Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eur J Haematol ; 111(5): 748-756, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37565648

RESUMO

OBJECTIVE: Ibrutinib treatment is associated with cardiovascular side effects, in particular atrial fibrillation (AF) and hypertension, as well as increased risk of bleeding. Here, we aimed at describing the incidence of these events during long-term follow-up in patients with chronic lymphocytic leukemia treated outside clinical trials as well as identifying clinical factors predictive of developing AF. Additionally, other reasons for treatment withdrawal were analyzed. METHODS: The study was retrospective, data were collected from medical records. RESULTS: A total of 134 patients were identified. Median follow-up was 32 months (range 3-103) and median duration of ibrutinib treatment was 26 months (range 1-103). Of 110 patients with no prior history of AF, 24.5% were diagnosed during treatment. Newly diagnosed or worsening of pre-existing hypertension occurred in 15.7%. Sixty-six % of the patients experienced bleeding events, of which 7.5% grade 3-4. Treatment discontinuation and dose reduction occurred in 68% and 47% of the patients, respectively, mostly due to toxicity. CONCLUSIONS: The incidence of AF was high and at a median follow-up of 2.5 years, two-thirds of the patients discontinued treatment mostly due to bleeding and infections. Treatment-related toxicity of any grade should be regarded as a concern of prolonged ibrutinib therapy.


Assuntos
Fibrilação Atrial , Hipertensão , Leucemia Linfocítica Crônica de Células B , Humanos , Leucemia Linfocítica Crônica de Células B/complicações , Leucemia Linfocítica Crônica de Células B/diagnóstico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Estudos Retrospectivos , Incidência , Hemorragia/induzido quimicamente , Hemorragia/diagnóstico , Hemorragia/epidemiologia , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/epidemiologia , Hipertensão/induzido quimicamente , Inibidores de Proteínas Quinases/uso terapêutico
2.
iScience ; 26(1): 105857, 2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36624836

RESUMO

Cardiomyocytes play key roles during cardiogenesis, but have poorly understood features, especially in prenatal stages. Here, we characterized human prenatal cardiomyocytes, 6.5-7 weeks post-conception, by integrating single-cell RNA sequencing, spatial transcriptomics, and ligand-receptor interaction information. Using a computational workflow developed to dissect cell type heterogeneity, localize cell types, and explore their molecular interactions, we identified eight types of developing cardiomyocyte, more than double compared to the ones identified in the Human Developmental Cell Atlas. These have high variability in cell cycle activity, mitochondrial content, and connexin gene expression, and are differentially distributed in the ventricles, including outflow tract, and atria, including sinoatrial node. Moreover, cardiomyocyte ligand-receptor crosstalk is mainly with non-cardiomyocyte cell types, encompassing cardiogenesis-related pathways. Thus, early prenatal human cardiomyocytes are highly heterogeneous and develop unique location-dependent properties, with complex ligand-receptor crosstalk. Further elucidation of their developmental dynamics may give rise to new therapies.

3.
Eur Heart J ; 43(27): 2562-2569, 2022 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34951629

RESUMO

AIMS: Trastuzumab and anthracyclines, often used in the treatment of breast cancer, may impair myocardial function, and reduce left ventricular ejection fraction (LVEF), potentially causing heart failure. Randomized controlled trials (RCTs) have evaluated the effects of beta-blockers (BBs), angiotensin receptor blockers (ARBs), and angiotensin-converting enzyme inhibitors (ACEI) on trastuzumab- and anthracycline-associated cardiotoxicity. We report a meta-analysis of these RCTs in patients with breast cancer. METHODS AND RESULTS: The primary analysis was on the effect of BBs and ACEI/ARBs on LVEF in patients treated with either trastuzumab or anthracyclines. A secondary analysis was done investigating the effect of BBs or ACEI/ARBs on LVEF in trastuzumab and anthracycline treatments. Only RCTs were included using the search term 'ARBs, ACEIs, BBs, anthracyclines, trastuzumab, and breast cancer' in PubMed, Embase, and CENTRAL up to 31 March 2021. A meta-analysis was conducted to estimate the mean difference (MD) in LVEF between intervention and placebo groups at follow-up. A total of nine RCTs (n = 1362) were included in the analysis. All patients were women. BBs and ACEI/ARBs were shown to attenuate the decline in LVEF during trastuzumab and anthracycline treatments [MD: 2.4; 95% confidence interval (CI): 0.3-4.2 and MD: 1.5; 95% CI: -0.6 to 3.7]. Compared with placebo, LVEF was significantly higher in patients assigned to BB or ACEI/ARB on trastuzumab (MD: 2.3; 95% CI: 0.0-4.6) but not on anthracyclines (MD: 1.9; 95% CI: -0.5 to 4.2). CONCLUSION: Both BB and ACEI/ARB therapies were associated with the preservation of LVEF during trastuzumab and anthracycline-containing regimens as compared with placebo, suggesting both to be beneficial.


Assuntos
Neoplasias da Mama , Disfunção Ventricular Esquerda , Antagonistas Adrenérgicos beta/efeitos adversos , Antagonistas de Receptores de Angiotensina/efeitos adversos , Inibidores da Enzima Conversora de Angiotensina/efeitos adversos , Antraciclinas/efeitos adversos , Antibióticos Antineoplásicos/farmacologia , Anti-Hipertensivos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Masculino , Sistema Renina-Angiotensina , Volume Sistólico , Trastuzumab/efeitos adversos , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/tratamento farmacológico , Disfunção Ventricular Esquerda/prevenção & controle
4.
ESC Heart Fail ; 7(2): 445-455, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31981321

RESUMO

AIMS: Neuregulin1-ß (NRG1-ß) is released from microvascular endothelial cells in response to inflammation with compensatory cardioprotective effects. Circulating NRG1-ß is elevated in heart failure (HF) with reduced ejection fraction (HFrEF) but not studied in HF with preserved EF (HFpEF). METHODS AND RESULTS: Circulating NRG1-ß was quantified in 86 stable patients with HFpEF (EF ≥45% and N-terminal pro-brain natriuretic peptide >300 ng/L), in 86 patients with HFrEF prior to and after left ventricular assist device (LVAD) and/or heart transplantation (HTx) and in 21 healthy controls. Association between NRG1-ß and the composite outcome of all-cause mortality/HF hospitalization in HFpEF and all-cause mortality/HTx/LVAD implantation in HFrEF with and without ischaemia assessed as macrovascular coronary artery disease was assessed. In HFpEF, median (25th-75th percentile) NRG1-ß was 6.5 (2.1-11.3) ng/mL; in HFrEF, 3.6 (2.1-7.6) ng/mL (P = 0.035); after LVAD, 1.7 (0.9-3.6) ng/mL; after HTx 2.1 (1.4-3.6) ng/mL (overall P < 0.001); and in controls, 29.0 (23.1-34.3) ng/mL (P = 0.001). In HFrEF, higher NRG1-ß was associated with worse outcomes (hazard ratio per log increase 1.45, 95% confidence interval 1.04-2.03, P = 0.029), regardless of ischaemia. In HFpEF, the association of NRG1-ß with outcomes was modified by ischaemia (log-rank P = 0.020; Pinteraction = 0.553) such that only in ischaemic patients, higher NRG1-ß was related to worse outcomes. In contrast, in patients without ischaemia, higher NRG1-ß trended towards better outcomes (hazard ratio 0.71, 95% confidence interval 0.48-1.05, P = 0.085). CONCLUSIONS: Neuregulin1-ß was reduced in HFpEF and further reduced in HFrEF. The opposing relationships of NRG1-ß with outcomes in non-ischaemic HFpEF compared with HFrEF and ischaemic HFpEF may indicate compensatory increases of cardioprotective NRG1-ß from microvascular endothelial dysfunction in the former (non-ischaemic HFpEF), but this compensatory mechanism is overwhelmed by the presence of ischaemia in the latter (HFrEF and ischaemic HFpEF).


Assuntos
Insuficiência Cardíaca , Células Endoteliais , Humanos , Prognóstico , Volume Sistólico , Função Ventricular Esquerda
5.
Cell ; 179(7): 1647-1660.e19, 2019 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-31835037

RESUMO

The process of cardiac morphogenesis in humans is incompletely understood. Its full characterization requires a deep exploration of the organ-wide orchestration of gene expression with a single-cell spatial resolution. Here, we present a molecular approach that reveals the comprehensive transcriptional landscape of cell types populating the embryonic heart at three developmental stages and that maps cell-type-specific gene expression to specific anatomical domains. Spatial transcriptomics identified unique gene profiles that correspond to distinct anatomical regions in each developmental stage. Human embryonic cardiac cell types identified by single-cell RNA sequencing confirmed and enriched the spatial annotation of embryonic cardiac gene expression. In situ sequencing was then used to refine these results and create a spatial subcellular map for the three developmental phases. Finally, we generated a publicly available web resource of the human developing heart to facilitate future studies on human cardiogenesis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Miócitos Cardíacos/metabolismo , Análise de Célula Única , Transcriptoma , Feminino , Humanos , Masculino , Morfogênese , Miócitos Cardíacos/citologia , RNA-Seq
6.
J Cardiothorac Surg ; 13(1): 74, 2018 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-29921307

RESUMO

BACKGROUND: The use of left ventricular assist devices (LVADs) has increased in the last decade. Major complications have been well described, but there is no data on device alarms and actual or threatening malfunction which impair quality of life and may impair outcomes. This study describes the technical problems related to the use of the HVAD® left ventricular assist device in a single center. METHODS: We retrospectively reviewed device malfunctions and outcomes in 22 patients with HVAD® left ventricular assist device followed at Karolinska University Hospital between 2011 and 2016. Device malfunction was defined by INTERMACS as a failure of one or more of the components of the LVAD system. The primary outcome was defined as death or hospitalization or unplanned urgent clinic visit due to device alarm of unknown significance or actual or threatening malfunction. Separate secondary outcomes were malfunction resulting in controller exchange and malfunction resulting in battery change. Exploratory outcomes were death, transplantation, or explantation because of recovery. RESULTS: Median age was 59 years and 19% were women. Over a mean follow-up time of 1.7 years (37 patient-years), the primary outcome occurred 30 times (0.8 events per patient-year; 0 deaths, 2 hospitalizations and 28 un-planned clinic visits). Secondary outcomes were 41 device malfunctions for 14 patients requiring 45 controller exchanges in 12 patients (1.1 events per patient-year) and 128 battery changes in 12 patients (3.5 events per patient-year). Exploratory outcomes were 8 deaths (36.4%), 7 transplantations (31.8%) and 2 explants due to recovery (9.1%). CONCLUSION: The use of HVAD® was associated with technical problems requiring frequent un-planned clinic visits and changes of controller and/or batteries. There were no deaths due to device malfunction. Further studies are warranted to evaluate the risk of device malfunction and associated reductions in quality of life and cost.


Assuntos
Equipamentos e Provisões Elétricas/efeitos adversos , Insuficiência Cardíaca/cirurgia , Coração Auxiliar/efeitos adversos , Falha de Prótese/efeitos adversos , Adulto , Idoso , Feminino , Seguimentos , Insuficiência Cardíaca/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida , Estudos Retrospectivos , Resultado do Tratamento
7.
Lakartidningen ; 1132016 06 08.
Artigo em Sueco | MEDLINE | ID: mdl-27299327

RESUMO

Modern cancer therapy has noticeably improved the prognosis for various cancer diseases, but cardiovascular side effects are not uncommon, both in short and long term. The individual patient's cardiovascular risk profile affects the risk of developing side effects. Potential underestimation of this risk can lead to life-long severe cardiac disease for a patient that has been cured from cancer. Overestimation of the risk can lead to withdrawal of life-saving cancer treatment because of potentially reversible or mild cardiac side effects. A multidisciplinary approach will lead to better handling of cardio-oncologic patients.


Assuntos
Antineoplásicos/efeitos adversos , Cardiotoxicidade , Cardiotoxicidade/diagnóstico , Cardiotoxicidade/etiologia , Cardiotoxicidade/prevenção & controle , Cardiopatias/induzido quimicamente , Humanos , Relações Interprofissionais , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Radioterapia/efeitos adversos , Fatores de Risco
8.
Stem Cell Reports ; 6(4): 607-617, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27052314

RESUMO

The intrinsic regenerative capacity of human fetal cardiac mesenchymal stromal cells (MSCs) has not been fully characterized. Here we demonstrate that we can expand cells with characteristics of cardiovascular progenitor cells from the MSC population of human fetal hearts. Cells cultured on cardiac muscle laminin (LN)-based substrata in combination with stimulation of the canonical Wnt/ß-catenin pathway showed increased gene expression of ISL1, OCT4, KDR, and NKX2.5. The majority of cells stained positive for PDGFR-α, ISL1, and NKX2.5, and subpopulations also expressed the progenitor markers TBX18, KDR, c-KIT, and SSEA-1. Upon culture of the cardiac MSCs in differentiation media and on relevant LNs, portions of the cells differentiated into spontaneously beating cardiomyocytes, and endothelial and smooth muscle-like cells. Our protocol for large-scale culture of human fetal cardiac MSCs enables future exploration of the regenerative functions of these cells in the context of myocardial injury in vitro and in vivo.


Assuntos
Proliferação de Células/genética , Células-Tronco Mesenquimais/metabolismo , Células-Tronco/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/genética , Sistema Cardiovascular/citologia , Diferenciação Celular/genética , Células Cultivadas , Coração Fetal/citologia , Perfilação da Expressão Gênica/métodos , Proteína Homeobox Nkx-2.5/genética , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Laminina/metabolismo , Células-Tronco Mesenquimais/citologia , Microscopia de Fluorescência , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-26421632

RESUMO

The human fetal heart is formed early during embryogenesis as a result of cell migrations, differentiation, and formative blood flow. It begins to beat around gestation day 22. Progenitor cells are derived from mesoderm (endocardium and myocardium), proepicardium (epicardium and coronary vessels), and neural crest (heart valves, outflow tract septation, and parasympathetic innervation). A variety of molecular disturbances in the factors regulating the specification and differentiation of these cells can cause congenital heart disease. This review explores the contribution of different cardiac progenitors to the embryonic heart development; the pathways and transcription factors guiding their expansion, migration, and functional differentiation; and the endogenous regenerative capacity of the adult heart including the plasticity of cardiomyocytes. Unfolding these mechanisms will become the basis for understanding the dynamics of specific congenital heart disease as well as a means to develop therapy for fetal as well as postnatal cardiac defects and heart failure.


Assuntos
Coração Fetal/embriologia , Células-Tronco Fetais/citologia , Cardiopatias Congênitas/embriologia , Mesoderma/citologia , Crista Neural/citologia , Diferenciação Celular , Movimento Celular , Vasos Coronários/citologia , Vasos Coronários/embriologia , Endocárdio/citologia , Endocárdio/embriologia , Coração Fetal/citologia , Humanos , Miocárdio/citologia , Pericárdio/citologia , Pericárdio/embriologia
10.
J Cardiovasc Transl Res ; 8(6): 381-4, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26149227

RESUMO

Endomyocardial delivery in the setting of active left ventricular assist device (LVAD) support has rarely been studied. The objective was to establish a protocol for endomyocardial injections during LVAD support without compromising mechanical circulation. LVAD implantation was performed in four pigs. A curved needle catheter was percutaneously inserted into the right carotid artery and positioned into the left ventricle under fluoroscopic guidance. In the setting of increasing LVAD flows (2.3-3.1 l/min), percutaneous methylene blue dye administration into the myocardium proceeded without complications in all pigs. Transection of excised hearts revealed an anterior, lateral, inferior, and septal wall distribution of methylene blue documenting injections in all four regions of the left ventricle. Ex vivo, the catheter could be maneuvered close to the LVAD inflow cannula despite augmentation of LVAD flow up to 5 l/min. Endomyocardial injections during LVAD support was found to be feasible and safe with the curved needle catheter.


Assuntos
Cateterismo Cardíaco/métodos , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/fisiopatologia , Coração Auxiliar , Injeções , Miocárdio , Radiografia Intervencionista , Animais , Modelos Animais de Doenças , Fluoroscopia , Azul de Metileno , Suínos
11.
PLoS One ; 10(3): e0120176, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25763592

RESUMO

Generation of new cardiomyocytes is critical for cardiac repair following myocardial injury, but which kind of stimuli is most important for cardiomyocyte regeneration is still unclear. Here we explore if apoptotic stimuli, manifested through caspase activation, influences cardiac progenitor up-regulation and cardiomyocyte differentiation. Using mouse embryonic stem cells as a cellular model, we show that sublethal activation of caspases increases the yield of cardiomyocytes while concurrently promoting the proliferation and differentiation of c-Kit+/α-actininlow cardiac progenitor cells. A broad-spectrum caspase inhibitor blocked these effects. In addition, the caspase inhibitor reversed the mRNA expression of genes expressed in cardiomyocytes and their precursors. Our study demonstrates that sublethal caspase-activation has an important role in cardiomyocyte differentiation and may have significant implications for promoting cardiac regeneration after myocardial injury involving exogenous or endogenous cell sources.


Assuntos
Caspase 3/metabolismo , Caspase 9/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Miócitos Cardíacos/metabolismo , Actinina/metabolismo , Animais , Apoptose , Linhagem Celular , Membrana Celular/metabolismo , Camundongos , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
13.
Coron Artery Dis ; 21(2): 65-71, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20016374

RESUMO

BACKGROUND AND AIMS: To investigate whether, in the subacute phase of acute myocardial infarction, in the peri-infarcted area the expressions of the vascular endothelial growth factor (VEGF-A) and angiopoietin (Ang) ligand receptors are depressed, and whether overexpression of these angiogens counteracts a downregulation of myocardial function. METHODS: Acute myocardial infarction was induced by left anterior descending artery ligation and overexpression through injection of human VEGF-A165 and Ang-1 plasmids. The capillary and arteriolar densities, Akt-1 phosphorylation and citrate synthase activity were measured concurrent with the expression of VEGF-A, VEGFR1 and R2, Ang-1, Ang-2 and Tie-2. RESULTS: One day after AMI, VEGR-2 was unchanged but all other measured factors in the two families were upregulated. After day 2, the Ang-2 expression increased but other measured factors decreased. After gene transfer, the vascular supply, Akt phosphorylation and citrate synthase activity were higher in the peri-infarcted area, where also the endogenous angiogenic growth factor expressions were increased. CONCLUSION: A rapid decrease in angiogenic stimulating factors occurs in the subacute phase of AMI and is related to a progressive decrease in myocardial contraction. A negative consequence of such a circuit is a successive reduction in the vascular supply and contractility in areas with reduced perfusion. These negative adaptations can be counteracted by angiogen overexpression.


Assuntos
Proteínas Angiogênicas/metabolismo , Infarto do Miocárdio/metabolismo , Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Neovascularização Fisiológica , Remodelação Ventricular , Proteínas Angiogênicas/genética , Angiopoietina-1/biossíntese , Angiopoietina-1/genética , Angiopoietina-2/metabolismo , Animais , Citrato (si)-Sintase/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Terapia Genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Isquemia Miocárdica/genética , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2 , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
Biochem Biophys Res Commun ; 373(3): 355-9, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18571496

RESUMO

Eph/ephrin signaling is pivotal in prenatal angiogenesis while its potential role in postnatal angiogenesis largely remains to be explored. Therefore its putative angiogenic and therapeutic effects were explored in endothelium and in myocardial ischemia. In culture of human aortic endothelial cells the fusion protein ephrinB2-Fc induced cell proliferation (p<0.0005) and in the murine aortic ring model ephrinB2-Fc induced increased sprouting (p<0.05). Myocardial infarction was induced by ligation of the left anterior descending artery in mouse. During the following 2 weeks mRNA of the receptor/ligand pair EphB4/ephrinB2 was expressed dichotomously (p<0.05) and other Eph/ephrin pairs were expressed to a lesser degree. Twenty-four hours after intraperitoneal administration of ephrinB2-Fc it was detected in abundance throughout the myocardium along capillaries, showing signs of increased mitosis. After 4 weeks the capillary density was increased 28% in the periinfarcted area (p<0.05) to a level not different from healthy regions of the heart where no change was observed. These results implicate that EphB4/ephrinB2 is an important signaling pathway in ischemic heart disease and its modulation may induce therapeutic angiogenesis.


Assuntos
Endotélio Vascular/metabolismo , Efrina-B2/metabolismo , Infarto do Miocárdio/metabolismo , Neovascularização Fisiológica , Animais , Aorta , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Efrina-B2/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitose/efeitos dos fármacos , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/farmacologia
15.
Stem Cells ; 26(7): 1850-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18467659

RESUMO

In order to study the ability of costimulation blockade to induce tolerance to human embryonic stem cells (HESC), severe combined immunodeficient (SCID), and immunocompetent C57BL/6 mice treated with costimulation blockade received intratesticular and intramyocardial HESC transplants. All SCID mice with intratesticular HESC transplants developed teratoma. When SCID mice were transplanted intramyocardially, only two of five mice developed teratoma-like tumors. C57BL/6 mice transplanted intratesticularly and treated with costimulation blockade all developed teratoma and were surrounded by CD4(+)CD25(+)Foxp3(+) T-cells, while isotype control treated recipients rejected their grafts. Most C57BL/6 mice transplanted intramyocardially and treated with costimulation blockade demonstrated lymphocytic infiltrates 1 month after transplantation, whereas one maintained its graft. Isolation of regulatory T-cells from intramyocardial transplanted recipients treated with costimulation blockade demonstrated specificity toward undifferentiated HESC and down-regulated naive T-cell activation toward HESC. These results demonstrate that costimulation blockade is sufficiently robust to induce tolerance to HESC in the immune-privileged environment of the testis. HESC specific regulatory T-cells developed to HESC transplanted to the heart and the success of transplantation was similar to that seen in SCID mice.


Assuntos
Transplante de Células/métodos , Células-Tronco Embrionárias/citologia , Miocárdio/metabolismo , Linfócitos T Reguladores/citologia , Testículo/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Ligante de CD40/biossíntese , Células-Tronco Embrionárias/metabolismo , Humanos , Tolerância Imunológica/imunologia , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID
16.
Cardiovasc Res ; 75(1): 178-85, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17481597

RESUMO

OBJECTIVE: This study investigates whether local sequential delivery of vascular endothelial growth factor-A(165) (VEGF-A(165)) followed by platelet-derived growth factor-BB (PDGF-BB) with alginate hydrogels could induce an angiogenic effect and functional improvement greater than single factors after myocardial infarction. METHODS: Alginate hydrogels were prepared by combining high and low molecular weight alginate. Growth factor release rates were monitored over time in vitro with 125I-labelled VEGF-A(165) and PDGF-BB included in the gels. One week after myocardial infarction was induced in Fisher rats, gels with VEGF-A(165), PDGF-BB, or both were given intra-myocardially along the border of the myocardial infarction. Vessel density was analysed in hearts and cardiac function was determined by Tissue Doppler Echocardiography. In addition, the angiogenic effect of sequenced delivery was studied in vitro in aortic rings from C57B1/6 mice. RESULTS: Alginate gels were capable of delivering VEGF-A(165) and PDGF-BB in a sustainable manner, and PDGF-BB was released more slowly than VEGF-A(165). Sequential growth factor administration led to a higher density of alpha-actin positive vessels than single factors, whereas no further increment was found in capillary density. Sequential protein delivery increased the systolic velocity-time integral and displayed a superior effect than single factors. In the aortic ring model, sequential delivery led to a higher angiogenic effect than single factor administration. CONCLUSIONS: The alginate hydrogel is an effective and promising injectable delivery system in a myocardial infarction model. Sequential growth factor delivery of VEGF-A(165) and PDGF-BB induces mature vessels and improves cardiac function more than each factor singly. This may indicate clinical utility.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Neovascularização Fisiológica , Fator de Crescimento Derivado de Plaquetas/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Alginatos , Animais , Aorta/efeitos dos fármacos , Becaplermina , Ecocardiografia , Ácido Glucurônico , Coração/diagnóstico por imagem , Coração/fisiopatologia , Ácidos Hexurônicos , Hidrogéis , Técnicas In Vitro , Injeções , Radioisótopos do Iodo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Proteínas Proto-Oncogênicas c-sis , Radiografia , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/tratamento farmacológico
17.
Cardiovasc Res ; 73(3): 481-7, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17134685

RESUMO

OBJECTIVE: To compare gene transfer of plasmid (P) and adenovirus (Ad) encoding human vascular endothelial growth factor-A165 (hVEGF-A165) angiogenic efficacy and adverse effects as regards apoptosis and ectopic expression of the transgene in a rat myocardial infarction model. METHODS: Myocardial infarction was provoked in Fisher rats. One week later, PhVEGF-A165, PLacZ, AdhVEGF-A165, or AdLacZ was transferred intramyocardially along the border of the myocardial infarction. hVEGF-A expression was detected with ELISA. Myocardial vessel density was analyzed 1 and 4 weeks after gene transfer. Apoptosis was detected by TUNEL staining. Cardiac function was assessed with Tissue Doppler Velocity Imaging. RESULTS: Although AdhVEGF-A165 had substantially higher myocardial hVEGF-A expression than PhVEGF-A165, AdhVEGF-A165 and PhVEGF-A165 induced angiogenic effects to a similar extent with maintained increased arteriolar density after 4 weeks of gene transfer (p < 0.05). The two treatments also improved left ventricular function similarly. Adenoviral gene transfer induced a higher number of TUNEL positive cells than plasmid (p < 0.02). Ectopic expression of the transgene was present with both vectors but substantially higher after adenoviral gene transfer. CONCLUSIONS: AdhVEGF-A165 has no obvious angiogenic advantage over PhVEGF-A165 but more side effects at least in a rat myocardial infarction model. This indicates that PhVEGF-A165 might be more applicable for therapeutic angiogenesis than AdhVEGF-A165.


Assuntos
Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/genética , Adenoviridae/genética , Animais , Apoptose , Arteríolas , Vasos Coronários , DNA/administração & dosagem , Ensaio de Imunoadsorção Enzimática/métodos , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Injeções , Óperon Lac , Fluxometria por Laser-Doppler , Masculino , Modelos Animais , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Fragmentos de Peptídeos , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes de Fusão/administração & dosagem , Fluxo Sanguíneo Regional , Transdução Genética/métodos , Transfecção/métodos , Transgenes , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Reprod Biomed Online ; 13(5): 712-24, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17169186

RESUMO

Recent studies have suggested that human embryonic stem cells (HESC) are immune-privileged and may thereby circumvent rejection. The expression of immunologically active molecules was studied by DNA microarray analysis and by flow cytometry. HESC were transplanted into immunologically competent mice and traced by fluorescence in-situ hybridization (FISH) and immunohistochemistry. The ability of HESC to directly and indirectly induce immune responses in CD4+ T-cells from naive and transplanted mice was studied. Their ability to induce immune responses of human CD4+ T-cells, when cultured in the presence of dendritic cells (DC) syngeneic to responder T-cells, was also analysed. HESC demonstrated expression of HLA class I and HLA class II genes, but the cell surface expression of HLA class II molecules was low even after incubation with IFNgamma. In wild-type mice, HESC could be demonstrated by FISH until 3 days after transplantation and were surrounded by heavy infiltrates of T-cells and macrophages. HESC induced a similar immune response as human fibroblast cells (HFib) on naive and immunized T-cells, both directly and in the presence of syngeneic DC. A similar response was observed in the allogeneic setting. It is concluded that HESC are immunologically inert and do not inhibit immune responses during direct or indirect antigen presentation, and they were acutely rejected in a xenogeneic setting.


Assuntos
Células-Tronco Embrionárias/imunologia , Antígenos HLA/metabolismo , Transplante de Células-Tronco/efeitos adversos , Linfócitos T/imunologia , Animais , Apresentação de Antígeno , Células Cultivadas/imunologia , Células-Tronco Embrionárias/transplante , Antígenos HLA/imunologia , Humanos , Hibridização in Situ Fluorescente , Leucócitos Mononucleares/imunologia , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Transplante Heterólogo , Transplante Homólogo
19.
Ann Med ; 38(2): 144-53, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16581700

RESUMO

AIM: As the capability of human mesenchymal stem cells (hMSC) to engraft, differentiate and improve myocardial function cannot be studied in humans, exploration was performed in a xenomodel. METHODS: The rats were divided into three groups depending on the type of rats used (Rowett nude (RNU) or Fischer rats +/- immunosuppression). Different groups were treated with intramyocardial injection of hMSC (1-2 million) either directly or three days after ligation of the left anterior descending artery (LAD). Myocardial function was investigated by echocardiography. The hMSC were identified with fluorescence in situ hybridization and myocardial differentiation was assessed by immunohistochemistry. RESULTS: When hMSC were injected directly after LAD ligation they could be identified in half (8/16) of the RNU rats (without immunosuppression) at 4 weeks. When injected 3 days after LAD ligation in immunosuppressed RNU rats they were identified in all (6/6) rats at 6 weeks. The surviving hMSC showed signs of differentiation into fibroblasts. No cardiomyocyte differentiation was observed. There was no difference in myocardial function in treated animals compared to controls. CONCLUSIONS: The hMSC survived in this xenomodel up to 6 weeks. However, hMSC required implantation into immunoincompetent animals as well as immunosuppression to survive, indicating that these cells are otherwise rejected. Furthermore, these cells did not differentiate into cardiomyocytes nor did they improve heart function in this xenomodel.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Infarto do Miocárdio/terapia , Miocárdio , Miócitos Cardíacos/imunologia , Animais , Modelos Animais de Doenças , Sobrevivência de Enxerto , Humanos , Hibridização in Situ Fluorescente , Injeções , Teste de Cultura Mista de Linfócitos , Infarto do Miocárdio/patologia , Isquemia Miocárdica/patologia , Miocárdio/patologia , Ratos , Ratos Endogâmicos F344 , Ratos Nus
20.
Biochem Biophys Res Commun ; 322(1): 292-6, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15313205

RESUMO

Therapeutic angiogenesis is a potential treatment modality for myocardial ischemia. phVEGF-A(165), phPDGF-BB, or a combination of the two were injected into the myocardial infarct border zone in rats 7 days after ligation of the coronary left anterior descending artery. Cardiac function was measured by echocardiography. Hearts were harvested 1 and 4 weeks after plasmid injection. phVEGF-A(165) increased capillary density more than phPDGF-BB, and phPDGF-BB preferentially stimulated arteriolar growth. The combination increased both capillaries and arterioles but did not enhance angiogenesis any more than single plasmid treatments did. VEGF-A(165) and the combination of phVEGF-A(165) and phPDGF-BB counteracted left ventricular dilatation after 1 week but did not counteract further deterioration.


Assuntos
Técnicas de Transferência de Genes , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Neovascularização Fisiológica , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Becaplermina , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Regulação da Expressão Gênica , Isquemia Miocárdica/genética , Isquemia Miocárdica/terapia , Plasmídeos/administração & dosagem , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...