Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Genome Med ; 15(1): 61, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37563727

RESUMEN

BACKGROUND: The immune response is a crucial factor for mediating the benefit of cardiac cell therapies. Our previous research showed that cardiomyocyte transplantation alters the cardiac immune response and, when combined with short-term pharmacological CCR2 inhibition, resulted in diminished functional benefit. However, the specific role of innate immune cells, especially CCR2 macrophages on the outcome of cardiomyocyte transplantation, is unclear. METHODS: We compared the cellular, molecular, and functional outcome following cardiomyocyte transplantation in wildtype and T cell- and B cell-deficient Rag2del mice. The cardiac inflammatory response was assessed using flow cytometry. Gene expression profile was assessed using single-cell and bulk RNA sequencing. Cardiac function and morphology were determined using magnetic resonance tomography and immunohistochemistry respectively. RESULTS: Compared to wildtype mice, Rag2del mice show an increased innate immune response at steady state and disparate macrophage response after MI. Subsequent single-cell analyses after MI showed differences in macrophage development and a lower prevalence of CCR2 expressing macrophages. Cardiomyocyte transplantation increased NK cells and monocytes, while reducing CCR2-MHC-IIlo macrophages. Consequently, it led to increased mRNA levels of genes involved in extracellular remodelling, poor graft survival, and no functional improvement. Using machine learning-based feature selection, Mfge8 and Ccl7 were identified as the primary targets underlying these effects in the heart. CONCLUSIONS: Our results demonstrate that the improved functional outcome following cardiomyocyte transplantation is dependent on a specific CCR2 macrophage response. This work highlights the need to study the role of the immune response for cardiomyocyte cell therapy for successful clinical translation.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Ratones , Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Macrófagos/metabolismo , Monocitos/metabolismo , Ratones Endogámicos C57BL
2.
Int J Mol Sci ; 23(16)2022 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-36012110

RESUMEN

Ventricular arrhythmias associated with myocardial infarction (MI) have a significant impact on mortality in patients following heart attack. Therefore, targeted reduction of arrhythmia represents a therapeutic approach for the prevention and treatment of severe events after infarction. Recent research transplanting mesenchymal stem cells (MSC) showed their potential in MI therapy. Our study aimed to investigate the effects of MSC injection on post-infarction arrhythmia. We used our murine double infarction model, which we previously established, to more closely mimic the clinical situation and intramyocardially injected hypoxic pre-conditioned murine MSC to the infarction border. Thereafter, various types of arrhythmias were recorded and analyzed. We observed a homogenous distribution of all types of arrhythmias after the first infarction, without any significant differences between the groups. Yet, MSC therapy after double infarction led to a highly significant reduction in simple and complex arrhythmias. Moreover, RNA-sequencing of samples from stem cell treated mice after re-infarction demonstrated a significant decline in most arrhythmias with reduced inflammatory pathways. Additionally, following stem-cell therapy we found numerous highly expressed genes to be either linked to lowering the risk of heart failure, cardiomyopathy or sudden cardiac death. Moreover, genes known to be associated with arrhythmogenesis and key mutations underlying arrhythmias were downregulated. In summary, our stem-cell therapy led to a reduction in cardiac arrhythmias after MI and showed a downregulation of already established inflammatory pathways. Furthermore, our study reveals gene regulation pathways that have a potentially direct influence on arrhythmogenesis after myocardial infarction.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Infarto del Miocardio , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/terapia , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas/metabolismo , Ratones , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia
3.
Cells ; 10(12)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34943774

RESUMEN

Stem/progenitor cell transplantation is a potential novel therapeutic strategy to induce angiogenesis in ischemic tissue, which can prevent major amputation in patients with advanced peripheral artery disease (PAD). Thus, clinicians can use cell therapies worldwide to treat PAD. However, some cell therapy studies did not report beneficial outcomes. Clinical researchers have suggested that classical risk factors and comorbidities may adversely affect the efficacy of cell therapy. Some studies have indicated that the response to stem cell therapy varies among patients, even in those harboring limited risk factors. This suggests the role of undetermined risk factors, including genetic alterations, somatic mutations, and clonal hematopoiesis. Personalized stem cell-based therapy can be developed by analyzing individual risk factors. These approaches must consider several clinical biomarkers and perform studies (such as genome-wide association studies (GWAS)) on disease-related genetic traits and integrate the findings with those of transcriptome-wide association studies (TWAS) and whole-genome sequencing in PAD. Additional unbiased analyses with state-of-the-art computational methods, such as machine learning-based patient stratification, are suited for predictions in clinical investigations. The integration of these complex approaches into a unified analysis procedure for the identification of responders and non-responders before stem cell therapy, which can decrease treatment expenditure, is a major challenge for increasing the efficacy of therapies.


Asunto(s)
Inteligencia Artificial/tendencias , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Enfermedad Arterial Periférica/terapia , Transcriptoma/genética , Estudio de Asociación del Genoma Completo/tendencias , Humanos , Enfermedad Arterial Periférica/genética , Medicina de Precisión/tendencias , Factores de Riesgo , Secuenciación Completa del Genoma/tendencias
4.
Sci Rep ; 11(1): 11625, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34079005

RESUMEN

Novel therapeutic strategies aiming at improving the healing process after an acute myocardial infarction are currently under intense investigation. The mouse model plays a central role for deciphering the underlying mechanisms on a molecular and cellular level. Therefore, we intended to assess in-vivo post-infarct remodeling as comprehensively as possible using an expedient native magnetic resonance imaging (MRI) in the two most prominent infarct models, permanent ligation (PL) of the left anterior descending artery (LAD) versus ischemia reperfusion (I/R). Mice were subjected to either permanent or transient (45 min) occlusion of the LAD. After 3 weeks, examinations were performed with a 7-Tesla small animal MRI system. Data analysis was performed with the freely available software Segment. PL resulted in a massive dilation of the left ventricle, accompanied by hypertrophy of the non-infarcted myocardium and a decline of contractile function. These effects were less pronounced following I/R compared to healthy animals. Single plane assessments were not sufficient to capture the specific differences of left ventricular (LV) properties between the two infarct models. Bulls-eye plots were found to be an ideal tool for qualitative LV wall assessment, whereas a multi-slice sector-based analysis of wall regions is ideal to determine differences in hypertrophy, lateral wall thinning and wall thickening on a quantitative level. We combine the use of polar map-based analysis of LV wall properties with volumetric measurements using simple CINE CMR imaging. Our strategy represents a versatile and easily available tool for serial assessment of the LV during the remodeling process. Our study contributes to a better understanding of the effects of novel therapies targeting the healing of damaged myocardium.


Asunto(s)
Trastornos Cerebrovasculares/diagnóstico por imagen , Ventrículos Cardíacos/diagnóstico por imagen , Corazón/diagnóstico por imagen , Infarto de la Arteria Cerebral Anterior/diagnóstico por imagen , Infarto del Miocardio/diagnóstico por imagen , Daño por Reperfusión/diagnóstico por imagen , Animales , Trastornos Cerebrovasculares/fisiopatología , Modelos Animales de Enfermedad , Corazón/fisiopatología , Ventrículos Cardíacos/fisiopatología , Infarto de la Arteria Cerebral Anterior/fisiopatología , Ligadura/métodos , Imagen por Resonancia Cinemagnética/métodos , Ratones , Infarto del Miocardio/fisiopatología , Daño por Reperfusión/fisiopatología , Factores de Tiempo , Remodelación Ventricular
6.
Cells ; 9(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32756334

RESUMEN

We investigated the influence of syngeneic cardiomyocyte transplantation after myocardial infarction (MI) on the immune response and cardiac function. Methods and Results: We show for the first time that the immune response is altered as a result of syngeneic neonatal cardiomyocyte transplantation after MI leading to improved cardiac pump function as observed by magnetic resonance imaging in C57BL/6J mice. Interestingly, there was no improvement in the capillary density as well as infarct area as observed by CD31 and Sirius Red staining, respectively. Flow cytometric analysis revealed a significantly different response of monocyte-derived macrophages and regulatory T cells after cell transplantation. Interestingly, the inhibition of monocyte infiltration accompanied by cardiomyocyte transplantation diminished the positive effect of cell transplantation alone. The number of CD68+ macrophages in the remote area of the heart observed after four weeks was also different between the groups. Transcriptome analysis showed several changes in the gene expression involving circadian regulation, mitochondrial metabolism and immune responses after cardiomyocyte transplantation. Conclusion: Our work shows that cardiomyocyte transplantation alters the immune response after myocardial infarction with the recruited monocytes playing a role in the beneficial effect of cell transplantation. It also paves the way for further optimization of the efficacy of cardiomyocyte transplantation and their successful translation in the clinic.


Asunto(s)
Infarto del Miocardio/terapia , Miocardio/inmunología , Miocitos Cardíacos/trasplante , Animales , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Corazón/fisiología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/inmunología , Receptores CCR2/inmunología , Linfocitos T Reguladores/inmunología
7.
EBioMedicine ; 57: 102862, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32629392

RESUMEN

BACKGROUND: Bone marrow stem cell clonal dysfunction by somatic mutation is suspected to affect post-infarction myocardial regeneration after coronary bypass surgery (CABG). METHODS: Transcriptome and variant expression analysis was studied in the phase 3 PERFECT trial post myocardial infarction CABG and CD133+ bone marrow derived hematopoetic stem cells showing difference in left ventricular ejection fraction (∆LVEF) myocardial regeneration Responders (n=14; ∆LVEF +16% day 180/0) and Non-responders (n=9; ∆LVEF -1.1% day 180/0). Subsequently, the findings have been validated in an independent patient cohort (n=14) as well as in two preclinical mouse models investigating SH2B3/LNK antisense or knockout deficient conditions. FINDINGS: 1. Clinical: R differed from NR in a total of 161 genes in differential expression (n=23, q<0•05) and 872 genes in coexpression analysis (n=23, q<0•05). Machine Learning clustering analysis revealed distinct RvsNR preoperative gene-expression signatures in peripheral blood acorrelated to SH2B3 (p<0.05). Mutation analysis revealed increased specific variants in RvsNR. (R: 48 genes; NR: 224 genes). 2. Preclinical:SH2B3/LNK-silenced hematopoietic stem cell (HSC) clones displayed significant overgrowth of myeloid and immune cells in bone marrow, peripheral blood, and tissue at day 160 after competitive bone-marrow transplantation into mice. SH2B3/LNK-/- mice demonstrated enhanced cardiac repair through augmenting the kinetics of bone marrow-derived endothelial progenitor cells, increased capillary density in ischemic myocardium, and reduced left ventricular fibrosis with preserved cardiac function. 3. VALIDATION: Evaluation analysis in 14 additional patients revealed 85% RvsNR (12/14 patients) prediction accuracy for the identified biomarker signature. INTERPRETATION: Myocardial repair is affected by HSC gene response and somatic mutation. Machine Learning can be utilized to identify and predict pathological HSC response. FUNDING: German Ministry of Research and Education (BMBF): Reference and Translation Center for Cardiac Stem Cell Therapy - FKZ0312138A and FKZ031L0106C, German Ministry of Research and Education (BMBF): Collaborative research center - DFG:SFB738 and Center of Excellence - DFG:EC-REBIRTH), European Social Fonds: ESF/IV-WM-B34-0011/08, ESF/IV-WM-B34-0030/10, and Miltenyi Biotec GmbH, Bergisch-Gladbach, Germany. Japanese Ministry of Health : Health and Labour Sciences Research Grant (H14-trans-001, H17-trans-002) TRIAL REGISTRATION: ClinicalTrials.gov NCT00950274.


Asunto(s)
Antígeno AC133/genética , Trasplante de Médula Ósea/métodos , Enfermedad de la Arteria Coronaria/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Isquemia Miocárdica/terapia , Adolescente , Adulto , Anciano , Células de la Médula Ósea/citología , Senescencia Celular/genética , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/fisiopatología , Femenino , Corazón/crecimiento & desarrollo , Corazón/fisiopatología , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Persona de Mediana Edad , Isquemia Miocárdica/genética , Isquemia Miocárdica/patología , Regeneración/genética , Adulto Joven
8.
Biomedicines ; 8(6)2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32545336

RESUMEN

Several cell populations derived from bone marrow (BM) have been shown to possess cardiac regenerative potential. Among these are freshly isolated CD133+ hematopoietic as well as culture-expanded mesenchymal stem cells. Alternatively, by purifying CD271+ cells from BM, mesenchymal progenitors can be enriched without an ex vivo cultivation. With regard to the limited available number of freshly isolated BM-derived stem cells, the effect of the dosage on the therapeutic efficiency is of particular interest. Therefore, in the present pre-clinical study, we investigated human BM-derived CD133+ and CD271+ stem cells for their cardiac regenerative potential three weeks post-myocardial infarction (MI) in a dose-dependent manner. The improvement of the hemodynamic function as well as cardiac remodeling showed no therapeutic difference after the transplantation of both 100,000 and 500,000 stem cells. Therefore, beneficial stem cell transplantation post-MI is widely independent of the cell dose and detrimental stem cell amplification in vitro can likely be avoided.

9.
Cells ; 9(6)2020 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-32486211

RESUMEN

Angiogenesis plays a central role in the healing process following acute myocardial infarction. The PET tracer [68Ga]-NODAGA-RGD, which is a ligand for the αvß3 integrin, has been investigated for imaging angiogenesis in the process of healing myocardium in both animal and clinical studies. It´s value as a prognostic marker of functional outcome remains unclear. Therefore, the aim of this work was to establish [68Ga]-NODAGA-RGD for imaging angiogenesis in the murine infarct model and evaluate the tracer as a predictor for cardiac remodeling in the context of cardiac stem cell therapy. [68Ga]-NODAGA-RGD PET performed seven days after left anterior descending coronary artery (LAD) occlusion in 129S6 mice showed intense tracer accumulation within the infarct region. The specificity was shown in a sub-group of animals by application of the competitive inhibitor cilengitide prior to tracer injection in a subgroup of animals. Myocardial infarction (MI) significantly reduced cardiac function and resulted in pronounced left ventricular remodeling after three weeks, as measured by cardiac MRI in a separate group. Cardiac induced cells (CiC) that were derived from mESC injected intramyocardially in the therapy group significantly improved left ventricular ejection fraction (LVEF). Surprisingly, CiC transplantation resulted in significantly lower tracer accumulation seven days after MI induction. Accordingly, we successfully established the PET tracer [68Ga]-NODAGA-RGD for the assessment of αvß3 integrin expression in the healing process after MI in the mouse model. Yet, our results indicate that the mere extent of angiogenesis following MI does not serve as a sufficient prognostic marker for functional outcome.


Asunto(s)
Acetatos/química , Radioisótopos de Galio/química , Compuestos Heterocíclicos con 1 Anillo/química , Infarto del Miocardio/diagnóstico por imagen , Neovascularización Fisiológica , Oligopéptidos/química , Tomografía de Emisión de Positrones , Trasplante de Células Madre , Remodelación Ventricular , Animales , Integrina alfaVbeta3/metabolismo , Imagen por Resonancia Magnética , Ratones , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia
10.
Adv Drug Deliv Rev ; 165-166: 96-104, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32305352

RESUMEN

In biomedical research, enormous progress is being made and new candidates for putative medicinal products emerge. However, most published preclinical data are not conducted according to the standard Good Laboratory Practice (GLP). GLP is mandatory for preclinical analysis of Advanced Therapy Medicinal Products (ATMP) and thereby a prerequisite for planning and conduction of clinical trials. Not inconsiderable numbers of clinical trials are terminated earlier or fail - do inadequate testing strategies or missing specialized assays during the preclinical development contribute to this severe complex of problems? Unfortunately, there is also a lack of access to GLP testing results and OECD (Organisation for Economic Co-operation and Development) GLP guidelines are not yet adjusted to ATMP specialties. Ultimately, GLP offers possibilities to generate reliable and reproducible data. Therefore, this review elucidates different GLP aspects in drug development, speculates on reasons of putative low GLP acceptance in the scientific community and mentions solution proposals.


Asunto(s)
Desarrollo de Medicamentos/organización & administración , Descubrimiento de Drogas/organización & administración , Evaluación Preclínica de Medicamentos/métodos , Laboratorios/organización & administración , Enfermedades Cardiovasculares/tratamiento farmacológico , Desarrollo de Medicamentos/normas , Descubrimiento de Drogas/normas , Evaluación Preclínica de Medicamentos/normas , Guías como Asunto , Humanos , Laboratorios/normas
11.
Cells ; 8(12)2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31835854

RESUMEN

Cellular inflammation following acute myocardial infarction has gained increasing importance as a target mechanism for therapeutic approaches. We sought to investigate the effect of syngeneic cardiac induced cells (CiC) on myocardial inflammation using 18F-FDG PET (Positron emission tomography)-based imaging and the resulting effect on cardiac pump function using cardiac magnetic resonance (CMR) imaging in a mouse model of myocardial infarction. Mice underwent permanent left anterior descending coronary artery (LAD) ligation inducing an acute inflammatory response. The therapy group received an intramyocardial injection of 106 CiC into the border zone of the infarction. Five days after myocardial infarction, 18F-FDG PET was performed under anaesthesia with ketamine and xylazine (KX) to image the inflammatory response in the heart. Flow cytometry of the mononuclear cells in the heart was performed to analyze the inflammatory response. The effect of CiC therapy on cardiac function was determined after three weeks by CMR. The 18F-FDG PET imaging of the heart five days after myocardial infarction (MI) revealed high focal tracer accumulation in the border zone of the infarcted myocardium, whereas no difference was observed in the tracer uptake between infarct and remote myocardium. The CiC transplantation induced a shift in 18F-FDG uptake pattern, leading to significantly higher 18F-FDG uptake in the whole heart, as well as the remote area of the heart. Correspondingly, high numbers of CD11+ cells could be measured by flow cytometry in this region. The CiC transplantation significantly improved the left ventricular ejection function (LVEF) three weeks after myocardial infarction. The CiC transplantation after myocardial infarction leads to an improvement in pump function through modulation of the cellular inflammatory response five days after myocardial infarction. By combining CiC transplantation and the cardiac glucose uptake suppression protocol with KX in a mouse model, we show for the first time, that imaging of cellular inflammation after myocardial infarction using 18F-FDG PET can be used as an early prognostic tool for assessing the efficacy of cardiac stem cell therapies.


Asunto(s)
Antígenos CD11/metabolismo , Fluorodesoxiglucosa F18/administración & dosificación , Corazón/diagnóstico por imagen , Células Madre Embrionarias de Ratones/trasplante , Infarto del Miocardio/terapia , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Corazón/fisiopatología , Humanos , Imagen por Resonancia Cinemagnética , Ratones , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/inmunología , Infarto del Miocardio/fisiopatología , Tomografía de Emisión de Positrones , Resultado del Tratamiento , Función Ventricular Izquierda
12.
Cells ; 8(12)2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31757119

RESUMEN

BACKGROUND: Ventricular arrhythmias (VA) are a common cause of sudden death after myocardial infarction (MI). Therefore, developing new therapeutic methods for the prevention and treatment of VA is of prime importance. METHODS: Human bone marrow derived CD271+ mesenchymal stem cells (MSC) were tested for their antiarrhythmic effect. This was done through the development of a novel mouse model using an immunocompromised Rag2-/- γc-/- mouse strain subjected to myocardial "infarction-reinfarction". The mice underwent a first ischemia-reperfusion through the left anterior descending (LAD) artery closure for 45 minutes with a subsequent second permanent LAD ligation after seven days from the first infarct. RESULTS: This mouse model induced various types of VA detected with continuous electrocardiogram (ECG) monitoring via implanted telemetry device. The immediate intramyocardial delivery of CD271+ MSC after the first MI significantly reduced VA induced after the second MI. CONCLUSIONS: In addition to the clinical relevance, more closely reflecting patients who suffer from severe ischemic heart disease and related arrhythmias, our new mouse model bearing reinfarction warrants the time required for stem cell engraftment and for the first time enables us to analyze and verify significant antiarrhythmic effects of human CD271+ stem cells in vivo.


Asunto(s)
Adapaleno/inmunología , Antiarrítmicos/uso terapéutico , Modelos Animales de Enfermedad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/terapia , Adapaleno/análisis , Animales , Femenino , Humanos , Inmunofenotipificación , Ratones , Ratones Noqueados
13.
Ann Thorac Surg ; 108(6): 1913-1921, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31398356

RESUMEN

BACKGROUND: Although the safety and feasibility of combined coronary artery bypass grafting (CABG) and bone marrow stem cell (BMSC) transplantation have been proven, the efficacy of this approach remains controversial. Therefore, we conducted an updated meta-analysis of randomized controlled trials to evaluate the efficacy of this procedure. METHODS: Electronic databases were systematically searched for randomized trials comparing 4-month to 6-month follow-up outcomes in patients who underwent isolated CABG (CABG group) and patients who received BMSC transplantation with CABG (BMSC group). A random-effects meta-analysis was conducted across eligible studies. Meta-regression and subgroup analyses were utilized to identify sources of data heterogeneity. RESULTS: Thirteen trials were eligible, with a total number of 292 patients in the BMSC group and 247 patients in the CABG group. Compared with the CABG group, the BMSC group showed significant improvement of follow-up left ventricular ejection fraction (n = 539, 4.8%; 95% confidence interval [CI], 2.3%-7.3%; P = .001). The analyzed data showed significant heterogeneity (I2 = 74.2%, P < .001). The reduction in scar size (n = 120; -2.2 mL; 95% CI, -18.2 mL to 13.7 mL; P = .44) and the improvement in the 6-minute walk test (n = 212; 41 m; 95% CI, -13 m to 95 m; P = .10) did not reach statistical significance. No significant correlation was found between the number of the injected BMSCs or the method of injection and the change in ejection fraction. CONCLUSIONS: The present evidence suggests that combined CABG and BMSC transplantation is associated with improvement of left ventricular ejection fraction. However, the heterogeneity in the data suggests variations in patient response to this therapy. Further studies are required to understand these variations.


Asunto(s)
Trasplante de Médula Ósea/métodos , Puente de Arteria Coronaria/métodos , Isquemia Miocárdica/cirugía , Trasplante de Células Madre/métodos , Humanos , Isquemia Miocárdica/fisiopatología , Función Ventricular Izquierda
14.
Cell Physiol Biochem ; 53(2): 337-354, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31373783

RESUMEN

BACKGROUND/AIMS: The availability of truly maturated cardiomyocytic subtypes is a major prerequisite for cardiovascular cell replacement therapies. Pluripotent stem cells provide a suitable source for the development of new strategies to overcome enormous hurdles such as yield, purity and safety of in vitro generated cells. METHODS: To address these issues, we have refined existing forward programming protocols by combining forced exogenous overexpression of the early cardiovascular transcription factor Nkx2.5 with a αMHC-promoter-based antibiotic selection step. Additionally, we applied small molecules such as ascorbic acid to enhance cardiomyogenic differentiation efficiency. Subsequently, we evaluated the cell fate of the resulting cardiomyocytes on the mRNA as well as protein levels. The latter was performed using high-resolution confocal microscopy. Furthermore, we examined the response of the cells` beating activities to pharmacological substance administration. RESULTS: Our results reveal an apparent influence of Nkx2.5 on the cell fate of ESC-derived cardiomyocytes. Resulting single cells exhibit characteristics of early ventricular cardiomyocytes, such as sarcomeric marker expression, spontaneous beating frequency, and distinct L-type calcium channel occurrence. CONCLUSION: Therefore, we demonstrate cardiovascular subtype forward programming of ESCs using a combination of transcription factors along with small molecule administration. However, our findings also underline current assumptions, that a terminal maturation of PSC derived cardiomyocytes in vitro is still an unsolved problem which urgently needs to be addressed in the field.


Asunto(s)
Reprogramación Celular , Células Madre Embrionarias/metabolismo , Proteína Homeótica Nkx-2.5/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Ácido Ascórbico/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Proteína Homeótica Nkx-2.5/antagonistas & inhibidores , Proteína Homeótica Nkx-2.5/genética , Ratones , Microscopía Confocal , Miocitos Cardíacos/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Verapamilo/farmacología
15.
Cell Mol Life Sci ; 76(9): 1681-1695, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30721319

RESUMEN

Despite significant developments in medical and surgical strategies, cardiac diseases remain the leading causes of morbidity and mortality worldwide. Numerous studies involving preclinical and clinical trials have confirmed that stem cell transplantation can help improve cardiac function and regenerate damaged cardiac tissue, and stem cells isolated from bone marrow, heart tissue, adipose tissue and umbilical cord are the primary candidates for transplantation. During the past decade, menstrual blood-derived endometrial stem cells (MenSCs) have gradually become a promising alternative for stem cell-based therapy due to their comprehensive advantages, which include their ability to be periodically and non-invasively collected, their abundant source material, their ability to be regularly donated, their superior proliferative capacity and their ability to be used for autologous transplantation. MenSCs have shown positive therapeutic potential for the treatment of various diseases. Therefore, aside from a brief introduction of the biological characteristics of MenSCs, this review focuses on the progress being made in evaluating the functional improvement of damaged cardiac tissue after MenSC transplantation through preclinical and clinical studies. Based on published reports, we conclude that the paracrine effect, transdifferentiation and immunomodulation by MenSC promote both regeneration of damaged myocardium and improvement of cardiac function.


Asunto(s)
Rehabilitación Cardiaca/métodos , Enfermedades Cardiovasculares/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Endometrio/citología , Trasplante de Células Madre , Células Madre/citología , Adulto , Anciano , Transdiferenciación Celular/fisiología , Femenino , Humanos , Masculino , Menstruación/sangre , Adulto Joven
16.
Cells ; 9(1)2019 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-31892273

RESUMEN

BACKGROUND: Bone marrow (BM)-derived stem cells with their various functions and characteristics have become a well-recognized source for the cell-based therapies. However, knowledge on their therapeutic potential and the shortage for a cross-link between distinct BM-derived stem cells, primed after the onset of myocardial infarction (MI), seems to be still rudimentary. Therefore, the post-examination of the therapeutic characteristics of such primed hematopoietic CD133+ and mesenchymal CD271+ stem cells was the object of the present study. METHODS AND RESULTS: The effects of respective CD133+ and CD271+ mononuclear cells alone as well as in the co-culture model have been explored with focus on their angiogenic potential. The phenotypic analysis revealed a small percentage of isolated cells expressing both surface markers. Moreover, target stem cells isolated with our standardized immunomagnetic isolation procedure did not show any negative alterations following BM storage in regard to cell numbers and/or quality. In vitro network formation relied predominantly on CD271+ stem cells when compared with single CD133+ culture. Interestingly, CD133+ cells contributed in the tube formation, only if they were cultivated in combination with CD271+ cells. Additional to the in vitro examination, therapeutic effects of the primed stem cells were investigated 48 h post MI in a murine model. Hence, we have found a lower expression of transforming growth factor ßeta 3 (TGFß3) as well as an increase of the proangiogenic factors after CD133+ cell treatment in contrast to CD271+ cell treatment. On the other hand, the CD271+ cell therapy led to a lower expression of the inflammatory cytokines. CONCLUSION: The interactions between CD271+ and CD133+ subpopulations the extent to which the combination may enhance cardiac regeneration has still not been investigated so far. We expect that the multiple characteristics and various regenerative effects of CD271+ cells alone as well as in combination with CD133+ will result in an improved therapeutic impact on ischemic heart disease.


Asunto(s)
Antígeno AC133/metabolismo , Adapaleno/metabolismo , Diferenciación Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Neovascularización Fisiológica , Animales , Biomarcadores , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Inmunofenotipificación , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Ratones Transgénicos , Infarto del Miocardio/etiología , Regeneración
17.
J Vis Exp ; (136)2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29985305

RESUMEN

While CD133+ hematopoietic stem cells (SCs) have been proven to provide high potential in the field of regenerative medicine, their low retention rates after injection into injured tissues as well as the observed massive cell death rates lead to very restricted therapeutic effects. To overcome these limitations, we sought to establish a non-viral based protocol for suitable cell engineering prior to their administration. The modification of human CD133+ expressing SCs using microRNA (miR) loaded magnetic polyplexes was addressed with respect to uptake efficiency and safety as well as the targeting potential of the cells. Relying on our protocol, we can achieve high miR uptake rates of 80-90% while the CD133+ stem cell properties remain unaffected. Moreover, these modified cells offer the option of magnetic targeting. We describe here a safe and highly efficient procedure for the modification of CD133+ SCs. We expect this approach to provide a standard technology for optimization of therapeutic stem cell effects and for monitoring of the administered cell product via magnetic resonance imaging (MRI).


Asunto(s)
Células de la Médula Ósea/metabolismo , Ingeniería Celular/métodos , Células Madre Hematopoyéticas/metabolismo , MicroARNs/metabolismo , Adulto , Células de la Médula Ósea/citología , Humanos , Transfección
18.
Dis Model Mech ; 11(6)2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29752300

RESUMEN

Ischemic heart failure is the leading cause of mortality worldwide. An early boost of intracardiac regenerative key mechanisms and angiogenetic niche signaling in cardiac mesenchymal stem cells (MSCs) could improve myocardial infarction (MI) healing. Epicardial erythropoietin (EPO; 300 U kg-1) was compared with intraperitoneal and intramyocardial EPO treatments after acute MI in rats (n=156). Real-time PCR and confocal microscopy revealed that epicardial EPO treatment enhanced levels of intracardiac regenerative key indicators (SDF-1, CXCR4, CD34, Bcl-2, cyclin D1, Cdc2 and MMP2), induced transforming growth factor ß (TGF-ß)/WNT signaling in intramyocardial MSC niches through the direct activation of AKT and upregulation of upstream signals FOS and Fzd7, and augmented intracardiac mesenchymal proliferation 24 h after MI. Cardiac catheterization and tissue analysis showed superior cardiac functions, beneficial remodeling and increased capillary density 6 weeks after MI. Concomitant fluorescence-activated cell sorting, co-cultures with neonatal cardiomyocytes, angiogenesis assays, ELISA, western blotting and RAMAN spectroscopy demonstrated that EPO could promote cardiomyogenic differentiation that was specific of tissue origin and enhance paracrine angiogenetic activity in cardiac CD45-CD44+DDR2+ MSCs. Epicardial EPO delivery might be the optimal route for efficient upregulation of regenerative key signals after acute MI. Early EPO-mediated stimulation of mesenchymal proliferation, synergistic angiogenesis with cardiac MSCs and direct induction of TGF-ß/WNT signaling in intramyocardial cardiac MSCs could initiate an accelerated healing process that enhances cardiac recovery.


Asunto(s)
Eritropoyetina/uso terapéutico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/terapia , Miocardio/patología , Neovascularización Fisiológica , Pericardio/metabolismo , Enfermedad Aguda , Animales , Antígenos CD/metabolismo , Capilares/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Pruebas de Función Cardíaca , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Mesodermo/patología , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Regeneración/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
19.
Stem Cells Int ; 2018: 1909346, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29535769

RESUMEN

During the past decades, stem cell-based therapy has acquired a promising role in regenerative medicine. The application of novel cell therapeutics for the treatment of cardiovascular diseases could potentially achieve the ambitious aim of effective cardiac regeneration. Despite the highly positive results from preclinical studies, data from phase I/II clinical trials are inconsistent and the improvement of cardiac remodeling and heart performance was found to be quite limited. The major issues which cardiac stem cell therapy is facing include inefficient cell delivery to the site of injury, accompanied by low cell retention and weak effectiveness of remaining stem cells in tissue regeneration. According to preclinical and clinical studies, various stem cells (adult stem cells, embryonic stem cells, and induced pluripotent stem cells) represent the most promising cell types so far. Beside the selection of the appropriate cell type, researchers have developed several strategies to produce "second-generation" stem cell products with improved regenerative capacity. Genetic and nongenetic modifications, chemical and physical preconditioning, and the application of biomaterials were found to significantly enhance the regenerative capacity of transplanted stem cells. In this review, we will give an overview of the recent developments in stem cell engineering with the goal to facilitate stem cell delivery and to promote their cardiac regenerative activity.

20.
Cells Tissues Organs ; 206(1-2): 35-45, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30630170

RESUMEN

In-depth knowledge of the mechanisms induced by early postischemic cardiac endogenous mesenchymal stem cells (MSCs) in the acutely ischemic heart could advance our understanding of cardiac regeneration. Herein, we aimed to identify, isolate, and initially characterize the origin, kinetics and fate of cardiac MSCs. This was facilitated by in vivo genetic cell fate mapping through green fluorescent protein (GFP) expression under the control of vimentin induction after acute myocardial infarction (MI). Following permanent ligation of the left anterior descending coronary artery in CreER+ mTom/mGFP+ mice, vimentin/GFP+ cells revealed ischemia-responsive activation, survival, and local enrichment inside the peri-infarction border zone. Fluorescence-activated cell sorting (FACS)-isolated vimentin/GFP+ cells could be strongly expanded in vitro with clonogenic precursor formation and revealed MSC-typical cell morphology. Flow-cytometric analyses demonstrated an increase in cardiac vimentin/GFP+ cells in the ischemic heart, from a 0.6% cardiac mononuclear cell (MNC) fraction at 24 h to 1.6% at 72 h following MI. Sca-1+CD45- cells within the vimentin/GFP+ subtype of this MNC fraction increased from 35.2% at 24 h to 74.6% at 72 h after MI. The cardiac postischemic vimentin/GFP+ MNC subtype showed multipotent adipogenic, chondrogenic, and osteogenic differentiation potential, which is distinctive for MSCs. In conclusion, we demonstrated a seemingly proliferative first response of vimentin- induced cardiac endogenous MSCs in the acutely ischemic heart. Genetically, GFP-targeted in vivo cell tracking, isolation, and in vitro expansion of this cardiac MSC subtype could help to clarify their reparative status in inflammation, fibrogenesis, cell turnover, tissue homeostasis, and myocardial regeneration.


Asunto(s)
Células Madre Mesenquimatosas/citología , Infarto del Miocardio/patología , Miocardio/patología , Vimentina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Separación Celular , Supervivencia Celular , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/metabolismo , Antígenos Comunes de Leucocito/análisis , Antígenos Comunes de Leucocito/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Vimentina/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA