Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Stem Cell Res Ther ; 15(1): 119, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38659070

RESUMO

BACKGROUND: Adipose stromal cells (ASC) are a form of mesenchymal stromal cells that elicit effects primarily via secreted factors, which may have advantages for the treatment of injury or disease. Several previous studies have demonstrated a protective role for MSC/ASC on mitigating acute kidney injury but whether ASC derived factors could hasten recovery from established injury has not been evaluated. METHODS: We generated a concentrated secretome (CS) of human ASC under well-defined conditions and evaluated its ability to improve the recovery of renal function in a preclinical model of acute kidney injury (AKI) in rats. 24 h following bilateral ischemia/reperfusion (I/R), rats were randomized following determination of plasma creatinine into groups receiving vehicle -control or ASC-CS treatment by subcutaneous injection (2 mg protein/kg) and monitored for evaluation of renal function, structure and inflammation. RESULTS: Renal function, assessed by plasma creatinine levels, recovered faster in ASC-CS treated rats vs vehicle. The most prominent difference between the ASC-CS treated vs vehicle was observed in rats with the most severe degree of initial injury (Pcr > 3.0 mg/dl 24 h post I/R), whereas rats with less severe injury (Pcr < 2.9 mg/dl) recovered quickly regardless of treatment. The quicker recovery of ASC-treated rats with severe injury was associated with less tissue damage, inflammation, and lower plasma angiopoietin 2. In vitro, ASC-CS attenuated the activation of the Th17 phenotype in lymphocytes isolated from injured kidneys. CONCLUSIONS: Taken together, these data suggest that ASC-CS represents a potent therapeutic option to improve established AKI.


Assuntos
Injúria Renal Aguda , Inflamação , Injúria Renal Aguda/terapia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Ratos , Humanos , Inflamação/patologia , Inflamação/metabolismo , Masculino , Secretoma/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Ratos Sprague-Dawley , Injeções Subcutâneas , Rim/metabolismo , Rim/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/terapia , Células Estromais/metabolismo
2.
Angiogenesis ; 26(3): 423-436, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36977946

RESUMO

Severe inflammatory stress often leads to vessel rarefaction and fibrosis, resulting in limited tissue recovery. However, signaling pathways mediating these processes are not completely understood. Patients with ischemic and inflammatory conditions have increased systemic Activin A level, which frequently correlates with the severity of pathology. Yet, Activin A's contribution to disease progression, specifically to vascular homeostasis and remodeling, is not well defined. This study investigated vasculogenesis in an inflammatory environment with an emphasis on Activin A's role. Exposure of endothelial cells (EC) and perivascular cells (adipose stromal cells, ASC) to inflammatory stimuli (represented by blood mononuclear cells from healthy donors activated with lipopolysaccharide, aPBMC) dramatically decreased EC tubulogenesis or caused vessel rarefaction compared to control co-cultures, concurrent with increased Activin A secretion. Both EC and ASC upregulated Inhibin Ba mRNA and Activin A secretion in response to aPBMC or their secretome. We identified TNFα (in EC) and IL-1ß (in EC and ASC) as the exclusive inflammatory factors, present in aPBMC secretome, responsible for induction of Activin A. Similar to ASC, brain and placental pericytes upregulated Activin A in response to aPBMC and IL-1ß, but not TNFα. Both these cytokines individually diminished EC tubulogenesis. Blocking Activin A with neutralizing IgG mitigated detrimental effects of aPBMC or TNFα/IL-1ß on tubulogenesis in vitro and vessel formation in vivo. This study delineates the signaling pathway through which inflammatory cells have a detrimental effect on vessel formation and homeostasis, and highlights the central role of Activin A in this process. Transitory interference with Activin A during early phases of inflammatory or ischemic insult, with neutralizing antibodies or scavengers, may benefit vasculature preservation and overall tissue recovery.


Assuntos
Células Endoteliais , Placenta , Humanos , Feminino , Gravidez , Células Endoteliais/metabolismo , Ativinas/metabolismo , Diferenciação Celular , Células Cultivadas
3.
Stem Cells Dev ; 32(11-12): 301-313, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36924295

RESUMO

Vascularization of ischemic and fabricated tissues is essential for successful tissue repair and replacement therapies. Endothelial cells (ECs) and mesenchymal stem/stromal cells (MSCs) in close proximity spontaneously organize into vessels after coimplantation in semisolid matrices. Thus, local injection of EC mixed with MSC may facilitate tissue (re)vascularization. The organization of these cells into vessels is accompanied by induction of a key regulator of vasculogenesis, activin A, in MSC through juxtacrine pathway. Mechanisms regulating activin A expression are poorly understood; therefore, the contributions of notch signaling pathways were evaluated in EC-adipose mesenchymal stromal cells (ASC) cocultures. Disruption of notch signaling in EC + ASC cocultures with a γ-secretase inhibitor, DAPT, completely abrogated both activin A induction and production, depending on the stage of vasculogenesis. While DAPT stimulated EC proliferation concurrent with increased secretion of vasculogenic factors, it also prevented the crucial transition of ASC from progenitor to smooth muscle cell phenotype, collectively resulting in ineffective tubulogenesis. Silencing Notch2 in ASC abolished activin A production in cocultures, but resulted in normal ASC maturation. In contrast, silencing Notch3 in ASC led to autonomous upregulation of mural cell markers, and intercellular contact with EC further enhanced upregulation of these markers, concurrent with amplified activin A secretion. Strong induction of activin A expression was achieved by exposing ASC to immobilized notch ligand jagged1, whereas jagged1 IgG, added to EC + ASC incubation media, prevented activin A expression. Overall, this study revealed that EC control activin A expression in ASC through trans juxtacrine notch signaling pathways, and uninterrupted notch signaling is required for activin A production, although signaling through Notch2 and Notch3 produce opposing effects.


Assuntos
Células-Tronco Mesenquimais , Pericitos , Pericitos/metabolismo , Células Endoteliais/metabolismo , Inibidores da Agregação Plaquetária/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo
4.
Mol Aspects Med ; 91: 101155, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36456416

RESUMO

Extracellular vesicles (EVs) are released from all cells in the body, forming an important intercellular communication network that contributes to health and disease. The contents of EVs are cell source-specific, inducing distinct signaling responses in recipient cells. The specificity of EVs and their accumulation in fluid spaces that are accessible for liquid biopsies make them highly attractive as potential biomarkers and therapies for disease. The duality of EVs as favorable (therapeutic) or unfavorable (pathological) messengers is context dependent and remains to be fully determined in homeostasis and various disease states. This review describes the use of EVs as biomarkers, drug delivery vehicles, and regenerative therapeutics, highlighting examples involving viral infections, cancer, and neurological diseases. There is growing interest to provide personalized therapy based on individual patient and disease characteristics. Increasing evidence suggests that EV biomarkers and therapeutic approaches are ideal for personalized medicine due to the diversity and multifunctionality of EVs.


Assuntos
Vesículas Extracelulares , Medicina de Precisão , Humanos , Sistemas de Liberação de Medicamentos , Biomarcadores , Biópsia Líquida
5.
Regen Med ; 17(10): 755-765, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35924471

RESUMO

As it begins to enter the clinic, regenerative medicine has the potential to revolutionize healthcare. Although there exists a growing need for individuals well-versed in the practice of regenerative medicine, few undergraduate institutions offer opportunities to learn about the topic. This article highlights the conception of two novel undergraduate courses in regenerative medicine developed through collaboration between students and faculty at our University to fill this void in the undergraduate curriculum. Lectures from scientists, healthcare professionals, regulatory experts and biotechnology leaders introduced students to regenerative medicine research and the translational process, and a certificate program incorporating relevant coursework and research experience is in development. This pipeline will guide promising undergraduate students to the field of regenerative medicine.


Regenerative medicine is a new medical discipline that aims to restore diseased or damaged tissue back to a healthy state. Stem cells, gene therapies and other regenerative approaches are now being used to treat patients, and, as a result, the field has recently entered the public eye. To implement these cutting-edge therapies, a well-trained workforce is required; however, regenerative medicine education, especially at the undergraduate level, is currently lacking. Faculty and students at our University worked together to address this issue by creating educational offerings that expose undergraduates to the work being done in the field, and opening opportunities for help them to engage in regenerative medicine-related research. Expanded utilization of this approach will encourage talented undergraduates to contribute to the development of safe, effective regenerative therapies.


Assuntos
Medicina Regenerativa , Estudantes , Currículo , Humanos , Medicina Regenerativa/educação
6.
Small ; 18(28): e2201330, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35670145

RESUMO

Current biomarkers for myocardial infarction (MI) diagnosis are typically late markers released upon cell death, incapable of distinguishing between ischemic and reperfusion injury and can be symptoms of other pathologies. Circulating microRNAs (miRNAs) have recently been proposed as alternative biomarkers for MI diagnosis; however, detecting the changes in the human cardiac miRNA profile during MI is extremely difficult. Here, to study the changes in miRNA levels during acute MI, a heart-on-chip model with a cardiac channel, containing human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes in human heart decellularized matrix and collagen, and a vascular channel, containing hiPSC-derived endothelial cells, is developed. This model is exposed to anoxia followed by normoxia to mimic ischemia and reperfusion, respectively. Using a highly sensitive miRNA biosensor that the authors developed, the exact same increase in miR-1, miR-208b, and miR-499 levels in the MI-on-chip and the time-matched human blood plasma samples collected before and after ischemia and reperfusion, is shown. That the surface marker profile of exosomes in the engineered model changes in response to ischemic and reperfusion injury, which can be used as biomarkers to detect MI, is also shown. Hence, the MI-on-chip model developed here can be used in biomarker discovery.


Assuntos
Exossomos , Células-Tronco Pluripotentes Induzidas , MicroRNAs , Infarto do Miocárdio , Traumatismo por Reperfusão , Biomarcadores/metabolismo , Células Endoteliais/metabolismo , Exossomos/metabolismo , Humanos , Hipóxia/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/metabolismo , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Reperfusão , Traumatismo por Reperfusão/diagnóstico
7.
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
8.
J Mol Cell Cardiol ; 164: 1-12, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34774548

RESUMO

BACKGROUND: Heart transplantation, a life-saving approach for patients with end-stage heart disease, is limited by shortage of donor organs. While prolonged storage provides more organs, it increases the extent of ischemia. Therefore, we seek to understand molecular mechanisms underlying pathophysiological changes of donor hearts during prolonged storage. Additionally, considering mesenchymal stromal cell (MSC)-derived paracrine protection, we aim to test if MSC secretome preserves myocardial transcriptome profile and whether MSC secretome from a certain source provides the optimal protection in donor hearts during cold storage. METHODS AND RESULTS: Isolated mouse hearts were divided into: no cold storage (control), 6 h cold storage (6 h-I), 6 h-I + conditioned media from bone marrow MSCs (BM-MSC CM), and 6 h-I + adipose-MSC CM (Ad-MSC CM). Deep RNA sequencing analysis revealed that compared to control, 6 h-I led to 266 differentially expressed genes, many of which were implicated in modulating mitochondrial performance, oxidative stress response, myocardial function, and apoptosis. BM-MSC CM and Ad-MSC CM restored these gene expression towards control. They also improved 6 h-I-induced myocardial functional depression, reduced inflammatory cytokine production, decreased apoptosis, and reduced myocardial H2O2. However, neither MSC-exosomes nor exosome-depleted CM recapitulated MSC CM-ameliorated apoptosis and CM-improved mitochondrial preservation during cold ischemia. Knockdown of Per2 by specific siRNA abolished MSC CM-mediated these protective effects in cardiomyocytes following 6 h cold storage. CONCLUSIONS: Our results demonstrated that using MSC secretome (BM-MSCs and Ad-MSCs) during prolonged cold storage confers preservation of the normal transcriptional "fingerprint", and reduces donor heart damage. MSC-released soluble factors and exosomes may synergistically act for donor heart protection.


Assuntos
Transplante de Coração , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Medula Óssea , Humanos , Peróxido de Hidrogênio/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Secretoma , Doadores de Tecidos , Transcriptoma
9.
Lab Chip ; 21(20): 3876-3887, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34546237

RESUMO

Micro RNAs (miRNAs) have shown great potential as rapid and discriminating biomarkers for acute myocardial infarction (AMI) diagnosis. We have developed a multiplexed ion-exchange membrane-based miRNA (MIX·miR) preconcentration/sensing amplification-free platform for quantifying in parallel a panel of miRNAs, including miR-1, miR-208b, and miR-499, from the same plasma samples from: 1) reference subjects with no evident coronary artery disease (NCAD); 2) subjects with stable coronary artery disease (CAD); and 3) subjects experiencing ST-elevation myocardial infarction (STEMI) prior to (STEMI-pre) and following (STEMI-PCI) percutaneous coronary intervention. The picomolar limit of detection from raw plasma and 3-decade dynamic range of MIX·miR permits detection of the miRNA panel in untreated samples from disease patients and its precise standard curve, provided by large 0.1 to 1 V signals and eliminates individual sensor calibration. The use of molecular concentration feature reduces the assay time to less than 30 minutes and increases the detection sensitivity by bringing all targets close to the sensors. miR-1 was low for NCAD patients but more than one order of magnitude above the normal value for all samples from three categories (CAD, STEMI-pre, and STEMI-PCI) of patients with CAD. In fact, miR-1 expression levels of stable CAD, STEMI-pre and STEMI-PCI are each more than 10-fold higher than the previous class, in that order, well above the 95% confidence level of MIX·miR. Its overexpression estimate is significantly higher than the PCR benchmark. This suggests that, in contrast to protein biomarkers of myocardial injury, miR-1 appears to differentiate ischemia from both reperfusion injury and non-AMI CAD patients. The battery-operated MIX·miR can be a portable and low-cost AMI diagnostic device, particularly useful in settings where cardiac catheterization is not readily available to determine the status of coronary reperfusion.


Assuntos
Doença da Artéria Coronariana , MicroRNAs , Infarto do Miocárdio , Intervenção Coronária Percutânea , Infarto do Miocárdio com Supradesnível do Segmento ST , Doença da Artéria Coronariana/diagnóstico , Doença da Artéria Coronariana/genética , Humanos , MicroRNAs/genética , Infarto do Miocárdio/diagnóstico
10.
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
11.
Stem Cells ; 39(2): 170-182, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33159685

RESUMO

Heart transplantation is a life-saving therapy for end-stage organ failure. Organ deterioration during transportation limits storage to 4 hours, limiting hearts available. Approaches ameliorating organ damage could increase the number of hearts acceptable for transplantation. Prior studies show that adipose-derived stem/stromal cell secretome (ASC-S) rescues tissues from postischemic damage in vivo. This study tested whether ASC-S preserved the function of mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes (iCM) exposed to organ transportation and transplantation conditions. Hearts were subjected to cold University of Wisconsin (UW) cardioplegic solution ± ASC-S for 6 hours followed by analysis using the Langendorff technique. In parallel, the effects of ASC-S on the recovery of iCM from UW solution were examined when provided either during or after cold cardioplegia. Exposure of hearts and iCM to UW deteriorated contractile activity and caused cell apoptosis, worsening in iCM as a function of exposure time; these were ameliorated by augmenting with ASC-S. Silencing of superoxide dismutase 3 and catalase expression prior to secretome generation compromised the ASC-S cardiomyocyte-protective effects. In this study, a novel in vitro iCM model was developed to complement a rodent heart model in assessing efficacy of approaches to improve cardiac preservation. ASC-S displays strong cardioprotective activity on iCM either with or following cold cardioplegia. This effect is associated with ASC-S-mediated cellular clearance of reactive oxygen species. The effect of ASC-S on the temporal recovery of iCM function supports the possibility of lengthening heart storage by augmenting cardioplegic transport solution with ASC-S, expanding the pool of hearts for transplantation.


Assuntos
Soluções Cardioplégicas/toxicidade , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Miócitos Cardíacos/metabolismo , Soluções para Preservação de Órgãos/toxicidade , Recuperação de Função Fisiológica/fisiologia , Adenosina/toxicidade , Alopurinol/toxicidade , Animais , Glutationa/toxicidade , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Insulina/toxicidade , Preparação de Coração Isolado/métodos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Rafinose/toxicidade , Recuperação de Função Fisiológica/efeitos dos fármacos
12.
Front Cell Dev Biol ; 8: 584653, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33102487

RESUMO

We have shown previously that adipose stromal cell (ASC)-derived conditioned media (CM) limited lung injury, endothelial barrier dysfunction, and apoptosis. Here, we used endothelial hyperpermeability and apoptosis assays to investigate how concentration processes affect endothelium-directed bioactivity of ASC-CM and to gain information on the nature of bioactive factors. Comparison of ASC-CM concentrated with differential molecular weight (MW) cutoff filters showed that endothelial barrier protection depended on the species-specific factors in ASC-CM fractionated with MW > 50 kDa. Known barrier regulators-keratin growth factor (KGF), vascular endothelial growth factor (VEGF), and hepatocyte growth factor (HGF)-were detected in ASC-CM fraction of > 100 kDa. Pretreatment of endothelial monolayers with concentrations of KGF, VEGF, and HGF detected in ASC-CM showed that only KGF and HGF protect the endothelium from barrier dysfunction. Depletion of KGF and HGF from ASC-CM attenuated ASC-CM's ability to protect the endothelial barrier. In contrast to barrier-protective factors, apoptosis-protective factors fractionated with MW < 3 kDa and were not species-specific. Application of donors of apoptosis-mitigating gases showed that the CO donor carbon monoxide-releasing molecule 2 (CORM2) protected the endothelium from apoptosis, while the H2S donor NaSH did not. Knockdown of CO-generating heme oxygenase 1 in ASC attenuated ASC-CM's ability to protect the endothelium from apoptosis. We have shown that tumor necrosis factor alpha (TNFα)-induced apoptosis in endothelium is c-Jun N-terminal kinase (JNK)-dependent, and JNK activation is inhibited by ASC-CM pretreatment of endothelial cells. ASC-CM from heme oxygenase 1-depleted ASC displayed attenuated ability to suppress endothelial JNK activation, suggesting that CO-mediated protection of the endothelium from apoptosis is achieved by the downregulation of the JNK pathway. Altogether, our results demonstrate that the concentration of ASC-CM with low MW cutoff filters significantly reduces its anti-apoptotic activity while preserving its barrier-protective activity.

13.
J Transl Med ; 18(1): 285, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32678051

RESUMO

BACKGROUND: Although a large body of information exists relating to cellular therapies, much of this information is either anecdotal or has been obtained from relatively small clinical trials, so that the level of evidence available to direct adoption of therapeutic approaches is quite limited. Despite this, a large number of clinics offer various cellular treatments without having gone through the processes of FDA approval. Florida is considered a "hotspot" of such sites, with a large number of clinics relative to the population. METHODS: To better understand the magnitude and scope of this issue with a specific focus on cardiovascular disease, we surveyed clinics in Florida advertising "cell therapy for heart failure". We identified only 8 clinics that "treat cardiac conditions, including heart failure." Data on administration route, cell type used, dose, success rate, cost, and training of persons performing procedures were collected when available, via email, telephone, or website information. RESULTS: A total of 20,135 patients were identified as treated: 2157 for cardiac conditions. All clinics reported administering cells intravenously, using either adipose- or umbilical-derived sources. Doses ranged from 30 to 150 million cells per treatment. The "success rate" ranged from 65 to 85%, with costs from $6000 to $20,700. Procedures were performed by PAs, MDs, and DOs. CONCLUSION: Large numbers of patients (> 10% of all 20,135 patients) have been and presumably are still are being treated for "cardiac conditions." We conclude that implementation of uniform data collection with an outcome registry, as well as creation of a public database listing FDA-approved cell-based clinical trials, would be useful to patients and the cardiovascular field at large.


Assuntos
Cardiopatias , Transplante de Células-Tronco , Tecido Adiposo , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Células-Tronco Multipotentes
14.
J Clin Med ; 9(1)2020 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-31963368

RESUMO

Over 26 million people worldwide suffer from heart failure, a disease associated with a 1 year mortality rate of 22%. Half of these patients present heart failure with preserved ejection fraction (HFpEF), for which there is no available therapy to improve prognosis. HFpEF is strongly associated with aging, inflammation, and comorbid burden, which are thought to play causal roles in disease development. Mesenchymal stromal/stem cells (MSCs) have potent immunomodulatory actions and promote tissue healing, thus representing an attractive therapeutic option in HFpEF. In this review, we summarize recent data suggesting that a two-hit model of immune dysregulation lies at the heart of the HFpEF. A first hit is represented by genetic mutations associated with clonal hematopoiesis of indeterminate potential (CHIP), which skew immune cells toward a pro-inflammatory phenotype, are associated with HFpEF development in animal models, and with immune dysregulation and risk of HF hospitalization in patients. A second hit is induced by cardiovascular risk factors, which cause subclinical cardiac dysfunction and production of danger signals. In mice, these attract proinflammatory macrophages, Th1 and Th17 cells into the myocardium, where they are required for the development of HFpEF. MSCs have been shown to reduce the pro-inflammatory activity of immune cell types involved in murine HFpEF in vitro, and to reduce myocardial fibrosis and improve diastolic function in vivo, thus they may efficiently target immune dysregulation in HFpEF and stop disease progression.

16.
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.

17.
Stem Cells Dev ; 29(5): 257-262, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31797749

RESUMO

Although adipose tissue and cells show considerable promise for clinical translation in the emerging field of regenerative medicine, they present a challenge to the regulatory community both nationally and internationally. This commentary evaluates the status of adipose-derived therapeutics and considers regulatory approaches designed to maximize patient safety while advancing clinical translation in accordance with evidence-based medical science.


Assuntos
Células Estromais/fisiologia , Tecido Adiposo/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Medicina Regenerativa/métodos
18.
J Gastrointest Surg ; 24(1): 67-75, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31745900

RESUMO

BACKGROUND: No specific therapy exists for acute pancreatitis (AP), and current treatment remains entirely supportive. Adipose stem cells (ASCs) have significant immunomodulatory and regenerative activities. We hypothesized that systemic administration of ASCs would mitigate inflammation in AP. METHODS: AP was induced in mice by 6 hourly intraperitoneal injections of cerulein. Twenty-four hours after AP induction, mice were randomized into four systemic treatment groups: sham group (no acute pancreatitis), vehicle, human ASCs, and human ASC-conditioned media. Mice were sacrificed at 48 h, and blood and organs were collected and analyzed. Pancreatic injury was quantified histologically using a published score (edema, inflammation, and necrosis). Pancreatic inflammation was also studied by immunohistochemistry and PCR. RESULTS: When using IV infusion of Hoechst-labeled ASCs, ASCs were found to localize to inflamed tissues: lungs and pancreas. Mice treated with ASCs had less severe AP, as shown by a significantly decreased histopathology score (edema, inflammation, and necrosis) (p = 0.001). ASCs infusion polarized pancreatic macrophages toward an anti-inflammatory M2 phenotype. ASC-conditioned media reduced pancreatic inflammation similarly to ASCs only, highlighting the importance of ASCs secreted factors in modulating inflammation. CONCLUSION: Intravenous delivery of human ASCs markedly reduces pancreatic inflammation in a murine model of AP ASCs which represent an effective therapy for AP.


Assuntos
Tecido Adiposo/citologia , Pancreatite/terapia , Transplante de Células-Tronco , Células Estromais/transplante , Animais , Ceruletídeo , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pancreatite/etiologia
19.
Restor Neurol Neurosci ; 36(5): 621-627, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30010155

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is devastating, leading to paralysis and death. Disease onset begins pre-symptomatically through spinal motor neuron (MN) axon die-back from musculature at ∼47 days of age in the mutant superoxide dismutase 1 (mSOD1G93A) transgenic ALS mouse model. This period may be optimal to assess potential therapies. We previously demonstrated that post-symptomatic adipose-derived stem cell conditioned medium (ASC-CM) treatment is neuroprotective in mSOD1G93A mice. We hypothesized that early disease onset treatment could ameliorate neuromuscular junction (NMJ) disruption. OBJECTIVE: To determine whether pre-symptom administration of ASC-CM prevents early NMJ disconnection. METHODS: We confirmed the NMJ denervation time course in mSOD1G93A mice using co-labeling of neurofilament and post-synaptic acetylcholine receptors (AchR) by α-bungarotoxin. We determined whether ASC-CM ameliorates early NMJ loss in mSOD1G93A mice by systemically administering 200µl ASC-CM or vehicle medium daily from post-natal days 35 to 47 and quantifying intact NMJs through co-labeling of neurofilament and synaptophysin with α-bungarotoxin in gastrocnemius muscle. RESULTS: Intact NMJs were significantly decreased in 47 day old mSOD1G93A mice (p < 0.05), and daily systemic ASC-CM prevented disease-induced NMJ denervation compared to vehicle treated mice (p < 0.05). CONCLUSIONS: Our results lay the foundation for testing the long-term neurological benefits of systemic ASC-CM therapy in the mSOD1G93A mouse model of ALS.


Assuntos
Adipócitos/metabolismo , Esclerose Lateral Amiotrófica/tratamento farmacológico , Meios de Cultivo Condicionados/farmacologia , Junção Neuromuscular/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Células-Tronco/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos Transgênicos , Molibdoferredoxina , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/inervação , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Junção Neuromuscular/patologia , Junção Neuromuscular/fisiopatologia , Distribuição Aleatória , Receptores Colinérgicos/metabolismo , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo
20.
Am Heart J ; 201: 54-62, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29910056

RESUMO

OBJECTIVES: SENECA (StEm cell iNjECtion in cAncer survivors) is a phase I, randomized, double-blind, placebo-controlled study to evaluate the safety and feasibility of delivering allogeneic mesenchymal stromal cells (allo-MSCs) transendocardially in subjects with anthracycline-induced cardiomyopathy (AIC). BACKGROUND: AIC is an incurable and often fatal syndrome, with a prognosis worse than that of ischemic or nonischemic cardiomyopathy. Recently, cell therapy with MSCs has emerged as a promising new approach to repair damaged myocardium. METHODS: The study population is 36 cancer survivors with a diagnosis of AIC, left ventricular (LV) ejection fraction ≤40%, and symptoms of heart failure (NYHA class II-III) on optimally-tolerated medical therapy. Subjects must be clinically free of cancer for at least two years with a ≤ 30% estimated five-year risk of recurrence. The first six subjects participated in an open-label, lead-in phase and received 100 million allo-MSCs; the remaining 30 will be randomized 1:1 to receive allo-MSCs or vehicle via 20 transendocardial injections. Efficacy measures (obtained at baseline, 6 months, and 12 months) include MRI evaluation of LV function, LV volumes, fibrosis, and scar burden; assessment of exercise tolerance (six-minute walk test) and quality of life (Minnesota Living with Heart Failure Questionnaire); clinical outcomes (MACE and cumulative days alive and out of hospital); and biomarkers of heart failure (NT-proBNP). CONCLUSIONS: This is the first clinical trial using direct cardiac injection of cells for the treatment of AIC. If administration of allo-MSCs is found feasible and safe, SENECA will pave the way for larger phase II/III studies with therapeutic efficacy as the primary outcome.


Assuntos
Antraciclinas/efeitos adversos , Sobreviventes de Câncer/estatística & dados numéricos , Insuficiência Cardíaca/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Neoplasias/tratamento farmacológico , Qualidade de Vida , Função Ventricular Esquerda/fisiologia , Adolescente , Adulto , Idoso , Antraciclinas/uso terapêutico , Método Duplo-Cego , Estudos de Viabilidade , Feminino , Seguimentos , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...