Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Acta Radiol ; 61(7): 894-902, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31752497

RESUMO

BACKGROUND: Pressure-volume analysis is the gold standard for quantifying pump function of the right ventricle (RV); however, volume measurements based on a conductive catheter may be imprecise. The reference method for volume assessment is cardiac magnetic resonance (CMR). PURPOSE: To determine the levels of agreement between RV volume measurements obtained by cine CMR, phase-contrast CMR (PC CMR), and a conductance catheter in an animal model. MATERIAL AND METHODS: CMR was performed in 20 sheep three months after pulmonary artery banding. Ejection fraction (EF), end-diastolic (EDV), end-systolic (ESV), and stroke volumes (SV) were obtained by cine CMR and conductance catheter. RESULTS: Statistically significant differences between cine CMR and conductance catheter derived volume measurements were found for EDV (P < 0.001), ESV (P < 0.05), and SV (P < 0.05). Bland-Altman analysis showed very poor agreement between the two methods: EDV, bias 36.27 mL, agreement of limits 1.96-70.57 mL; ESV, bias 15.33 mL, agreement of limits -6.89-37.55 mL; and SV, bias 20.69 mL, agreement of limits 8.01-49.10 mL. Good agreement was found for SV between cine CMR and PC CMR (bias -7.0 mL, agreement of limits -24.01-9.98 mL), while SV derived from PC CMR measurements showed poor agreement with conductance catheter (bias 27.76 mL, agreement of limits -3.84-59.26 mL). CONCLUSION: Poor agreement between the conductance catheter and CMR RV volume measurements was found. PC CMR and cine CMR measurements of SV agreed well.


Assuntos
Imagem Cinética por Ressonância Magnética/métodos , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/fisiopatologia , Disfunção Ventricular Direita/diagnóstico por imagem , Disfunção Ventricular Direita/fisiopatologia , Adaptação Fisiológica , Animais , Pressão Arterial , Cateterismo Cardíaco , Técnicas de Imagem de Sincronização Cardíaca , Modelos Animais de Doenças , Hemodinâmica/fisiologia , Interpretação de Imagem Assistida por Computador , Ligadura , Carneiro Doméstico , Volume Sistólico
2.
Cells Tissues Organs ; 206(1-2): 35-45, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30630170

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/patologia , Miocárdio/patologia , Vimentina/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Separação Celular , Sobrevivência Celular , Células Cultivadas , Feminino , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/metabolismo , Antígenos Comuns de Leucócito/análise , Antígenos Comuns de Leucócito/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Vimentina/análise
3.
Eur Surg Res ; 58(5-6): 341-353, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29073604

RESUMO

BACKGROUND/PURPOSE: Cardiac mesenchymal stem cells (MSCs) could stimulate cell-specific regenerative mechanisms after myocardial infarction (MI) depending on spatial origin, distribution, and niche regulation. We aimed at identifying and isolating tissue-specific cardiac MSCs that could contribute to regeneration. METHODS: Following permanent ligation of the left anterior descending coronary artery in rats (n = 16), early cardiac tissues and cardiac mononuclear cells (MNCs) were analyzed by immunohistology, confocal laser scanning microscopy, and flow cytometry, respectively. Early postischemic specific MSCs were purified by fluorescence-activated cell sorting, cultivated under standardized culture conditions, and tested for multipotent differentiation in functional identification kits. RESULTS: Cardiac MSC niches were detected intramyocardially in cell clusters after MI and characterized by positive expression for vimentin, CD29, CD44, CD90, CD105, PDGFRα, and DDR2. Following myocardial ischemia, proliferation was induced early and proliferation density was approximately 11% in intramyocardial MSC clusters of the peri-infarction border zone. Cluster sizes increased by 157 and 64% in the peri-infarction and noninfarcted areas of infarcted hearts compared with noninfarcted hearts 24 h following MI, respectively. Coincidentally, flow cytometry analyses illustrated postischemic moderate enrichments of CD45-CD44+ and CD45-DDR2+ cardiac MNCs. We enabled isolation of early postischemic culturable cardiac CD45-CD44+DDR2+ MSCs that demonstrated typical clonogenicity with colony-forming unit-fibroblast formation as well as adipogenic, chondrogenic, and osteogenic differentiation. CONCLUSIONS: MI triggered early proliferation in specific cardiac MSC niches that were organized in intramyocardial clusters. Following targeted isolation, early postischemic cardiac CD45-CD44+DDR2+ MSCs exhibited typical characteristics with multipotent differentiation capacity and clonogenic expansion.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Isquemia Miocárdica , Miocárdio/citologia , Regeneração , Animais , Proliferação de Células , Masculino , Ratos Endogâmicos Lew , Nicho de Células-Tronco
4.
J Cell Mol Med ; 19(8): 1975-85, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25991381

RESUMO

Myocardial infarction (MI) is a major condition causing heart failure (HF). After MI, the renin angiotensin system (RAS) and its signalling octapeptide angiotensin II (Ang II) interferes with cardiac injury/repair via the AT1 and AT2 receptors (AT1R, AT2R). Our study aimed at deciphering the mechanisms underlying the link between RAS and cellular components of the immune response relying on a rodent model of HF as well as HF patients. Flow cytometric analyses showed an increase in the expression of CD4(+) AT2R(+) cells in the rat heart and spleen post-infarction, but a reduction in the peripheral blood. The latter was also observed in HF patients. The frequency of rat CD4(+) AT2R(+) T cells in circulating blood, post-infarcted heart and spleen represented 3.8 ± 0.4%, 23.2 ± 2.7% and 22.6 ± 2.6% of the CD4(+) cells. CD4(+) AT2R(+) T cells within blood CD4(+) T cells were reduced from 2.6 ± 0.2% in healthy controls to 1.7 ± 0.4% in patients. Moreover, we characterized CD4(+) AT2R(+) T cells which expressed regulatory FoxP3, secreted interleukin-10 and other inflammatory-related cytokines. Furthermore, intramyocardial injection of MI-induced splenic CD4(+) AT2R(+) T cells into recipient rats with MI led to reduced infarct size and improved cardiac performance. We defined CD4(+) AT2R(+) cells as a T cell subset improving heart function post-MI corresponding with reduced infarction size in a rat MI-model. Our results indicate CD4(+) AT2R(+) cells as a promising population for regenerative therapy, via myocardial transplantation, pharmacological AT2R activation or a combination thereof.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Testes de Função Cardíaca , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/fisiopatologia , Receptor Tipo 2 de Angiotensina/metabolismo , Remodelação Ventricular , Animais , Cardiotônicos/metabolismo , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Imunomodulação , Interleucina-10/sangue , Infarto do Miocárdio/sangue , Infarto do Miocárdio/complicações , Isquemia Miocárdica/sangue , Isquemia Miocárdica/complicações , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/fisiopatologia , Ratos Wistar , Fator de Necrose Tumoral alfa/sangue
5.
J Cell Mol Med ; 15(6): 1310-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20477905

RESUMO

Matrigel promotes angiogenesis in the myocardium from ischemic injury and prevents remodelling of the left ventricle. We assessed the therapeutic efficacy of intracardiac matrigel injection and matrigel-mediated stem cell homing in a rat myocardial infarction (MI) model. Following MI, matrigel (250 µl) or phosphate-buffered solution (PBS) was delivered by intracardiac injection. Compared to the MI control group (MI-PBS), matrigel significantly improved left ventricular function (n= 11, P < 0.05) assessed by pressure-volume loops after 4 weeks. There is no significant difference in infarct size between MI-matrigel (MI-M; 21.48 ± 1.49%, n = 10) and MI-PBS hearts (20.98 ± 1.25%, n = 10). The infarct wall thickness of left ventricle is significantly higher (P < 0.01) in MI-M (0.72 ± 0.02 mm, n = 10) compared with MI-PBS (0.62 ± 0.02 mm, n = 10). MI-M hearts exhibited higher capillary density (border 130.8 ± 4.7 versus 115.4 ± 6.0, P < 0.05; vessels per high-power field [HPF; 400×], n = 6) than MI-PBS hearts. c-Kit(+) stem cells (38.3 ± 5.3 versus 25.7 ± 1.5 c-Kit(+) cells per HPF [630×], n = 5, P < 0.05) and CD34(+) cells (13.0 ± 1.51 versus 5.6 ± 0.68 CD34(+) cells per HPF [630×], n = 5, P < 0.01) were significantly more numerous in MI-M than in MI-PBS in the infarcted hearts (n = 5, P < 0.05). Intracardiac matrigel injection restores myocardial functions following MI, which may attribute to the improved recruitment of CD34(+) and c-Kit(+) stem cells.


Assuntos
Movimento Celular/efeitos dos fármacos , Colágeno , Laminina , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/patologia , Proteoglicanas , Animais , Aorta Torácica/fisiopatologia , Colágeno/administração & dosagem , Colágeno/uso terapêutico , Modelos Animais de Doenças , Combinação de Medicamentos , Hemodinâmica/efeitos dos fármacos , Injeções Intramusculares , Laminina/administração & dosagem , Laminina/uso terapêutico , Ligadura , Masculino , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Proteoglicanas/administração & dosagem , Proteoglicanas/uso terapêutico , Ratos , Ratos Endogâmicos , Células-Tronco/fisiologia , Função Ventricular Esquerda/efeitos dos fármacos
6.
Herz ; 35(5): 324-33, 2010 Aug.
Artigo em Alemão | MEDLINE | ID: mdl-20625690

RESUMO

Chronic ischemic heart disease patients are already being treated worldwide with bone marrow stem cells both in the context of clinical studies and in therapy trials. By combining this therapy with established revascularization procedures such as bypass surgery, a high level of patient safety can be achieved. To date, no stem cell-related cardiac complications following intramyocardial injection of bone marrow-derived stem cells during CABG (coronary artery bypass graft) surgery have been reported. The functional advantage conferred by surgical bone marrow stem cell therapy is a 7.2% increase in LVEF (left ventricular ejection fraction) compared to controls. Randomized placebo-controlled trials, like the German trial PERFECT, are needed to obtain a more evidence-based assessment of this therapy.


Assuntos
Transplante de Medula Óssea/métodos , Ponte de Artéria Coronária , Insuficiência Cardíaca/cirurgia , Infarto do Miocárdio/cirurgia , Isquemia Miocárdica/cirurgia , Doença Crônica , Terapia Combinada , Ensaios Clínicos Controlados como Assunto , Insuficiência Cardíaca/fisiopatologia , Humanos , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Volume Sistólico/fisiologia , Resultado do Tratamento , Disfunção Ventricular Esquerda/fisiopatologia , Disfunção Ventricular Esquerda/cirurgia , Função Ventricular Esquerda/fisiologia
7.
J Crit Care ; 56: 106-112, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31896443

RESUMO

Mechanical circulatory support (MCS) using left ventricular assist devices (LVAD) have considerably improved the quality of life and survival rate of patients with end-stage heart failure. Despite substantial technological progress, major challenges with regard to VAD-specific and VAD-related infections have hitherto hindered the broader application of this promising therapy approach. Driveline infections (DLI) range among the main adverse events experienced in LVAD patients. However, many centers still apply their own protocol for driveline exit site (DLES) care and an international standard on prevention, reduction and early treatment of DLI after the perioperative period has not yet been defined. In March 2019, VAD coordinators and cardiac surgeons from Germany and Austria met to develop a standard of care procedure (SOP) as well as a new staging approach with recommended actions for treatment of VAD carriers. In this Driveline Expert STagINg and carE (DESTINE) study group we developed a 10-step SOP for DLES care with emphasis on essentials such as clean and save preparation, sterile dressing change and secure driveline immobilization. An advanced wound staging approach was defined with recommended actions for prevention, early detection and stage-related management of DLI. Broad consensus was reached on the fact that an interdisciplinary approach both in DLES care and DLES healing disorder awareness is required to prolong infect-free survival times on MCS as well as to ensure high patient compliance and quality of life. In conclusion, a new detailed SOP for appropriate DLES care and an advanced wound staging approach for prevention and management of DLI were defined on an expert level applicable for VAD clinicians, practitioners and care givers in Central Europe.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Coração Auxiliar/efeitos adversos , Infecções Relacionadas à Prótese/prevenção & controle , Dermatopatias/prevenção & controle , Áustria , Cardiologia/normas , Feminino , Alemanha , Humanos , Cooperação Internacional , Masculino , Pessoa de Meia-Idade , Pacientes Ambulatoriais , Infecções Relacionadas à Prótese/diagnóstico , Qualidade de Vida , Dermatopatias/diagnóstico , Padrão de Cuidado , Taxa de Sobrevida
8.
EBioMedicine ; 57: 102862, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32629392

RESUMO

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.


Assuntos
Antígeno AC133/genética , Transplante de Medula Óssea/métodos , Doença da Artéria Coronariana/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Isquemia Miocárdica/terapia , Adolescente , Adulto , Idoso , Células da Medula Óssea/citologia , Senescência Celular/genética , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/fisiopatologia , Feminino , Coração/crescimento & desenvolvimento , Coração/fisiopatologia , Células-Tronco Hematopoéticas/citologia , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/genética , Isquemia Miocárdica/patologia , Regeneração/genética , Adulto Jovem
9.
J Cell Mol Med ; 13(4): 664-79, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19449462

RESUMO

Erythropoietin (EPO) protects the myocardium from ischaemic injury and promotes beneficial remodelling. We assessed the therapeutic efficacy of intracardiac EPO injection and EPO-mediated stem cell homing in a rat myocardial infarction (MI) model. Following MI, EPO (3000 U/kg) or saline was delivered by intracardiac injection. Compared to myocardial infarction control group (MIC), EPO significantly improved left ventricular function (n =11-14, P < 0.05) and decreased right ventricular wall stress (n = 8, P < 0.05) assessed by pressure-volume loops after 6 weeks. MI-EPO hearts exhibited smaller infarction size (20.1 +/- 1.1% versus 27.8 +/- 1.2%; n = 6-8, P < 0.001) and greater capillary density (338.5 +/- 14.7 versus 259.8 +/- 9.2 vessels per mm2; n = 6-8, P < 0.001) than MIC hearts. Direct EPO injection reduced post-MI myocardial apoptosis by approximately 41% (0.27 +/- 0.03% versus 0.42 +/- 0.03%; n = 6, P= 0.005). The chemoattractant SDF-1 was up-regulated significantly assessed by quantitative realtime PCR and immunohistology. c-Kit(+) and CD34(+) stem cells were significantly more numerous in MI-EPO than in MIC at 24 hrs in peripheral blood (n = 7, P < 0.05) and 48 hrs in the infarcted hearts (n = 6, P < 0.001). Further, the mRNAs of Akt, eNOS and EPO receptor were significantly enhanced in MI-EPO hearts (n = 7, P < 0.05). Intracardiac EPO injection restores myocardial functions following MI, which may attribute to the improved early recruitment of c-Kit(+) and CD34(+) stem cells via the enhanced expression of chemoattractant SDF-1.


Assuntos
Eritropoetina/administração & dosagem , Eritropoetina/uso terapêutico , Testes de Função Cardíaca , Mobilização de Células-Tronco Hematopoéticas , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Eritropoetina/farmacologia , Hematócrito , Humanos , Injeções , Metaloproteinase 2 da Matriz/metabolismo , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Receptores CXCR4/metabolismo , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Troponina T/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Cell Transplant ; 18(3): 319-31, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19558780

RESUMO

Bone marrow-derived mesenchymal stem cells (MSCs) are multipotent cells characterized by their self-renewal and differentiation potential. Accumulating clinical and preclinical evidence indicate MSCs are a promising cell source for regenerative medical therapies. However, undesirable immortalization, spontaneous transformation, and tumorigenic potential from long-term cultured MSCs have been reported in human and mouse. We report rat MSCs isolated from young donors could undergo transformation in early passage culture. We aimed to characterize the transformed population and determine their therapeutic effects after intracardiac transplantation in the infarcted myocardium. MSCs were isolated from bone marrow of Lewis rats according to standard protocols and cultured under standard conditions. Phenotype of growing cells was assessed by flow cytometry. Following acute myocardial infarction in rats, cells were delivered by intracardiac injection. Cardiac functions were assessed by pressure-volume loops. Infarction size and pathologic effects were evaluated after 6 weeks. The abnormal colonies were detected in culture as early at passage 3. They were noted to appear as distinctly different morphology from typical MSCs, which changed from a normal elongated spindle shape to a compact abnormal morphology. They exhibited rapid cell proliferation. Some subclones lost contact inhibition of cell division and formed multilayer aggregates. Chromosomal instability was detected. They were devoid of surface markers CD29, CD44, CD90, and CD117. Furthermore, there was no significant improvement on infarction size and cardiac function 6 weeks after cell transplantation. Our study highlights the need for establishment of biosafety criteria in regulating culture- expanded MSCs to achieve the full clinical therapeutic benefits.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/patologia , Miocárdio/patologia , Animais , Biomarcadores/metabolismo , Linhagem Celular Transformada , Células Cultivadas , Aberrações Cromossômicas , Cromossomos de Mamíferos/metabolismo , Testes de Função Cardíaca , Imuno-Histoquímica , Imunofenotipagem , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Ratos
11.
J Vis Exp ; (153)2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31814607

RESUMO

Antegrade endoscopic harvesting of autografts for bypass grafting may be an optimal strategy addressing excellent graft quality and reduced post-operative wound complications. This standardized protocol for antegrade endoscopic vein harvesting (EVH) from the lower leg has the potential to be introduced to routine coronary artery bypass grafting (CABG). Patients undergoing CABG surgery are positioned on a surgical table with two additional foam rollers below the extended legs, enabling antegrade EVH from the lower leg. Following minimally invasive surgical access through a bridging vein harvest technique, an endoscopic optical dissector is inserted antegrade into the wound. The main vessel and side branches are dissected under continuous optical control of vein quality status and the working channel. After, an endoscopic optical retractor is inserted with an internal bipolar electrocoagulation device for precise, safe, and tissue-protective interruption of side branches. After release of the vein, the vessel is cut off at the proximal and distal ends under optical control, retrieved from the wound, then cannulated and flushed with heparinized saline. Finally, all side branches of the vein graft are double-clipped. Vascular histology is analyzed in a randomized selection of vein samples. After applying this standardized EVH protocol, the learning curve was shown to be steep, and graft quality was sufficient for coronary artery bypass grafting in every case. There was no conversion to surgical harvesting and low risks for tissue damage and bleeding. Leg positioning and synergizing EVH with bridging vein harvesting improved procedural success and vein graft quality. In our hands, antegrade EVH from the lower leg was feasible, demonstrating straightforward graft dissection as well as adequate macroscopic and microscopic graft quality with preserved endothelial integrity. In conclusion, the introduced technique is safe, shows excellent vein autograft quality, and illustrates feasibility for elective and urgent isolated CABG and combined CABG scenarios.


Assuntos
Ponte de Artéria Coronária/métodos , Perna (Membro)/irrigação sanguínea , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Veia Safena/transplante , Coleta de Tecidos e Órgãos/métodos , Procedimentos Cirúrgicos Vasculares/métodos , Idoso , Ponte de Artéria Coronária/normas , Feminino , Humanos , Perna (Membro)/cirurgia , Masculino , Pessoa de Meia-Idade , Procedimentos Cirúrgicos Minimamente Invasivos/normas , Complicações Pós-Operatórias/prevenção & controle , Distribuição Aleatória , Veia Safena/cirurgia , Método Simples-Cego , Coleta de Tecidos e Órgãos/normas , Procedimentos Cirúrgicos Vasculares/normas
12.
J Cardiovasc Transl Res ; 12(5): 459-466, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30847657

RESUMO

This experimental study describes the adaptive processes of the right ventricular (RV) myocardium after pulmonary artery banding (PAB) evaluated by cine cardiac magnetic resonance (CMR), phase-contrast CMR (PC-CMR), and conductance catheter. Seven sheep were subjected to CMR 3 months after PAB. Conductance catheter measurements were performed before and 3 months after PAB. Four nonoperated, healthy, age-matched animals served as controls. Higher RV masses (p < 0.01), elevated RV end-systolic volumes (p < 0.05), and lower RV ejection fraction (p < 0.01) were observed in the operated group. The time-to-peak pulmonary artery flow was longer in the banding group (p < 0.01). RV maximal pressure and RV end-diastolic pressure correlated with the time-to-peak flow in the pulmonary artery (r = - 0.70 and - 0.69, respectively). In summary, PAB caused RV hypertrophy, increased myocardial contractility, and decreased RV-EF and cardiac output. The time-to-peak pulmonary artery flow correlated with RV pressures.


Assuntos
Cateterismo Cardíaco , Hemodinâmica , Hipertrofia Ventricular Direita/diagnóstico , Imagem Cinética por Ressonância Magnética , Hipertensão Arterial Pulmonar/diagnóstico , Artéria Pulmonar/fisiopatologia , Disfunção Ventricular Direita/diagnóstico , Função Ventricular Direita , Remodelação Ventricular , Adaptação Fisiológica , Animais , Pressão Arterial , Modelos Animais de Doenças , Feminino , Hipertrofia Ventricular Direita/etiologia , Hipertrofia Ventricular Direita/fisiopatologia , Ligadura , Valor Preditivo dos Testes , Hipertensão Arterial Pulmonar/etiologia , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/cirurgia , Carneiro Doméstico , Volume Sistólico , Disfunção Ventricular Direita/etiologia , Disfunção Ventricular Direita/fisiopatologia , Pressão Ventricular
13.
Dis Model Mech ; 11(6)2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29752300

RESUMO

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.


Assuntos
Eritropoetina/uso terapêutico , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Isquemia Miocárdica/patologia , Isquemia Miocárdica/terapia , Miocárdio/patologia , Neovascularização Fisiológica , Pericárdio/metabolismo , Doença Aguda , Animais , Antígenos CD/metabolismo , Capilares/patologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Eritropoetina/administração & dosagem , Eritropoetina/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Testes de Função Cardíaca , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Mesoderma/patologia , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Ratos Endogâmicos Lew , Regeneração/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
14.
ESC Heart Fail ; 4(2): 105-111, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28451446

RESUMO

AIMS: Standardization of stem cell therapy requires application of appropriate methods to evaluate safety and efficacy, including long-term pharmacovigilance. To accomplish this objective, a long-term registry programme was installed. METHODS AND RESULTS: We analysed 150 patients with ischemic cardiomyopathy, who received intramyocardial CD133+ bone marrow mononuclear stem cell treatment combined with coronary artery bypass grafting (CABG) or CABG alone. The mortality rate, major adverse cerebral and cardiac events, and functional outcome parameters were evaluated for the time period up to 14 years follow-up. As a result, we have stratified the patient population (96 patients) into responders and non-responders. Furthermore, the analysis of relevant predictors of good response to CD133+ bone marrow mononuclear stem cell treatment was performed. Several positive tendencies related to stem cells transplantation were demonstrated. First, no significant difference in major adverse cardiovascular and cerebral events was observed between stem cell and control group up to 14 years follow-up. Second, an improvement of left ventricle ejection fraction (LVEF) in stem cell group retained for 5 years in contrast with CABG-only group, where no significant changes in LVEF after 2 years were observed. In addition, LVEF under 30% and left ventricle end diastolic diameter above 60 mm were independent predictors of functional response to CD133+ cell therapy. CONCLUSIONS: Participants with overt heart failure benefit most from CABG combined with intramyocardial injection of CD133+ bone marrow mononuclear cell within the group. An improvement LVEF in stem cell group remained for 5 years in contrast with the CABG-only group. The patients, in whom the improvement of both LVEF and LVED was observed, have benefited by increased life expectancy.

15.
Biores Open Access ; 4(1): 278-87, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26309803

RESUMO

Applying cells in a spray can overcome current hurdles in coating tissue engineered constructs with a thin layer of endo- or epithelial cells. We report here a structured study on the influences of spray application with a medical spray device on vascular smooth muscle cells (vSMCs) and respiratory epithelial cells (RECs) with and without fibrin gel. Next to viability and cytotoxicity assays, the in vitro differentiation capacity after spray processing was analyzed. For vSMC, no influence of air pressures till 0.8 bar could be shown, whereas the viability decreased for higher pressures. The viability of RECs was reduced to 88.5% with 0.4 bar air pressure. Lactate dehydrogenase-levels in the culture medium increased the first day after spraying but normalized afterward. In the short term, no differences by means of morphology and expression-specific markers for vSMCs and RECs were seen between the control and study group. In addition, in a long-term study for 28 days with the air-liquid interface, RECs differentiated and built up an organized epithelial layer with ciliary development that was comparable to the control for cells sprayed without fibrin gel. When spraying within fibrin gel, ciliary development was lower at 28 days. Thus, spraying of vSMCs and RECs was proved to be a suitable method for tissue engineering. Especially for RECs, this application is of special significance when coating luminal structures or other unfavorable topographies.

16.
Cell Transplant ; 24(11): 2409-22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24800805

RESUMO

Stem cell transplantation is a viable strategy for regenerative medicine. However, it is inevitable to have cells undergo storage for several hours or days due to processing and transportation. Therefore, it is crucial to have rigidly controlled conditions ensuring the therapeutic benefit of isolated stem cells. In the present study, we investigated the impact of short-term storage on human CD133(+) cells. CD133(+) cells were isolated from human bone marrow and kept at standardized nonfreezing storage conditions for up to 72 h. Cell viability (apoptosis/necrosis) and expression of CD133 and CXCR4 were analyzed by flow cytometry. Metabolic activity was determined using an MTT assay; colony-forming ability, as well as endothelial-like differentiation, was further evaluated. A qRT-PCR array was employed to investigate the expression of stemness genes. CD133 and CXCR4 expressions were preserved at all time points. After 30 h, cell number and metabolic activity decreased, although no significant changes were detected in cell viability and proliferation as well as endothelial-like differentiation. Cell viability and proliferation decreased significantly only after 72 h of storage. Our results indicate that storage of isolated human CD133(+) bone marrow stem cells in liquid allows for high viability and functionality. However, storage time should be limited in order to avoid cell loss.


Assuntos
Sobrevivência Celular/fisiologia , Células-Tronco/citologia , Preservação de Tecido , Antígeno AC133 , Antígenos CD/genética , Antígenos CD/metabolismo , Células da Medula Óssea/citologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Peptídeos/genética , Peptídeos/metabolismo , Receptores CXCR4 , Transplante de Células-Tronco
17.
J Tissue Eng Regen Med ; 9(12): E177-90, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23208947

RESUMO

At present, intensive investigation aims at the creation of optimal valvular prostheses. We introduced and tested the applicability and functionality of two advanced cell-plus-matrix seeding technologies, spray-assisted bioprocessing (SaBP) and laser-assisted bioprocessing (LaBP), for autologous tissue engineering (TE) of bioresorbable artificial grafts. For SaBP, human mesenchymal stem cells (HMSCs), umbilical cord vein endothelial cells (HUVECs) and fibrin were simultaneously spray-administered on poly(ε-caprolactone) (PCL) substrates. For LaBP, HUVECs and HMSCs were separately laser-printed in stripes, followed by fibrin sealing. Three-leaflet valves were manufactured following TE of electrospun PCL tissue equivalents. Grafts were monitored in vitro under static and dynamic conditions in bioreactors. SaBP and LaBP resulted in TE of grafts with homogeneous cell distribution and accurate cell pattern, respectively. The engineered valves demonstrated immediate sufficient performance, complete cell coating, proliferation, engraftment, HUVEC-mediated invasion, HMSC differentiation and extracellular matrix deposition. SaBP revealed higher efficiency, with at least 12-fold shorter processing time than the applied LaBP set-up. LaBP realized coating with higher cell density and minimal cell-scaffold distance. Fibrin and PCL stability remain issues for improvement. The introduced TE technologies resulted in complete valvular cell-plus-matrix coating, excellent engraftment and HMSCs differentiation. SaBP might have potential for intraoperative table-side TE considering the procedural duration and ease of implementation. LaBP might accelerate engraftment with precise patterns.


Assuntos
Matriz Extracelular/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Poliésteres/química , Engenharia Tecidual/métodos , Feminino , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia
18.
J R Soc Interface ; 10(82): 20120820, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-23427092

RESUMO

When hungry, the wandering spider Cupiennius salei is frequently seen to catch flying insect prey. The success of its remarkable prey-capture jump from its sitting plant into the air obviously depends on proper timing and sensory guidance. In this study, it is shown that particular features of the airflow generated by the insect suffice to guide the spider. Vision and the reception of substrate vibrations and airborne sound are not needed. The behavioural reactions of blinded spiders were examined by exposing them to natural and synthetic flows imitating the fly-generated flow or particular features of it. Thus, the different roles of the three phases previously identified in the fly-generated flow and described in the companion paper could be demonstrated. When exposing the spider to phase I flow only (exponentially increasing flow velocity with very little fluctuation and typical of the fly's approach), an orienting behaviour could be observed but a prey-capture jump never be elicited. Remarkably, the spider reacted to the onset of phase II (highly fluctuating flow) of a synthetically generated flow field with a jump as frequently as it did when exposed to natural fly-generated flows. In all cases using either natural or artificial flows, the spider's jump was triggered before its flow sensors were hit by phase III flow (steadily decreasing airflow velocity). Phase III may tell the spider that the prey has passed by already in case of no prey-capture reaction. Our study underlines the relevance of airflow in spider behaviour. It also reflects the sophisticated workings of their flow sensors (trichobothria) previously studied in detail. Presumably, the information contained in prey-generated airflows plays a similar role in many other arthropods.


Assuntos
Comportamento Predatório/fisiologia , Aranhas/fisiologia , Animais
19.
J R Soc Interface ; 9(75): 2591-602, 2012 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-22572032

RESUMO

The hunting spider Cupiennius salei uses airflow generated by flying insects for the guidance of its prey-capture jump. We investigated the velocity field of the airflow generated by a freely flying blowfly close to the flow sensors on the spider's legs. It shows three characteristic phases (I-III). (I) When approaching, the blowfly induces an airflow signal near the spider with only little fluctuation (0.013 ± 0.006 m s(-1)) and a strength that increases nearly exponentially with time (maximum: 0.164 ± 0.051 m s(-1) s.d.). The spider detects this flow while the fly is still 38.4 ± 5.6 mm away. The fluctuation of the airflow above the sensors increases linearly up to 0.037 m s(-1) with the fly's altitude. Differences in the time of arrival and intensity of the fly signal at different legs probably inform the spider about the direction to the prey. (II) Phase II abruptly follows phase I with a much higher degree of fluctuation (fluctuation amplitudes: 0.114 ± 0.050 m s(-1)). It starts when the fly is directly above the sensor and corresponds to the time-dependent flow in the wake below and behind the fly. Its onset indicates to the spider that its prey is now within reach and triggers its jump. The spider derives information on the fly's position from the airflow characteristics, enabling it to properly time its jump. The horizontal velocity of the approaching fly is reflected by the time of arrival differences (ranging from 0.038 to 0.108 s) of the flow at different legs and the exponential velocity growth rate (16-79 s(-1)) during phase I. (III) The air flow velocity decays again after the fly has passed the spider.


Assuntos
Movimentos do Ar , Dípteros/fisiologia , Voo Animal/fisiologia , Comportamento Predatório/fisiologia , Aranhas/fisiologia , Animais
20.
PLoS One ; 6(2): e15652, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21347366

RESUMO

The possible different therapeutic efficacy of human mesenchymal stem cells (hMSC) derived from umbilical cord blood (CB), adipose tissue (AT) or bone marrow (BM) for the treatment of myocardial infarction (MI) remains unexplored. This study was to assess the regenerative potential of hMSC from different origins and to evaluate the role of CD105 in cardiac regeneration. Male SCID mice underwent LAD-ligation and received the respective cell type (400.000/per animal) intramyocardially. Six weeks post infarction, cardiac catheterization showed significant preservation of left ventricular functions in BM and CD105(+)-CB treated groups compared to CB and nontreated MI group (MI-C). Cell survival analyzed by quantitative real time PCR for human GAPDH and capillary density measured by immunostaining showed consistent results. Furthermore, cardiac remodeling can be significantly attenuated by BM-hMSC compared to MI-C. Under hypoxic conditions in vitro, remarkably increased extracellular acidification and apoptosis has been detected from CB-hMSC compared to BM and CD105 purified CB-derived hMSC. Our findings suggests that hMSC originating from different sources showed a different healing performance in cardiac regeneration and CD105(+) hMSC exhibited a favorable survival pattern in infarcted hearts, which translates into a more robust preservation of cardiac function.


Assuntos
Coração/fisiopatologia , Células-Tronco Mesenquimais/citologia , Regeneração , Cicatrização/fisiologia , Animais , Antígenos CD/metabolismo , Capilares/metabolismo , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Humanos , Ligadura/efeitos adversos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica , Receptores de Superfície Celular/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA