Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998413

RESUMO

IKr current, a major component of cardiac repolarization, is mediated by human Ether-à-go-go-Related Gene (hERG, Kv11.1) potassium channels. The blockage of these channels by pharmacological compounds is associated to drug-induced long QT syndrome (LQTS), which is a life-threatening disorder characterized by ventricular arrhythmias and defects in cardiac repolarization that can be illustrated using cardiomyocytes derived from human-induced pluripotent stem cells (hiPS-CMs). This study was meant to assess the modification in hiPS-CMs excitability and contractile properties by BeKm-1, a natural scorpion venom peptide that selectively interacts with the extracellular face of hERG, by opposition to reference compounds that act onto the intracellular face. Using an automated patch-clamp system, we compared the affinity of BeKm-1 for hERG channels with some reference compounds. We fully assessed its effects on the electrophysiological, calcium handling, and beating properties of hiPS-CMs. By delaying cardiomyocyte repolarization, the peptide induces early afterdepolarizations and reduces spontaneous action potentials, calcium transients, and contraction frequencies, therefore recapitulating several of the critical phenotype features associated with arrhythmic risk in drug-induced LQTS. BeKm-1 exemplifies an interesting reference compound in the integrated hiPS-CMs cell model for all drugs that may block the hERG channel from the outer face. Being a peptide that is easily modifiable, it will serve as an ideal molecular platform for the design of new hERG modulators displaying additional functionalities.


Assuntos
Cálcio/metabolismo , Canal de Potássio ERG1/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Potássio/metabolismo , Venenos de Escorpião/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Antiarrítmicos/farmacologia , Canais de Cálcio/metabolismo , Diferenciação Celular , Canal de Potássio ERG1/metabolismo , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Transporte de Íons , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/fisiopatologia , Modelos Biológicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Fenetilaminas/farmacologia , Piperidinas/farmacologia , Piridinas/farmacologia , Sulfonamidas/farmacologia
2.
Stem Cells ; 34(7): 1836-45, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26891455

RESUMO

Mesenchymal stem cell (MSC) immunosuppressive functions make them attractive candidates for anti-inflammatory therapy in allergic asthma. However, the mechanisms by which they ensure therapeutic effects remain to be elucidated. In an acute mouse model of house dust mite (Der f)-induced asthma, one i.v. MSC injection was sufficient to normalize and stabilize lung function in Der f-sensitized mice as compared to control mice. MSC injection decreased in vivo airway responsiveness and decreased ex vivo carbachol-induced bronchial contraction, maintaining bronchial expression of the inhibitory type 2 muscarinic receptor. To evaluate in vivo MSC survival, MSCs were labeled with PKH26 fluorescent marker prior to i.v. injection, and 1 to 10 days later total lungs were digested to obtain single-cell suspensions. 91.5 ± 2.3% and 86.6 ± 6.3% of the recovered PKH26(+) lung cells expressed specific macrophage markers in control and Der f mice, respectively, suggesting that macrophages had phagocyted in vivo the injected MSCs. Interestingly, only PKH26(+) macrophages expressed M2 phenotype, while the innate PKH26(-) macrophages expressed M1 phenotype. Finally, the remaining 0.5% PKH26(+) MSCs expressed 10- to 100-fold more COX-2 than before injection, suggesting in vivo MSC phenotype modification. Together, the results of this study indicate that MSCs attenuate asthma by being phagocyted by lung macrophages, which in turn acquire a M2 suppressive phenotype. Stem Cells 2016;34:1836-1845.


Assuntos
Asma/patologia , Macrófagos/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Fagocitose , Animais , Asma/complicações , Asma/fisiopatologia , Broncoconstrição , Polaridade Celular , Modelos Animais de Doenças , Hipersensibilidade/complicações , Hipersensibilidade/patologia , Hipersensibilidade/fisiopatologia , Inflamação/complicações , Inflamação/patologia , Inflamação/fisiopatologia , Injeções Intravenosas , Pulmão/patologia , Camundongos Endogâmicos BALB C , Fenótipo , Pyroglyphidae/fisiologia , Hipersensibilidade Respiratória/complicações , Hipersensibilidade Respiratória/patologia , Hipersensibilidade Respiratória/fisiopatologia
3.
Stem Cell Res ; 77: 103396, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38522388

RESUMO

Mutations in the DES gene, which encodes the intermediate filament desmin, lead to desminopathy, a rare disease characterized by skeletal muscle weakness and different forms of cardiomyopathies associated with cardiac conduction defects and arrhythmias. We generated human induced pluripotent stem cells (hiPSC) from a patient carrying the DES p.R406W mutation, and employed CRISPR/Cas9 to rectify the mutation in the patient's hiPSC line and introduced the mutation in an hiPSC line from a control individual unrelated to the patient. These hiPSC lines represent useful models for delving into the mechanisms of desminopathy and developing new therapeutic approaches.


Assuntos
Desmina , Células-Tronco Pluripotentes Induzidas , Mutação , Células-Tronco Pluripotentes Induzidas/metabolismo , Humanos , Desmina/metabolismo , Desmina/genética , Linhagem Celular , Sistemas CRISPR-Cas , Técnicas de Introdução de Genes , Diferenciação Celular
4.
Cells ; 11(23)2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36497174

RESUMO

Human heart development is governed by transcription factor (TF) networks controlling dynamic and temporal gene expression alterations. Therefore, to comprehensively characterize these transcriptional regulations, day-to-day transcriptomic profiles were generated throughout the directed cardiac differentiation, starting from three distinct human- induced pluripotent stem cell lines from healthy donors (32 days). We applied an expression-based correlation score to the chronological expression profiles of the TF genes, and clustered them into 12 sequential gene expression waves. We then identified a regulatory network of more than 23,000 activation and inhibition links between 216 TFs. Within this network, we observed previously unknown inferred transcriptional activations linking IRX3 and IRX5 TFs to three master cardiac TFs: GATA4, NKX2-5 and TBX5. Luciferase and co-immunoprecipitation assays demonstrated that these five TFs could (1) activate each other's expression; (2) interact physically as multiprotein complexes; and (3) together, finely regulate the expression of SCN5A, encoding the major cardiac sodium channel. Altogether, these results unveiled thousands of interactions between TFs, generating multiple robust hypotheses governing human cardiac development.


Assuntos
Redes Reguladoras de Genes , Coração , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica , Diferenciação Celular/genética
5.
Stem Cell Res ; 60: 102688, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35101670

RESUMO

Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) is an exercise and emotional stress-induced life-threatening inherited heart rhythm disorder, characterized by an abnormal cellular calcium homeostasis. Most reported cases have been linked to mutations in the gene encoding the type 2 ryanodine receptor gene, RYR2. We generated induced pluripotent stem cells (hiPSCs) from peripheral blood mononuclear cells (PBMC) from three CPVT-affected patients, two of them carrying p.R4959Q mutation and one carrying p.Y2476D mutation. These generated hiPSC lines are a useful model to study pathophysiological consequences of RYR2 dysfunction in humans and the molecular basis of CPVT.


Assuntos
Células-Tronco Pluripotentes Induzidas , Cálcio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Mutação/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Taquicardia Ventricular
6.
Cardiovasc Res ; 117(9): 2092-2107, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-32898233

RESUMO

AIMS: Several inherited arrhythmic diseases have been linked to single gene mutations in cardiac ion channels and interacting proteins. However, the mechanisms underlying most arrhythmias, are thought to involve altered regulation of the expression of multiple effectors. In this study, we aimed to examine the role of a transcription factor (TF) belonging to the Iroquois homeobox family, IRX5, in cardiac electrical function. METHODS AND RESULTS: Using human cardiac tissues, transcriptomic correlative analyses between IRX5 and genes involved in cardiac electrical activity showed that in human ventricular compartment, IRX5 expression strongly correlated to the expression of major actors of cardiac conduction, including the sodium channel, Nav1.5, and Connexin 40 (Cx40). We then generated human-induced pluripotent stem cells (hiPSCs) derived from two Hamamy syndrome-affected patients carrying distinct homozygous loss-of-function mutations in IRX5 gene. Cardiomyocytes derived from these hiPSCs showed impaired cardiac gene expression programme, including misregulation in the control of Nav1.5 and Cx40 expression. In accordance with the prolonged QRS interval observed in Hamamy syndrome patients, a slower ventricular action potential depolarization due to sodium current reduction was observed on electrophysiological analyses performed on patient-derived cardiomyocytes, confirming the functional role of IRX5 in electrical conduction. Finally, a cardiac TF complex was newly identified, composed by IRX5 and GATA4, in which IRX5 potentiated GATA4-induction of SCN5A expression. CONCLUSION: Altogether, this work unveils a key role for IRX5 in the regulation of human ventricular depolarization and cardiac electrical conduction, providing therefore new insights into our understanding of cardiac diseases.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/genética , Doenças Ósseas/genética , Ventrículos do Coração/metabolismo , Proteínas de Homeodomínio/genética , Hipertelorismo/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Deficiência Intelectual/genética , Mutação com Perda de Função , Miócitos Cardíacos/metabolismo , Miopia/genética , Fatores de Transcrição/genética , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Doenças Ósseas/metabolismo , Doenças Ósseas/fisiopatologia , Células Cultivadas , Conexinas/genética , Conexinas/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Frequência Cardíaca , Proteínas de Homeodomínio/metabolismo , Humanos , Hipertelorismo/metabolismo , Hipertelorismo/fisiopatologia , Deficiência Intelectual/metabolismo , Deficiência Intelectual/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Miopia/metabolismo , Miopia/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma , Proteína alfa-5 de Junções Comunicantes
7.
J Am Coll Cardiol ; 75(17): 2140-2152, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32354382

RESUMO

BACKGROUND: Patients with long QT syndrome (LQTS) are predisposed to life-threatening arrhythmias. A delay in cardiac repolarization is characteristic of the disease. Pharmacotherapy, implantable cardioverter-defibrillators, and left cardiac sympathetic denervation are part of the current treatment options, but no targeted therapy for LQTS exists to date. Previous studies indicate that induced autoimmunity against the voltage-gated KCNQ1 K+ channels accelerates cardiac repolarization. OBJECTIVES: However, a causative relationship between KCNQ1 antibodies and the observed electrophysiological effects has never been demonstrated, and thus presents the aim of this study. METHODS: The authors purified KCNQ1 antibodies and performed whole-cell patch clamp experiments as well as single-channel recordings on Chinese hamster ovary cells overexpressing IKs channels. The effect of purified KCNQ1 antibodies on human cardiomyocytes derived from induced pluripotent stem cells was then studied. RESULTS: The study demonstrated that KCNQ1 antibodies underlie the previously observed increase in repolarizing IKs current. The antibodies shift the voltage dependence of activation and slow the deactivation of IKs. At the single-channel level, KCNQ1 antibodies increase the open time and probability of the channel. In models of LQTS type 2 (LQTS2) using human induced pluripotent stem cell-derived cardiomyocytes, KCNQ1 antibodies reverse the prolonged cardiac repolarization and abolish arrhythmic activities. CONCLUSIONS: Here, the authors provide the first direct evidence that KCNQ1 antibodies act as agonists on IKs channels. Moreover, KCNQ1 antibodies were able to restore alterations in cardiac repolarization and most importantly to suppress arrhythmias in LQTS2. KCNQ1 antibody therapy may thus present a novel promising therapeutic approach for LQTS2.


Assuntos
Autoanticorpos/sangue , Imunoterapia/métodos , Canal de Potássio KCNQ1/sangue , Síndrome do QT Longo/sangue , Síndrome do QT Longo/terapia , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Células HEK293 , Humanos , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/imunologia , Síndrome do QT Longo/imunologia , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/metabolismo , Estudo de Prova de Conceito , Estrutura Secundária de Proteína , Coelhos
8.
J Cell Mol Med ; 13(9B): 3703-12, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19438811

RESUMO

Cell-based therapies have great potential for the treatment of cardiovascular diseases. Recently, using a transgenic mouse model Roell et al. reported that cardiac engraftment of connexin43 (Cx43)-overexpressing myoblasts in vivo prevents post-infarct arrhythmia, a common cause of death in patients following heart attack. We carried out a similar study but in a clinically relevant context via transplantation of autologous connexin43-overexpressing myoblasts in infarcted rats. Seven days after coronary ligation, rats were randomized into three groups: a control group injected with myoblasts, a null group injected with myoblasts transduced with an empty lentivirus vector (null) and a Cx43 group injected with myoblasts transduced with a lentivirus vector encoding connexin43. In contrast to Roell's report, arrhythmia occurrence was not statistically different between groups (58%, 64% and 48% for the control (n= 12), null (n= 14) and Cx43 (n= 23) groups, respectively, P= 0.92). Using ex vivo intramural monophasic action potential recordings synchronous electrical activity was observed between connexin43-overexpressing myoblasts and host cardiomyocytes, whereas such synchrony did not occur in the null-transduced group. This suggests that ex vivo connexin43 gene transfer and expression in myoblasts improved intercellular electrical coupling between myoblasts and cardiomyocytes. However, in our model such electrical coupling was not sufficient to decrease arrhythmia induction. Therefore, we would suggest a note of caution on the use of combined Cx43 gene and cell therapy to prevent post-infarct arrhythmias in heart failure patients.


Assuntos
Arritmias Cardíacas/patologia , Conexina 43/biossíntese , Regulação da Expressão Gênica , Ventrículos do Coração/metabolismo , Mioblastos Esqueléticos/citologia , Potenciais de Ação , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Técnicas de Transferência de Genes , Insuficiência Cardíaca/patologia , Masculino , Mioblastos Esqueléticos/metabolismo , Infarto do Miocárdio/patologia , Ratos , Ratos Wistar
9.
Cardiovasc Res ; 69(2): 348-58, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16376327

RESUMO

BACKGROUND: Small scale clinical trials suggested the feasibility and the efficacy of autologous myoblast transplantation to improve ventricular function after myocardial infarction. However, these trials were hampered by unexpected episodes of life-threatening ventricular tachyarrhythmias (VT). We investigated cardiac electrical stability after myoblast transplantation to the myocardium. METHODS AND RESULTS: Seven days after coronary ligation, Wistar rats were randomized into 3 groups: a control group receiving no further treatment, a vehicle group injected with culture medium into the infarcted myocardium, and a myoblast group injected with autologous myoblasts. Holter monitoring did not discriminate the myoblast from the vehicle groups. Programmed Electrical Stimulation (PES) was performed to evaluate further a cardiac substrate for arrhythmia susceptibility. The occurrence of sustained VT during PES was similar in control and vehicle groups (5/17 and 4/19 rats, respectively; p=0.50). In contrast, 13/20 rats (65%) from the myoblast group showed at least one episode of sustained VT during PES (p<0.05 and p<0.005 versus control and vehicle groups). As a further control group, rats injected with autologous bone marrow mononuclear cells into the infarcted myocardium did not show increased susceptibility to PES. CONCLUSIONS: In an infarcted rat model, myoblast transplantation but not bone marrow mononuclear cells or myocardial injection per se induces electrical ventricular instability. Because ventricular arrhythmias are life-threatening disorders, we suggest that such preclinical evaluation should be conducted for any new source of cells to be injected into the myocardium.


Assuntos
Mioblastos Cardíacos/transplante , Infarto do Miocárdio/cirurgia , Fibrilação Ventricular/etiologia , Animais , Transplante de Medula Óssea , Cardiomegalia/etiologia , Estimulação Elétrica , Eletrocardiografia Ambulatorial , Coração/fisiopatologia , Injeções , Masculino , Infarto do Miocárdio/fisiopatologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Autólogo , Fibrilação Ventricular/fisiopatologia
11.
Stem Cell Res Ther ; 4(6): 152, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24423369

RESUMO

INTRODUCTION: Although autologous bone marrow cell (BMC) therapy has emerged as a promising treatment for acute myocardial infarction (AMI), trials reported mixed results. In the BONAMI trial, active smoking reduced cardiac function recovery after reperfused AMI. Therefore, we hypothesized that variability in the functionality of BMCs retrieved from patients with cardiovascular risk factors may partly explain these mixed results. We investigated the characteristics of progenitor cells in active smokers and non-smokers with AMI and their potential impact on BMC therapy efficacy. METHODS: Bone marrow and blood samples from 54 smoking and 47 non-smoking patients enrolled in the BONAMI cell therapy trial were analyzed. RESULTS: The white BMC and CD45dimCD34+ cell numbers were higher in active smokers (P = 0.001, P = 0.03, respectively). In marked contrast, either bone marrow or blood endothelial progenitor CD45dimCD34 + KDR + cells (EPCs) were decreased in active smokers (P = 0.005, P = 0.04, respectively). Importantly, a multivariate analysis including cardiovascular risk factors confirmed the association between active smoking and lower EPC number in bone marrow (P = 0.04) and blood (P = 0.04). Furthermore, baseline circulating EPC count predicted infarct size decrease at three months post-AMI in non-smokers (P = 0.01) but not in active smokers. Interestingly, baseline circulating EPCs were no longer predictive of cardiac function improvement in the BMC therapy group. CONCLUSIONS: These data suggest that circulating EPCs play an important role in cardiac repair post-AMI only in non-smokers and that active smoking-associated EPC alterations may participate in the impairment of cardiac function recovery observed in smokers after AMI, an effect that was overridden by BMC therapy.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Infarto do Miocárdio/cirurgia , Fumar , Adolescente , Adulto , Idoso , Antígenos CD34/metabolismo , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Antígenos Comuns de Leucócito/metabolismo , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/patologia , Remodelação Ventricular , Adulto Jovem
12.
PLoS One ; 7(12): e51991, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284842

RESUMO

BACKGROUND: To improve the efficacy of bone marrow-derived mesenchymal stem cell (MSC) therapy targeted to infarcted myocardium, we investigated whether a self-setting silanized hydroxypropyl methylcellulose (Si-HPMC) hydrogel seeded with MSC (MSC+hydrogel) could preserve cardiac function and attenuate left ventricular (LV) remodeling during an 8-week follow-up study in a rat model of myocardial infarction (MI). METHODOLOGY/PRINCIPAL FINDING: Si-HPMC hydrogel alone, MSC alone or MSC+hydrogel were injected into the myocardium immediately after coronary artery ligation in female Lewis rats. Animals in the MSC+hydrogel group showed an increase in cardiac function up to 28 days after MI and a mid-term prevention of cardiac function alteration at day 56. Histological analyses indicated that the injection of MSC+hydrogel induced a decrease in MI size and an increase in scar thickness and ultimately limited the transmural extent of MI. These findings show that intramyocardial injection of MSC+hydrogel induced short-term recovery of ventricular function and mid-term attenuation of remodeling after MI. CONCLUSION/SIGNIFICANCE: These beneficial effects may be related to the specific scaffolding properties of the Si-HPMC hydrogel that may provide the ability to support MSC injection and engraftment within myocardium.


Assuntos
Hidrogel de Polietilenoglicol-Dimetacrilato , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Remodelação Ventricular , Animais , Técnicas de Cultura de Células , Sobrevivência Celular , Rastreamento de Células , Modelos Animais de Doenças , Eletrocardiografia , Feminino , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Infarto do Miocárdio/patologia , Miocárdio/patologia , Ratos , Fatores de Tempo
13.
Stem Cell Res Ther ; 1(1): 4, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20504285

RESUMO

INTRODUCTION: Early randomized clinical trials of autologous bone marrow cardiac stem cell therapy have reported contradictory results highlighting the need for a better evaluation of protocol designs. This study was designed to quantify and compare whole body and heart cell distribution after intracoronary or peripheral intravenous injection of autologous bone marrow mononuclear cells in a porcine acute myocardial infarction model with late reperfusion. METHODS: Myocardial infarction was induced using balloon inflation in the left coronary artery in domestic pigs. At seven days post-myocardial infarction, 1 x 10(8) autologous bone marrow mononuclear cells were labeled with fluorescent marker and/or 99mTc radiotracer, and delivered using intracoronary or peripheral intravenous injection (leg vein). RESULTS: Scintigraphic analyses and Upsilon-emission radioactivity counting of harvested organs showed a significant cell fraction retained within the heart after intracoronary injection (6 +/- 1.7% of injected radioactivity at 24 hours), whereas following peripheral intravenous cell injection, no cardiac homing was observed at 24 hours and cells were mainly detected within the lungs. Importantly, no difference was observed in the percentage of retained cells within the myocardium in the presence or absence of myocardial infarction. Histological evaluation did not show arterial occlusion in both animal groups and confirmed the presence of bone marrow mononuclear cells within the injected myocardium area. CONCLUSIONS: Intravenous bone marrow mononuclear cell injection was ineffective to target myocardium. Myocardial cell distribution following intracoronary injection did not depend on myocardial infarction presence, a factor that could be useful for cardiac cell therapy in patients with chronic heart failure of non-ischemic origin or with ischemic myocardium without myocardial infarction.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Monócitos/transplante , Infarto do Miocárdio/terapia , Miocárdio/citologia , Transplante de Células-Tronco , Animais , Células da Medula Óssea/citologia , Movimento Celular , Vasos Coronários/patologia , Modelos Animais de Doenças , Infarto do Miocárdio/induzido quimicamente , Miocárdio/metabolismo , Células-Tronco/citologia , Suínos , Tecnécio
14.
Hum Gene Ther ; 20(11): 1329-43, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19606934

RESUMO

Interest has increased in the use of exogenous stem cells to optimize lung repair and serve as carriers of a therapeutic gene for genetic airway diseases such as cystic fibrosis. We investigated the survival and engraftment of exogenous stem cells after intratracheal injection, in a murine model of acute epithelial airway injury already used in gene therapy experiments on cystic fibrosis. Embryonic stem cells and mesenchymal stem cells were intratracheally injected 24 hr after 2% polidocanol administration, when epithelial airway injury was maximal. Stem cells were transfected with reporter genes immediately before administration. Reporter gene expression was analyzed in trachea-lungs and bronchoalveolar lavage, using nonfluorescence, quantitative, and sensitive methods. Enzyme-linked immunosorbent assay quantitative results showed that 0.4 to 5.5% of stem cells survived in the injured airway. Importantly, no stem cells survived in healthy airway or in the epithelial lining fluid. Using 5-bromo-4-chloro-3-indolyl-beta-d-galactopyranoside staining, transduced mesenchymal stem cells were detected in injured trachea and bronchi lumen. When the epithelium was spontaneously regenerated, the in vivo amount of engrafted mesenchymal stem cells from cell lines decreased dramatically. No stem cells from primary culture were located within the lungs at 7 days. This study demonstrated the feasibility of intratracheal cell delivery for airway diseases with acute epithelial injury.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Embrionárias/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Doenças Respiratórias/terapia , Traqueia/citologia , Análise de Variância , Animais , Lavagem Broncoalveolar , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Galactosídeos , Engenharia Genética/métodos , Vetores Genéticos , Indóis , Masculino , Camundongos , Transfecção , beta-Galactosidase
15.
Cancer Immunol Immunother ; 54(1): 78-86, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15693142

RESUMO

We have shown recently that some indigestible carbohydrate (short-chain fructo-oligosaccharides [sc-FOS]) reduced colon tumor incidence in Apc+/Min mice, and that this effect depended on a functional local immune system. In addition, IL-15 mRNA was concomitantly modulated in the mucosa. Since intraepithelial lymphocytes (IELs) are in close contact with intestinal epithelial cells, these cells are the candidates most likely to be involved in early cancer immunosurveillance. The present study documents the effects of sc-FOS on large intestine IELs (LI-IELs) from Apc+/+ or Apc+/Min mice by analyzing markers related to their phenotype, their activation status, and the cell surface IL-15/IL-5R alpha. In the colons of Apc+/Min mice, fewer LI-IELs expressed surface IL-15/IL-15R alpha. In addition, a lower number of CD4+ LI-IELs expressed CD25, although more LI-IELs expressed CD69, as compared to normal mice. The sc-FOS enriched diet caused a decrease in the proportion of CD25+ LI-IELs and an increase in the percentage of LI-IELs bearing surface IL-15/IL-15R alpha, independently of the Apc gene status. The IL-15/IL-15R alpha increase was, however, higher in Min mice, and returned to a level very similar to that of Apc+/+ mice when the latter mice were fed a low-fiber diet. The sc-FOS-enriched diet specifically induced an increase in CD69+ cells in Apc+/+ mice, and a decrease in the proportion of CD4+ CD25+ LI-IELs in Apc+/Min mice. Some of these modulations could contribute to the development of a better immune anticancer response in the early steps of cancer development.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Carboidratos da Dieta/farmacologia , Interleucina-15/imunologia , Intestino Grosso/efeitos dos fármacos , Linfócitos/imunologia , Receptores de Interleucina-2/imunologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Carboidratos da Dieta/imunologia , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Genes APC , Interleucina-15/genética , Intestino Grosso/citologia , Intestino Grosso/imunologia , Linfócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Fenótipo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , Receptores de Interleucina-15 , Receptores de Interleucina-2/efeitos dos fármacos , Receptores de Interleucina-2/genética
16.
Nutr Cancer ; 45(1): 84-92, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12791508

RESUMO

Loss of function of the Apc gene product is an early and frequent event in colorectal carcinogenesis. Altered migration of intestinal epithelial cells has been described in vivo in the Min mouse Apc+/Min model. Using cell lines established from this model we show in vitro that Apc+/Min cells are less motile than Apc+/+ cells and exhibit a disordered actin cytoskeletal network. This would increase the probabilities of the initiated cell to acquire additional genetic alterations leading to neoplasia. Butyrate, a product of indigestible carbohydrate fermentation by the colonic flora, is able to restore both motility and actin cytoskeletal organization. This feature may contribute to explain the protective effect exerted by butyrogenic diets on colon carcinogenesis in animal models.


Assuntos
Actinas/metabolismo , Proteína da Polipose Adenomatosa do Colo/fisiologia , Butiratos/farmacologia , Neoplasias Colorretais/prevenção & controle , Citoesqueleto/fisiologia , Genes APC , Actinas/efeitos dos fármacos , Actinas/genética , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Células Epiteliais , Citometria de Fluxo , Genes Supressores de Tumor , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Mutação , Reação em Cadeia da Polimerase , Sensibilidade e Especificidade , Células Tumorais Cultivadas
17.
Cell Immunol ; 223(1): 70-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12914760

RESUMO

Adenomatous polyposis coli (APC) functions are involved in the heterotypic interactions occurring between intestinal epithelial cells (IECs) and intra-epithelial lymphocytes (IELs). These interactions may be of interest in cancer prevention, since recent data provide evidence for lymphocyte mediated immunosurveillance of epithelial cancers. The present study attempts to determine if APC inactivation induces changes in the cross-talk between IEC and large intestine IEL (LI-IEL) through intercellular adhesion molecule (ICAM-1)/leukocyte function-associated (LFA-1) interactions. Mouse Apc+/+ and Apc+/Min colonocytes were co-cultivated with LI-IEL. When co-cultured with LI-IEL Apc+/Min IEC but not Apc+/+ IEC expressed high levels of ICAM-1. The presence of ICAM-1 was linked to TNFalpha production in both co-cultures and TNFR expression only in co-cultivated Apc+/Min IEC. Finally, butyrate enhanced the expression of ICAM-1 in Apc+/Min IEC co-cultured with LI-IEL, and the secretion of TNFalpha by both types of co-cultures. These events could participate in determining the Apc+/Min IEC immunogenicity under different in vivo conditions.


Assuntos
Comunicação Celular/imunologia , Genes APC/fisiologia , Molécula 1 de Adesão Intercelular/biossíntese , Intestino Grosso/citologia , Linfócitos/citologia , Receptores do Fator de Necrose Tumoral/biossíntese , Animais , Butiratos/farmacologia , Técnicas de Cocultura , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Citometria de Fluxo , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Interferon gama/imunologia , Intestino Grosso/efeitos dos fármacos , Intestino Grosso/imunologia , Intestino Grosso/metabolismo , Antígeno-1 Associado à Função Linfocitária/biossíntese , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA