties, whose place in ARDS treatment is not established. This phase 2b trial was undertaken to assess the efficacy of UC-MSCs in patients with SARS-CoV-2-induced ARDS. METHODS: This multicentre, double-blind, randomized, placebo-controlled trial (STROMA-CoV-2) recruited adults (≥ 18 years) with SARS-CoV-2-induced early (< 96 h) mild-to-severe ARDS in 10 French centres. Patients were randomly assigned to receive three intravenous infusions of 106 UC-MSCs/kg or placebo (0.9% NaCl) over 5 days after recruitment. For the modified intention-to-treat population, the primary endpoint was the partial pressure of oxygen to fractional inspired oxygen (PaO2/FiO2)-ratio change between baseline (day (D) 0) and D7. RESULTS: Among the 107 patients screened for eligibility from April 6, 2020, to October 29, 2020, 45 were enrolled, randomized and analyzed. PaO2/FiO2 changes between D0 and D7 did not differ significantly between the UC-MSCs and placebo groups (medians [IQR] 54.3 [- 15.5 to 93.3] vs 25.3 [- 33.3 to 104.6], respectively; ANCOVA estimated treatment effect 7.4, 95% CI - 44.7 to 59.7; P = 0.77). Six (28.6%) of the 21 UC-MSCs recipients and six of 24 (25%) placebo-group patients experienced serious adverse events, none of which were related to UC-MSCs treatment. CONCLUSIONS: D0-to-D7 PaO2/FiO2 changes for intravenous UC-MSCs-versus placebo-treated adults with SARS-CoV-2-induced ARDS did not differ significantly. Repeated UC-MSCs infusions were not associated with any serious adverse events during treatment or thereafter (until D28). Larger trials enrolling patients earlier during the course of their ARDS are needed to further assess UC-MSCs efficacy in this context. TRIAL REGISTRATION: NCT04333368. Registered 01 April 2020, https://clinicaltrials.gov/ct2/history/NCT04333368 .


Assuntos
COVID-19 , Células-Tronco Mesenquimais , Síndrome do Desconforto Respiratório , Método Duplo-Cego , Humanos , Síndrome do Desconforto Respiratório/terapia , SARS-CoV-2 , Resultado do Tratamento
3.
Circ Res ; 124(1): 161-169, 2019 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-30605412

RESUMO

On March 1 and 2, 2018, the National Institutes of Health 2018 Progenitor Cell Translational Consortium, Cardiovascular Bioengineering Symposium, was held at the University of Alabama at Birmingham. Convergence of life sciences and engineering to advance the understanding and treatment of heart failure was the theme of the meeting. Over 150 attendees were present, and >40 scientists presented their latest work on engineering human functional myocardium for disease modeling, drug development, and heart failure research. The scientists, engineers, and physicians in the field of cardiovascular sciences met and discussed the most recent advances in their work and proposed future strategies for overcoming the major roadblocks of cardiovascular bioengineering and therapy. Particular emphasis was given for manipulation and using of stem/progenitor cells, biomaterials, and methods to provide molecular, chemical, and mechanical cues to cells to influence their identity and fate in vitro and in vivo. Collectively, these works are profoundly impacting and progressing toward deciphering the mechanisms and developing novel treatments for left ventricular dysfunction of failing hearts. Here, we present some important perspectives that emerged from this meeting.


Assuntos
Disciplinas das Ciências Biológicas , Engenharia Biomédica , Pesquisa Biomédica , Insuficiência Cardíaca , Comunicação Interdisciplinar , Animais , Comportamento Cooperativo , Difusão de Inovações , Coração/fisiopatologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Humanos , Miocárdio/metabolismo , Miocárdio/patologia , Recuperação de Função Fisiológica , Regeneração
4.
Eur Heart J ; 41(38): 3702-3710, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32860406

RESUMO

AIMS: Bone marrow-derived mononuclear cell (BM-MNC) therapy may improve myocardial recovery in patients following acute myocardial infarction (AMI), though existing trial results are inconsistent. METHODS AND RESULTS: Originally an open-label, multicentre Phase III trial, BAMI was designed to demonstrate the safety and efficacy of intracoronary infusion of BM-MNCs in reducing the time to all-cause mortality in patients with reduced left ventricular ejection fraction (LVEF, ≤45%) after primary angioplasty (PPCI) for ST-elevation AMI. Unexpectedly low recruitment means the trial no longer qualifies as a hypothesis-testing trial, but is instead an observational study with no definitive conclusions possible from statistical analysis. In total, 375 patients were recruited: 185 patients were randomized to the treatment arm (intracoronary infusion of BM-MNCs 2-8 days after PPCI) and 190 patients to the control arm (optimal medical therapy). All-cause mortality at 2 years was 3.26% [6 deaths; 95% confidence interval (CI): 1.48-7.12%] in the BM-MNC group and 3.82% (7 deaths; 95% CI: 1.84-7.84%) in the control group. Five patients (2.7%, 95% CI: 1.0-5.9%) in the BM-MNC group and 15 patients (8.1%, CI : 4.7-12.5%) in the control group were hospitalized for heart failure during 2 years of follow-up. Neither adverse events nor serious adverse events differed between the two groups. There were no patients hospitalized for stroke in the control group and 4 (2.2%) patients hospitalized for stroke in the BM-MNC group. CONCLUSIONS: Although BAMI is the largest trial of autologous cell-based therapy in the treatment of AMI, unexpectedly low recruitment and event rates preclude any meaningful group comparisons and interpretation of the observed results.


Assuntos
Infarto do Miocárdio , Função Ventricular Esquerda , Medula Óssea , Transplante de Medula Óssea , Humanos , Infarto do Miocárdio/terapia , Volume Sistólico , Transplante Autólogo , Resultado do Tratamento
5.
J Mol Cell Cardiol ; 141: 1-10, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32169551

RESUMO

BACKGROUND: We have shown that genetic overexpression of cell cycle proteins can increase the proliferation of transplanted cardiomyocytes derived from human induced-pluripotent stem cells (hiPSC-CMs) in animal models of myocardial infarction (MI). Here, we introduce a new, non-genetic approach to promote hiPSC-CM cell cycle activity and proliferation in transplanted human cardiomyocyte patches (hCMPs). METHODS: Mice were randomly distributed into 5 experimental groups (n = 10 per group). One group underwent Sham surgery, and the other 4 groups underwent MI induction surgery followed by treatment with hCMPs composed of hiPSC-CMs and nanoparticles that contained CHIR99021 and FGF1 (the NPCF-hCMP group), with hCMPs composed of hiPSC-CMs and empty nanoparticles (the NPE-hCMP group); with patches containing the CHIR99021/FGF-loaded nanoparticles but lacking hiPSC-CMs (the NPCF-Patch group), or patches lacking both the nanoparticles and cells (the E-Patch group). Cell cycle activity was evaluated via Ki67 and Aurora B expression, bromodeoxyuridine incorporation, and phosphorylated histone 3 levels (immunofluorescence); engraftment via human cardiac troponin T or human nuclear antigen expression (immunofluorescence) and bioluminescence imaging; cardiac function via echocardiography; infarct size and wall thickness via histology; angiogenesis via isolectin B4 expression (immunofluorescence); and apoptosis via TUNEL and caspace 3 expression (immunofluorescence). RESULTS: Combined CHIR99021- and FGF1-treatment significantly increased hiPSC-CM cell cycle activity both in cultured cells (by 4- to 6-fold) and in transplanted hCMPs, and compared to treatment with NPE-hCMPs, NPCF-hCMP transplantation increased hiPSC-CM engraftment by ~4-fold and was associated with significantly better measurements of cardiac function, infarct size, wall thickness, angiogenesis, and hiPSC-CM apoptosis four weeks after MI induction. CONCLUSIONS: Nanoparticle-mediated CHIR99021 and FGF1 delivery promotes hiPSC-CM cell cycle activity and proliferation, as well as the engraftment and regenerative potency of transplanted hCMPs, in a mouse MI model.


Assuntos
Fator 1 de Crescimento de Fibroblastos/farmacologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Regeneração/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Camundongos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Nanopartículas/química , Neovascularização Fisiológica/efeitos dos fármacos , Transplante de Células-Tronco
6.
Circ Res ; 123(1): 100-106, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29592957

RESUMO

RATIONALE: A rapid and massive influx of inflammatory cells occurs into ischemic area after myocardial infarction (MI), resulting in local release of cytokines and growth factors. Yet, the mechanisms regulating their production are not fully explored. The release of extracellular vesicles (EVs) in the interstitial space curbs important biological functions, including inflammation, and influences the development of cardiovascular diseases. To date, there is no evidence for in situ release of cardiac EVs after MI. OBJECTIVE: The present study tested the hypothesis that local EV generation in the infarcted heart coordinates cardiac inflammation after MI. METHODS AND RESULTS: Coronary artery ligation in mice transiently increases EV levels in the left ventricle when compared with sham animals. EVs from infarcted hearts were characterized as large vesicles (252±18 nm) expressing cardiomyocyte and endothelial markers and small EVs (118±4 nm) harboring exosomal markers, such as CD (cluster of differentiation) 63 and CD9. Cardiac large EVs generated after MI, but not small EVs or sham EVs, increased the release of IL (interleukin)-6, CCL (chemokine ligand) 2, and CCL7 from fluorescence-activated cell-sorted Ly6C+ cardiac monocytes. EVs of similar diameter were also isolated from fragments of interventricular septum obtained from patients undergoing aortic valve replacement, thus supporting the clinical relevance of our findings in mice. CONCLUSIONS: The present study demonstrates that acute MI transiently increases the generation of cardiac EVs characterized as both exosomes and microvesicles, originating mainly from cardiomyocytes and endothelial cells. EVs accumulating in the ischemic myocardium are rapidly taken up by infiltrating monocytes and regulate local inflammatory responses.


Assuntos
Vesículas Extracelulares/patologia , Infarto do Miocárdio/patologia , Miocardite/etiologia , Animais , Biomarcadores/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Vasos Coronários , Células Endoteliais/metabolismo , Exossomos , Vesículas Extracelulares/metabolismo , Interleucina-6/metabolismo , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia
7.
Eur Heart J ; 39(20): 1835-1847, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29420830

RESUMO

Aims: We have shown that extracellular vesicles (EVs) secreted by embryonic stem cell-derived cardiovascular progenitor cells (Pg) recapitulate the therapeutic effects of their parent cells in a mouse model of chronic heart failure (CHF). Our objectives are to investigate whether EV released by more readily available cell sources are therapeutic, whether their effectiveness is influenced by the differentiation state of the secreting cell, and through which mechanisms they act. Methods and results: The total EV secreted by human induced pluripotent stem cell-derived cardiovascular progenitors (iPSC-Pg) and human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) were isolated by ultracentrifugation and characterized by Nanoparticle Tracking Analysis, western blot, and cryo-electron microscopy. In vitro bioactivity assays were used to evaluate their cellular effects. Cell and EV microRNA (miRNA) content were assessed by miRNA array. Myocardial infarction was induced in 199 nude mice. Three weeks later, mice with left ventricular ejection fraction (LVEF) ≤ 45% received transcutaneous echo-guided injections of iPSC-CM (1.4 × 106, n = 19), iPSC-Pg (1.4 × 106, n = 17), total EV secreted by 1.4 × 106 iPSC-Pg (n = 19), or phosphate-buffered saline (control, n = 17) into the peri-infarct myocardium. Seven weeks later, hearts were evaluated by echocardiography, histology, and gene expression profiling, blinded to treatment group. In vitro, EV were internalized by target cells, increased cell survival, cell proliferation, and endothelial cell migration in a dose-dependent manner and stimulated tube formation. Extracellular vesicles were rich in miRNAs and most of the 16 highly abundant, evolutionarily conserved miRNAs are associated with tissue-repair pathways. In vivo, EV outperformed cell injections, significantly improving cardiac function through decreased left ventricular volumes (left ventricular end systolic volume: -11%, P < 0.001; left ventricular end diastolic volume: -4%, P = 0.002), and increased LVEF (+14%, P < 0.0001) relative to baseline values. Gene profiling revealed that EV-treated hearts were enriched for tissue reparative pathways. Conclusion: Extracellular vesicles secreted by iPSC-Pg are effective in the treatment of CHF, possibly, in part, through their specific miRNA signature and the associated stimulation of distinct cardioprotective pathways. The processing and regulatory advantages of EV could make them effective substitutes for cell transplantation.


Assuntos
Vesículas Extracelulares/transplante , Insuficiência Cardíaca/terapia , Animais , Proliferação de Células , Sobrevivência Celular , Células-Tronco Embrionárias/ultraestrutura , Vesículas Extracelulares/genética , Insuficiência Cardíaca/patologia , Humanos , Camundongos Nus , MicroRNAs/análise , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/ultraestrutura , Células-Tronco Pluripotentes/ultraestrutura , Resultado do Tratamento
8.
J Vasc Surg ; 68(3): 849-857, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29074110

RESUMO

OBJECTIVE: Various animal models of critical limb ischemia have been developed in the past. However, there is no animal model that can undergo endovascular treatment, while providing reproducible true critical limb ischemia with arterial ulcers and rest pain. We evaluated the efficacy of a new model of rabbit hindlimb ischemia created through a percutaneous approach using embolization with calibrated particles. METHODS: Through a percutaneous transauricular artery approach and selective catheterization of the superficial femoral artery, embolization of distal limb vessels was performed using a mixture of 300- to 500-µm calibrated microparticles (Embosphere, Merit Medical, Salt Lake City, Utah), saline solution, and iodine contrast. Clinical and ultrasound imaging-based blood flow evaluation was performed before embolization and during follow-up. Histologic evaluation was performed at humane killing 14 days after the procedure. RESULTS: The model was successfully created in 10 rabbits (10 limbs). One rabbit died of sudden death at 8 days after the procedure. The nine surviving rabbits developed hind ulcers. All rabbits had a higher pain score in the follow-up compared to baseline value (P < .0001). Blood flow in the saphenous artery decreased significantly after the procedure and later at 14 days follow-up (baseline value 63.4 ± 31.3 µL per cardiac cycle vs 32.0 ± 28.4 µL per cardiac cycle postprocedure [P = .0013] and 32.0 ± 28.4 µL per cardiac cycle at 14 days [P = .0015]). Pathology showed signs of severe limb ischemia in all rabbits with subacute and chronic injury patterns. CONCLUSIONS: A rabbit hind limb ischemia model created by percutaneous transauricular distal femoral artery embolization with calibrated particles may overcome some of the limitations of existing animal models. As such, this model could prove useful for assessing therapies designed to improve arterial perfusion and collateral growth.


Assuntos
Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Animais , Modelos Animais de Doenças , Procedimentos Endovasculares , Artéria Femoral/diagnóstico por imagem , Artéria Femoral/fisiopatologia , Artéria Femoral/cirurgia , Membro Posterior/diagnóstico por imagem , Isquemia/diagnóstico por imagem , Isquemia/cirurgia , Medição da Dor , Coelhos , Reprodutibilidade dos Testes , Ultrassonografia
9.
Eur J Vasc Endovasc Surg ; 56(3): 373-380, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30005965

RESUMO

OBJECTIVE/BACKGROUND: To evaluate the experimental feasibility of endovascular fenestration using specific endovascular scissor prototypes in an ovine model of acute aortic dissection (AD). METHODS: A previously described endovascular technique was used to create a model of acute type B AD in sheep. Endovascular fenestrations using either endovascular scissor prototypes or a long sheath were compared. Four prototypes of endovascular fenestration scissors were evaluated. Both validity of the experimental model of AD and technical success of endovascular fenestration were assessed by haemodynamic criteria, completion angiography, transesophageal echocardiography, and post-procedural analysis of harvested aortas. RESULTS: Experimental acute AD was created by endovascular means in 17 sheep, with a technical success rate of 82%. Systolic blood pressure was lower in the false lumen than in the true lumen (58 ± 5 vs. 79 ± 3 mmHg, respectively; p < .001). Endovascular fenestration was performed in 11 models (endovascular scissors n = 8; long sheath n = 3). Controlled endovascular fenestration was obtained by the use of endovascular scissors (n = 5/8), resulting in a significant rise in false lumen systolic blood pressure after fenestration (60 ± 2 vs. 67 ± 9 mmHg before and after fenestration, respectively; p < .047). Long sheath fenestration resulted in an uncontrolled flap motion, leading to either pseudo-coarctation syndrome or aortic rupture (58 ± 6 vs. 40 ± 2 mmHg before and after fenestration, respectively; p < .001). CONCLUSION: In this experimental study, a reproducible AD model has been developed in sheep using endovascular procedures exclusively to evaluate endovascular fenestration techniques. Endovascular fenestration using a long sheath appeared hazardous and risky in vivo. Endovascular scissors constitute a dedicated and suitable tool to perform a safe controlled and effective endovascular fenestration in an ovine model.


Assuntos
Aneurisma Aórtico/cirurgia , Dissecção Aórtica/cirurgia , Procedimentos Endovasculares/instrumentação , Instrumentos Cirúrgicos , Dissecção Aórtica/patologia , Dissecção Aórtica/fisiopatologia , Animais , Aneurisma Aórtico/patologia , Aneurisma Aórtico/fisiopatologia , Pressão Arterial , Modelos Animais de Doenças , Desenho de Equipamento , Estudos de Viabilidade , Teste de Materiais , Carneiro Doméstico
10.
JAMA ; 318(6): 548-556, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28787507

RESUMO

Importance: Low cardiac output syndrome after cardiac surgery is associated with high morbidity and mortality in patients with impaired left ventricular function. Objective: To assess the ability of preoperative levosimendan to prevent postoperative low cardiac output syndrome. Design, Setting, and Participants: Randomized, double-blind, placebo-controlled trial conducted in 13 French cardiac surgical centers. Patients with a left ventricular ejection fraction less than or equal to 40% and scheduled for isolated or combined coronary artery bypass grafting with cardiopulmonary bypass were enrolled from June 2013 until May 2015 and followed during 6 months (last follow-up, November 30, 2015). Interventions: Patients were assigned to a 24-hour infusion of levosimendan 0.1 µg/kg/min (n = 167) or placebo (n = 168) initiated after anesthetic induction. Main Outcomes and Measures: Composite end point reflecting low cardiac output syndrome with need for a catecholamine infusion 48 hours after study drug initiation, need for a left ventricular mechanical assist device or failure to wean from it at 96 hours after study drug initiation when the device was inserted preoperatively, or need for renal replacement therapy at any time postoperatively. It was hypothesized that levosimendan would reduce the incidence of this composite end point by 15% in comparison with placebo. Results: Among 336 randomized patients (mean age, 68 years; 16% women), 333 completed the trial. The primary end point occurred in 87 patients (52%) in the levosimendan group and 101 patients (61%) in the placebo group (absolute risk difference taking into account center effect, -7% [95% CI, -17% to 3%]; P = .15). Predefined subgroup analyses found no interaction with ejection fraction less than 30%, type of surgery, and preoperative use of ß-blockers, intra-aortic balloon pump, or catecholamines. The prevalence of hypotension (57% vs 48%), atrial fibrillation (50% vs 40%), and other adverse events did not significantly differ between levosimendan and placebo. Conclusions and Relevance: Among patients with low ejection fraction who were undergoing coronary artery bypass grafting with cardiopulmonary bypass, levosimendan compared with placebo did not result in a significant difference in the composite end point of prolonged catecholamine infusion, use of left ventricular mechanical assist device, or renal replacement therapy. These findings do not support the use of levosimendan for this indication. Trial Registration: EudraCT Number: 2012-000232-25; clinicaltrials.gov Identifier: NCT02184819.


Assuntos
Baixo Débito Cardíaco/prevenção & controle , Cardiotônicos/uso terapêutico , Ponte de Artéria Coronária/efeitos adversos , Hidrazonas/uso terapêutico , Pré-Medicação , Piridazinas/uso terapêutico , Idoso , Ponte Cardiopulmonar , Cardiotônicos/efeitos adversos , Catecolaminas/administração & dosagem , Método Duplo-Cego , Feminino , Coração Auxiliar , Humanos , Hidrazonas/efeitos adversos , Infusões Intravenosas , Análise de Intenção de Tratamento , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/prevenção & controle , Piridazinas/efeitos adversos , Terapia de Substituição Renal , Simendana , Volume Sistólico/efeitos dos fármacos , Falha de Tratamento
11.
Stem Cells ; 33(4): 1021-35, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25537366

RESUMO

Tissue engineering aims at recapitulating permissive conditions that enable cells to collaborate and form functional tissues. Applications range from human tissue modeling for diagnostic purposes to therapeutic solutions in regenerative medicine and surgery. Across this spectrum, human stem cells are the active ingredient, expandable virtually indefinitely and with the propensity to generate new tissue. Engaging lineage-specific differentiation requires a precise concerto of key spatial and temporal factors, such as soluble molecules and growth factors, but also physical and mechanical stimuli. These stimuli compete to modulate distinct developmental signaling pathways and ultimately affect the differentiation efficiency. The heart is a chemo-mechano-electrical biological system that behaves as both a sensor and an actuator. It can transduce electrical inputs to generate mechanical contraction and electrical wave propagation. Such a complex organ arises from multipart developmental events that interact with one another to self-regulate. Here, we overview the main events of heart development and the role of mechanical forces in modifying the microenvironment of the progenitor cells. We analyze the cascades regulating cardiac gene activation to illustrate how mechanotransduction is already involved in the most popular protocols for stem cell differentiation (SCD) into cardiomyocytes. We then review how forces are transmitted to embryonic stem cells by cell-substrate or cell-cell communications, and how biomaterials can be designed to mimic these interactions and help reproduce key features of the developmental milieu. Putting this back in a clinical perspective, many challenges need to be overcome before biomaterials-based SCD protocols can be scaled up and marketed.


Assuntos
Materiais Biomiméticos/administração & dosagem , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/transplante , Coração/crescimento & desenvolvimento , Animais , Materiais Biomiméticos/química , Comunicação Celular/fisiologia , Humanos , Mecanotransdução Celular/fisiologia , Miócitos Cardíacos/fisiologia , Engenharia Tecidual/métodos
12.
Biomacromolecules ; 17(5): 1662-72, 2016 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-27064341

RESUMO

The formation of chitosan hydrogels without any external cross-linking agent was successfully achieved by inducing the gelation of a viscous chitosan solution with aqueous NaOH or gaseous NH3. The hydrogels produced from high molecular weight (Mw ≈ 640 000 g mol(-1)) and extensively deacetylated chitosan (DA ≈ 2.8%) at polymer concentrations above ∼2.0% exhibited improved mechanical properties due to the increase of the chain entanglements and intermolecular junctions. The results also show that the physicochemical and mechanical properties of chitosan hydrogels can be controlled by varying their polymer concentration and by controlling the gelation conditions, that is, by using different gelation routes. The biological evaluation of such hydrogels for regeneration of infarcted myocardium revealed that chitosan hydrogels prepared from 1.5% polymer solutions were perfectly incorporated onto the epicardial surface of the heart and presented partial degradation accompanied by mononuclear cell infiltration.


Assuntos
Quitosana/química , Hidrogéis/química , Infarto do Miocárdio/prevenção & controle , Polímeros/química , Regeneração/fisiologia , Animais , Materiais Biocompatíveis , Reagentes de Ligações Cruzadas/química , Feminino , Teste de Materiais , Ratos , Ratos Wistar , Função Ventricular Esquerda , Água
13.
Eur Heart J ; 36(30): 2011-7, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25990469

RESUMO

AIMS: Comparative studies suggest that stem cells committed to a cardiac lineage are more effective for improving heart function than those featuring an extra-cardiac phenotype. We have therefore developed a population of human embryonic stem cell (ESC)-derived cardiac progenitor cells. METHODS AND RESULTS: Undifferentiated human ESCs (I6 line) were amplified and cardiac-committed by exposure to bone morphogenetic protein-2 and a fibroblast growth factor receptor inhibitor. Cells responding to these cardio-instructive cues express the cardiac transcription factor Isl-1 and the stage-specific embryonic antigen SSEA-1 which was then used to purify them by immunomagnetic sorting. The Isl-1(+) SSEA-1(+) cells were then embedded into a fibrin scaffold which was surgically delivered onto the infarct area in a 68-year-old patient suffering from severe heart failure [New York Heart Association [NYHA] functional Class III; left ventricular ejection fraction (LVEF): 26%]. A coronary artery bypass was performed concomitantly in a non-infarcted area. The implanted cells featured a high degree of purity (99% were SSEA-1(+)), had lost the expression of Sox-2 and Nanog, taken as markers for pluripotency, and strongly expressed Isl-1. The intraoperative delivery of the patch was expeditious. The post-operative course was uncomplicated either. After 3 months, the patient is symptomatically improved (NYHA functional Class I; LVEF: 36%) and a new-onset contractility is echocardiographically evident in the previously akinetic cell/patch-treated, non-revascularized area. There have been no complications such as arrhythmias, tumour formation, or immunosuppression-related adverse events. CONCLUSION: This observation demonstrates the feasibility of generating a clinical-grade population of human ESC-derived cardiac progenitors and combining it within a tissue-engineered construct. While any conclusion pertaining to efficacy would be meaningless, the patient's functional outcome yet provides an encouraging hint. Beyond this case, the platform that has been set could be useful for generating different ESC-derived lineage-specific progenies.


Assuntos
Insuficiência Cardíaca/terapia , Células-Tronco Embrionárias Humanas/transplante , Feminino , Humanos , Pessoa de Meia-Idade , Isquemia Miocárdica/terapia , Alicerces Teciduais , Resultado do Tratamento , Disfunção Ventricular Esquerda/terapia
14.
Eur Heart J ; 36(12): 743-50, 2015 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24835485

RESUMO

AIM: There is now compelling evidence that cells committed to a cardiac lineage are most effective for improving the function of infarcted hearts. This has been confirmed by our pre-clinical studies entailing transplantation of human embryonic stem cell (hESC)-derived cardiac progenitors in rat and non-human primate models of myocardial infarction. These data have paved the way for a translational programme aimed at a phase I clinical trial. METHODS AND RESULTS: The main steps of this programme have included (i) the expansion of a clone of pluripotent hESC to generate a master cell bank under good manufacturing practice conditions (GMP); (ii) a growth factor-induced cardiac specification; (iii) the purification of committed cells by immunomagnetic sorting to yield a stage-specific embryonic antigen (SSEA)-1-positive cell population strongly expressing the early cardiac transcription factor Isl-1; (iv) the incorporation of these cells into a fibrin scaffold; (v) a safety assessment focused on the loss of teratoma-forming cells by in vitro (transcriptomics) and in vivo (cell injections in immunodeficient mice) measurements; (vi) an extensive cytogenetic and viral testing; and (vii) the characterization of the final cell product and its release criteria. The data collected throughout this process have led to approval by the French regulatory authorities for a first-in-man clinical trial of transplantation of these SSEA-1(+) progenitors in patients with severely impaired cardiac function. CONCLUSION: Although several facets of this manufacturing process still need to be improved, these data may yet provide a useful platform for the production of hESC-derived cardiac progenitor cells under safe and cost-effective GMP conditions.


Assuntos
Células-Tronco Embrionárias Humanas/transplante , Separação Imunomagnética/métodos , Bancos de Tecidos/organização & administração , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ensaios Clínicos Fase I como Assunto , Análise Citogenética , Estudos de Avaliação como Assunto , Humanos , Camundongos SCID , Miócitos Cardíacos/citologia , Miócitos Cardíacos/transplante , Preservação de Tecido/métodos , Alicerces Teciduais
15.
Curr Opin Cardiol ; 30(2): 179-185, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25500893

RESUMO

PURPOSE OF REVIEW: Outcomes of stem cell trials in patients with advanced heart failure have been divergent, which has raised some scepticism about this therapy and led to recommending slowing clinical trials until basic issues have been more thoroughly addressed. It is therefore timely and relevant to examine the current data and discuss how recent findings may change the perspectives of stem cell therapy. RECENT FINDINGS: The most important recent change has been a shift in the mechanistic paradigm. Although the initial objective of stem cells was to physically replace dead cardiomyocytes and build a new electromechanically integrated myocardial tissue, it is now recognized that the unavoidable death of most of the transplanted cells makes this objective unrealistic. Indeed, the primary mechanism of action of the cells seems to be paracrine through the release of factors activating the endogenous signalling pathways, leading to cardioprotection. This hypothesis has several implications. First, it leads to focus on the efficiency of early retention, rather than on sustained survival, which, in turn, implies improving delivery approaches, largely through an increased reliance on adjunctive biomaterials; second, it may rationalize the use of allogeneic cells as long as their rejection is delayed to give them enough time for releasing the signalling biomolecules; and, finally, it raises the possibility that transplantation of cells could be replaced by the delivery of their sole secretome, possibly under the form of microvesicles. SUMMARY: Put together, these approaches could streamline the translational process and enhance large-scale clinical applications.

17.
Circ J ; 79(1): 70-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25482295

RESUMO

BACKGROUND: Little is known about mid-term (3-month) postoperative atrial fibrillation (MT-POAF) in patients treated with bioprosthetic aortic valve replacement (BAVR). The aim of this study was to describe the natural history, identify the predictors and investigate the potential consequences in terms of anti-thrombotic therapy. METHODS AND RESULTS: During a longitudinal, prospective study, 219 patients were treated with BAVR early (7 days) and at mid-term postoperatively (30 and 90 days). POAF was monitored and risk factors were identified on logistic regression analysis. History of previous AF (OR, 3.08; 95% CI: 1.35-6.98), early POAF (OR, 5.93; 95% CI: 2.96-11.8), and BMI (per 5 kg/m(2): OR, 1.46; 95% CI: 1.03-2.09), were independent predictors for MT-POAF whereas sex, age and Euroscore were not. Results were identical when restricted to the 176 patients free from preoperative AF. In this subgroup, 36 patients (20.4%) had MT-POAF; 33 out of 174 (18.7%) would have required anticoagulation (CHA2DS2VASc score ≥ 1). Conversely, patients with BMI <27.7 and sinus rhythm at early follow-up had a very low risk of MT-POAF (OR, 0.16; 95% CI: 0.06-0.42). CONCLUSIONS: There was a higher than expected occurrence of MT-POAF in patients treated with BAVR, particularly in overweight patients with early POAF. This raises the question of implementing an anti-thrombotic therapy in these patients at higher risk of delayed atrial arrhythmia.


Assuntos
Valva Aórtica/cirurgia , Fibrilação Atrial/etiologia , Bioprótese , Fibrinolíticos/uso terapêutico , Implante de Prótese de Valva Cardíaca , Próteses Valvulares Cardíacas , Complicações Pós-Operatórias/etiologia , Adiposidade , Idoso , Idoso de 80 Anos ou mais , Fibrilação Atrial/epidemiologia , Índice de Massa Corporal , Comorbidade , Diabetes Mellitus/epidemiologia , Feminino , Fibrinolíticos/administração & dosagem , Humanos , Hipertensão/epidemiologia , Masculino , Obesidade/complicações , Complicações Pós-Operatórias/epidemiologia , Período Pós-Operatório , Estudos Prospectivos , Recidiva , Fatores de Risco , Índice de Gravidade de Doença , Fumar/epidemiologia , Tromboembolia/epidemiologia , Tromboembolia/etiologia , Trombofilia/epidemiologia , Trombofilia/etiologia
18.
EBioMedicine ; 103: 105145, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38713924

RESUMO

BACKGROUND: There is increased evidence that the effects of stem cells can mostly be duplicated by administration of their secretome which might streamline the translation towards the clinics. METHODS: The 12-patient SECRET-HF phase 1 trial has thus been designed to determine the feasibility and safety of repeated intravenous injections of the extracellular vesicle (EV)-enriched secretome of cardiovascular progenitor cells differentiated from pluripotent stem cells in severely symptomatic patients with drug-refractory left ventricular (LV) dysfunction secondary to non-ischemic dilated cardiomyopathy. Here we report the case of the first treated patient (baseline NYHA class III; LV Ejection Fraction:25%) in whom a dose of 20 × 109 particles/kg was intravenously infused three times three weeks apart. FINDINGS: In addition to demonstrating the feasibility of producing a cardiac cell secretome compliant with Good Manufacturing Practice standards, this case documents the excellent tolerance of its repeated delivery, without any adverse events during or after infusions. Six months after the procedure, the patient is in NYHA Class II with improved echo parameters, a reduced daily need for diuretics (from 240 mg to 160 mg), no firing from the previously implanted automatic internal defibrillator and no alloimmunization against the drug product, thereby supporting its lack of immunogenicity. INTERPRETATION: The rationale underlying the intravenous route is that the infused EV-enriched secretome may act by rewiring endogenous immune cells, both circulating and in peripheral organs, to take on a reparative phenotype. These EV-modified immune cells could then traffic to the heart to effect tissue repair, including mitigation of inflammation which is a hallmark of cardiac failure. FUNDING: This trial is funded by the French Ministry of Health (Programme Hospitalier de Recherche CliniqueAOM19330) and the "France 2030" National Strategy Program (ANR-20-F2II-0003). It is sponsored by Assistance Publique-Hôpitaux de Paris.


Assuntos
Insuficiência Cardíaca , Secretoma , Humanos , Insuficiência Cardíaca/terapia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/etiologia , Secretoma/metabolismo , Masculino , Vesículas Extracelulares/metabolismo , Pessoa de Meia-Idade , Resultado do Tratamento
19.
Stem Cell Res Ther ; 15(1): 109, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38637891

RESUMO

BACKGROUND: The STROMA-CoV-2 study was a French phase 2b, multicenter, double-blind, randomized, placebo-controlled clinical trial that did not identify a significant efficacy of umbilical cord-derived mesenchymal stromal cells in patients with SARS-CoV-2-induced acute respiratory distress syndrome. Safety on day 28 was found to be good. The aim of our extended study was to assess the 6- and 12-month safety of UC-MSCs administration in the STROMA-CoV-2 cohort. METHODS: A detailed multi-domain assessment was conducted at 6 and 12 months following hospital discharge focusing on adverse events, lung computed tomography-scan, pulmonary and muscular functional status, and quality of life in the STROMA-CoV-2 cohort including SARS-CoV-2-related early (< 96 h) mild-to-severe acute respiratory distress syndrome. RESULTS: Between April 2020 and October 2020, 47 patients were enrolled, of whom 19 completed a 1-year follow-up. There were no significant differences in any endpoints or adverse effects between the UC-MSCs and placebo groups at the 6- and 12-month assessments. Ground-glass opacities persisted at 1 year in 5 patients (26.3%). Furthermore, diffusing capacity for carbon monoxide remained altered over 1 year, although no patient required oxygen or non-invasive ventilatory support. Quality of life revealed declines in mental, emotional and physical health throughout the follow-up period, and the six-minute walking distance remained slightly impaired at the 1-year patient assessment. CONCLUSIONS: This study suggests a favorable safety profile for the use of intravenous UC-MSCs in the context of the first French wave of SARS-CoV-2-related moderate-to-severe acute respiratory distress syndrome, with no adverse effects observed at 1 year.


Assuntos
COVID-19 , Células-Tronco Mesenquimais , Síndrome do Desconforto Respiratório , Humanos , COVID-19/terapia , Método Duplo-Cego , Qualidade de Vida , Síndrome do Desconforto Respiratório/tratamento farmacológico , SARS-CoV-2 , Resultado do Tratamento , Cordão Umbilical
20.
Circulation ; 126(23): 2720-7, 2012 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-23139296

RESUMO

BACKGROUND: Undersized ring annuloplasty for ischemic mitral regurgitation (MR) is associated with variable results and >30% MR recurrence. We tested whether subvalvular repair by severing second-order mitral chordae can improve annuloplasty by reducing papillary muscle tethering. METHODS AND RESULTS: Posterolateral myocardial infarction known to produce chronic remodeling and MR was created in 28 sheep. At 3 months, sheep were randomized to sham surgery versus isolated undersized annuloplasty versus isolated bileaflet chordal cutting versus the combined therapy (n=7 each). At baseline, chronic myocardial infarction (3 months), and euthanasia (6.6 months), we measured left ventricular (LV) volumes and ejection fraction, wall motion score index, MR regurgitation fraction and vena contracta, mitral annulus area, and posterior leaflet restriction angle (posterior leaflet to mitral annulus area) by 2-dimensional and 3-dimensional echocardiography. All groups were comparable at baseline and chronic myocardial infarction, with mild to moderate MR (MR vena contracta, 4.6±0.1 mm; MR regurgitation fraction, 24.2±2.9%) and mitral annulus dilatation (P<0.01). At euthanasia, MR progressed to moderate to severe in controls but decreased to trace with ring plus chordal cutting versus trace to mild with chordal cutting alone versus mild to moderate with ring alone (MR vena contracta, 5.9±1.1 mm in controls, 0.5±0.08 with both, 1.0±0.3 with chordal cutting alone, 2.0±0.4 with ring alone; P<0.01). In addition, LV end-systolic volume increased by 108% in controls versus 28% with ring plus chordal cutting, less than with each intervention alone (P<0.01). In multivariate analysis, LV end-systolic volume and mitral annulus area most strongly predicted MR (r(2)=0.82, P<0.01). CONCLUSIONS: Comprehensive annular and subvalvular repair improves long-term reduction of both chronic ischemic MR and LV remodeling without decreasing global or segmental LV function at follow-up.


Assuntos
Anuloplastia da Valva Mitral/métodos , Insuficiência da Valva Mitral/fisiopatologia , Insuficiência da Valva Mitral/cirurgia , Remodelação Ventricular/fisiologia , Animais , Seguimentos , Valva Mitral/diagnóstico por imagem , Valva Mitral/cirurgia , Insuficiência da Valva Mitral/diagnóstico por imagem , Distribuição Aleatória , Ovinos , Fatores de Tempo , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA