Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomedicines ; 8(6)2020 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-32545336

RESUMO

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.

2.
Sci Rep ; 7(1): 9755, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28852100

RESUMO

Different subtypes of bone marrow-derived stem cells are characterized by varying functionality and activity after transplantation into the infarcted heart. Improvement of stem cell therapeutics requires deep knowledge about the mechanisms that mediate the benefits of stem cell treatment. Here, we demonstrated that co-transplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) led to enhanced synergistic effects on cardiac remodeling. While HSCs were associated with blood vessel formation, MSCs were found to possess transdifferentiation capacity. This cardiomyogenic plasticity of MSCs was strongly promoted by a gap junction-dependent crosstalk between myocytes and stem cells. The inhibition of cell-cell coupling significantly reduced the expression of the cardiac specific transcription factors NKX2.5 and GATA4. Interestingly, we observed that small non-coding RNAs are exchanged between MSCs and cardiomyocytes in a GJ-dependent manner that might contribute to the transdifferentiation process of MSCs within a cardiac environment. Our results suggest that the predominant mechanism of HSCs contribution to cardiac regeneration is based on their ability to regulate angiogenesis. In contrast, transplanted MSCs have the capability for intercellular communication with surrounding cardiomyocytes, which triggers the intrinsic program of cardiogenic lineage specification of MSCs by providing cardiomyocyte-derived cues.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Infarto do Miocárdio/terapia , Transdução de Sinais , Animais , Comunicação Celular , Transdiferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Junções Comunicantes/metabolismo , Humanos , Camundongos SCID , Miócitos Cardíacos/fisiologia , Neovascularização Fisiológica
3.
Biomaterials ; 135: 74-84, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28494265

RESUMO

BACKGROUND: Magnetic activated cell sorting (MACS®) is routinely used to isolate stem cell subpopulations intended for the treatment of cardiovascular diseases. In strong contrast, studies examining the amount, effect and intramyocardial distribution of iron nanoparticles used for magnetic cell labelling are missing, although iron excess can cause functional disorders in the heart. METHODS AND RESULTS: CD133+ haematopoietic and CD271+ mesenchymal stem cells were purified from bone marrow using automatically and manually MACS® based systems. Flow cytometric measurements demonstrated a rapid loss of MACS® MicroBeads from cells under culture conditions, while storage under hypothermic conditions decelerated their detachment. Moreover, an average loading of ∼11 fg iron/cell caused by magnetic labelling was determined in magnetic particle spectroscopy. Importantly, hemodynamic measurements as well as histological examinations using a myocardial ischemia/reperfusion mouse model showed no influence of MACS® MicroBeads on cardiac regeneration, while the transplantation of stem cells caused a significant improvement. Furthermore, immunostainings demonstrated the clearance of co-injected iron nanoparticles from stem cells and the surrounding heart tissue within 48 h post transplantation. CONCLUSIONS: Our results indicate that iron amounts typically co-injected with MACS® purified stem cells do not harm cardiac functions and are cleared from heart tissue within a few hours. Therefore, we conclude that MACS® MicroBeads exhibit a good compatibility in the cardiac environment.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Ferro/metabolismo , Miocárdio/metabolismo , Nanopartículas/metabolismo , Antígeno AC133/metabolismo , Adapaleno/metabolismo , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Humanos , Ferro/química , Leucócitos Mononucleares/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos SCID , Miocárdio/citologia , Nanopartículas/química
4.
Stem Cell Res Ther ; 8(1): 33, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28187777

RESUMO

BACKGROUND: CD133+ stem cells represent a promising subpopulation for innovative cell-based therapies in cardiovascular regeneration. Several clinical trials have shown remarkable beneficial effects following their intramyocardial transplantation. Yet, the purification of CD133+ stem cells is typically performed in centralized clean room facilities using semi-automatic manufacturing processes based on magnetic cell sorting (MACS®). However, this requires time-consuming and cost-intensive logistics. METHODS: CD133+ stem cells were purified from patient-derived sternal bone marrow using the recently developed automatic CliniMACS Prodigy® BM-133 System (Prodigy). The entire manufacturing process, as well as the subsequent quality control of the final cell product (CP), were realized on-site and in compliance with EU guidelines for Good Manufacturing Practice. The biological activity of automatically isolated CD133+ cells was evaluated and compared to manually isolated CD133+ cells via functional assays as well as immunofluorescence microscopy. In addition, the regenerative potential of purified stem cells was assessed 3 weeks after transplantation in immunodeficient mice which had been subjected to experimental myocardial infarction. RESULTS: We established for the first time an on-site manufacturing procedure for stem CPs intended for the treatment of ischemic heart diseases using an automatized system. On average, 0.88 × 106 viable CD133+ cells with a mean log10 depletion of 3.23 ± 0.19 of non-target cells were isolated. Furthermore, we demonstrated that these automatically isolated cells bear proliferation and differentiation capacities comparable to manually isolated cells in vitro. Moreover, the automatically generated CP shows equal cardiac regeneration potential in vivo. CONCLUSIONS: Our results indicate that the Prodigy is a powerful system for automatic manufacturing of a CD133+ CP within few hours. Compared to conventional manufacturing processes, future clinical application of this system offers multiple benefits including stable CP quality and on-site purification under reduced clean room requirements. This will allow saving of time, reduced logistics and diminished costs.


Assuntos
Automação Laboratorial/instrumentação , Separação Celular/instrumentação , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Infarto do Miocárdio/terapia , Regeneração/fisiologia , Antígeno AC133/genética , Antígeno AC133/metabolismo , Idoso , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Diferenciação Celular , Proliferação de Células , Separação Celular/métodos , Modelos Animais de Doenças , Feminino , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Masculino , Camundongos , Camundongos SCID , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Recuperação de Função Fisiológica/fisiologia , Doadores de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...