Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mutat ; 39(9): 1161-1172, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29858533

RESUMO

Mutations in FLNC for a long time are known in connection to neuromuscular disorders and only recently were described in association with various cardiomyopathies. Here, we report a new clinical phenotype of filaminopathy in four unrelated patients with early-onset restrictive cardiomyopathy (RCM) in combination with congenital myopathy due to FLNC mutations (NM_001458.4:c.3557C>T, p.A1186V, rs1114167361 in three probands and c.[3547G>C; 3548C>T], p.A1183L, rs1131692185 in one proband). In all cases, concurrent myopathy was confirmed by neurological examination, electromyography, and morphological studies. Three of the patients also presented with arthrogryposis. The pathogenicity of the described missense variants was verified by cellular and morphological studies and by in vivo modeling in zebrafish. Combination of in silico and experimental approaches revealed that FLNC missense variants localized in Ig-loop segments often lead to development of RCM. The described FLNC mutations associated with early-onset RCMP extend cardiac spectrum of filaminopathies and facilitate the differential diagnosis of restrictive cardiac phenotype associated with neuromuscular involvement in children.


Assuntos
Cardiomiopatia Restritiva/genética , Anormalidades Congênitas/genética , Filaminas/genética , Doenças Musculares/genética , Adolescente , Cardiomiopatia Restritiva/fisiopatologia , Pré-Escolar , Anormalidades Congênitas/fisiopatologia , Análise Mutacional de DNA , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Doenças Musculares/fisiopatologia , Mutação , Linhagem , Fenótipo
2.
Arterioscler Thromb Vasc Biol ; 35(11): 2401-11, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26381870

RESUMO

OBJECTIVE: MicroRNAs are involved in many critical functions, including angiogenesis. Ultrasound-targeted microbubble destruction (UTMD) is a noninvasive technique for targeted vascular transfection of plasmid DNA and may be well suited for proangiogenic microRNA delivery. We aimed to investigate UTMD of miR-126-3p for therapeutic angiogenesis in chronic ischemia. APPROACH AND RESULTS: The angiogenic potential of miR-126-3p was tested in human umbilical vein endothelial cells in vitro. UTMD of miR-126-3p was tested in vivo in Fischer-344 rats before and after chronic left femoral artery ligation, evaluating target knockdown, miR-126-3p and miR-126-5p expression, phosphorylated Tie2 levels, microvascular perfusion, and vessel density. In vitro, miR-126-3p-transfected human umbilical vein endothelial cells showed repression of sprouty-related protein-1 and phosphatidylinositol-3-kinase regulatory subunit 2, negative regulators of vascular endothelial growth factor and angiopoietin-1 signaling, increased phosphorylated Tie2 mediated by knockdown of phosphatidylinositol-3-kinase regulatory subunit 2 and greater angiogenic potential mediated by both vascular endothelial growth factor/vascular endothelial growth factor R2 and angiopoietin-1 /Tie2 effects. UTMD of miR-126-3p resulted in targeted vascular transfection, peaking early after delivery and lasting for >3 days, and resulting in inhibition of sprouty-related protein-1 and phosphatidylinositol-3-kinase regulatory subunit 2, with minimal uptake in remote organs. Finally, UTMD of miR-126-3p to chronic ischemic hindlimb muscle resulted in improved perfusion, vessel density, enhanced arteriolar formation, pericyte coverage, and phosphorylated Tie2 levels, without affecting miR-126-5p or delta-like 1 homolog levels. CONCLUSIONS: UTMD of miR-126 results in improved tissue perfusion and vascular density in the setting of chronic ischemia by repressing sprouty-related protein-1 and phosphatidylinositol-3-kinase regulatory subunit 2 and enhancing vascular endothelial growth factor and angiopoietin-1 signaling, with no effect on miR-126-5p. UTMD is a promising platform for microRNA delivery, with applications for therapeutic angiogenesis.


Assuntos
Terapia Genética/métodos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Isquemia/terapia , MicroRNAs/metabolismo , Microvasos/fisiopatologia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Transfecção/métodos , Ultrassom , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Membro Posterior , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatologia , Masculino , MicroRNAs/genética , Microbolhas , Microcirculação , Ratos Endogâmicos F344 , Fluxo Sanguíneo Regional , Fatores de Tempo
3.
Front Genet ; 10: 608, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31297131

RESUMO

Even though genetic studies of individuals with neuromuscular diseases have uncovered the molecular background of many cardiac disorders such as cardiomyopathies and inherited arrhythmic syndromes, the genetic cause of a proportion of cardiomyopathies associated with neuromuscular phenotype still remains unknown. Here, we present an individual with a combination of cardiomyopathy and limb-girdle type muscular dystrophy where whole exome sequencing identified myoferlin (MYOF)-a member of the Ferlin protein family and close homolog of DYSF-as the most likely candidate gene. The disease-causative role of the identified variant c.[2576delG; 2575G>C], p.G859QfsTer8 is supported by functional studies in vitro using the primary patient's skeletal muscle mesenchymal progenitor cells, including both RNA sequencing and morphological studies, as well as recapitulating the muscle phenotype in vivo in zebrafish. We provide the first evidence supporting a role of MYOF in human muscle disease.

4.
J Am Heart Assoc ; 6(2)2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28174168

RESUMO

BACKGROUND: Cardiomyocyte-specific transgenic mice overexpressing S100A6, a member of the family of EF-hand calcium-binding proteins, develop less cardiac hypertrophy, interstitial fibrosis, and myocyte apoptosis after permanent coronary ligation, findings that support S100A6 as a potential therapeutic target after acute myocardial infarction. Our purpose was to investigate S100A6 gene therapy for acute myocardial ischemia-reperfusion. METHODS AND RESULTS: We first performed in vitro studies to examine the effects of S100A6 overexpression and knockdown in rat neonatal cardiomyocytes. S100A6 overexpression improved calcium transients and protected against apoptosis induced by hypoxia-reoxygenation via enhanced calcineurin activity, whereas knockdown of S100A6 had detrimental effects. For in vivo studies, human S100A6 plasmid or empty plasmid was delivered to the left ventricular myocardium by ultrasound-targeted microbubble destruction in Fischer-344 rats 2 days prior to a 30-minute ligation of the left anterior descending coronary artery followed by reperfusion. Control animals received no therapy. Pretreatment with S100A6 gene therapy yielded a survival advantage compared to empty-plasmid and nontreated controls. S100A6-pretreated animals had reduced infarct size and improved left ventricular systolic function, with less myocyte apoptosis, attenuated cardiac hypertrophy, and less cardiac fibrosis. CONCLUSIONS: S100A6 overexpression by ultrasound-targeted microbubble destruction helps ameliorate myocardial ischemia-reperfusion, resulting in lower mortality and improved left ventricular systolic function post-ischemia-reperfusion via attenuation of apoptosis, reduction in cardiac hypertrophy, and reduced infarct size. Our results indicate that S100A6 is a potential therapeutic target for acute myocardial infarction.


Assuntos
Apoptose , Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Infarto do Miocárdio/genética , Traumatismo por Reperfusão Miocárdica/complicações , Miócitos Cardíacos/metabolismo , RNA/genética , Proteína A6 Ligante de Cálcio S100/genética , Animais , Animais Recém-Nascidos , Western Blotting , Proteínas de Ciclo Celular/biossíntese , Modelos Animais de Doenças , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/patologia , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Proteína A6 Ligante de Cálcio S100/biossíntese , Transdução de Sinais
5.
Oncotarget ; 7(43): 69489-69506, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27542226

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is characterized by an intense fibrotic reaction termed tumor desmoplasia, which is in part responsible for its aggressiveness. Endothelial cells have been shown to display cellular plasticity in the form of endothelial-to-mesenchymal transition (EndMT) that serves as an important source of fibroblasts in pathological disorders, including cancer. Angiogenic co-receptor, neuropilin-1 (NRP- 1) actively binds TGFß1, the primary mediator of EndMT and is involved in oncogenic processes like epithelial-to-mesenchymal transition (EMT). NRP-1 and TGFß1 signaling have been shown to be aberrantly up-regulated in PDAC. We report herein a positive correlation between NRP-1 levels, EndMT and fibrosis in human PDAC xenografts. Loss of NRP-1 in HUVECs limited TGFß1-induced EndMT as demonstrated by gain of endothelial and loss of mesenchymal markers, while maintaining endothelial cell architecture. Knockdown of NRP-1 down-regulated TGFß canonical signaling (pSMAD2) and associated pro-fibrotic genes. Overexpression of NRP-1 exacerbated TGFß1-induced EndMT and up-regulated TGFß signaling and expression of pro-fibrotic genes. In vivo, loss of NRP-1 attenuated tumor perfusion and size, accompanied by reduction in EndMT and fibrosis. This study defines a previously unrecognized role of NRP-1 in regulating TGFß1-induced EndMT and fibrosis, and advocates NRP-1 as a therapeutic target to reduce tumor fibrosis and PDAC progression.


Assuntos
Carcinoma Ductal Pancreático/genética , Transição Epitelial-Mesenquimal/genética , Neuropilina-1/genética , Neoplasias Pancreáticas/genética , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/terapia , Linhagem Celular Tumoral , Células Cultivadas , Quimiorradioterapia , Tratamento Farmacológico , Feminino , Fibrose/genética , Fibrose/metabolismo , Humanos , Masculino , Neuropilina-1/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Interferência de RNA , Ratos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Artigo em Inglês | MEDLINE | ID: mdl-26229686

RESUMO

BACKGROUND: Left ventricular hypertrophy (LVH) is commonly found in chronic dialysis (CD) recipients, and is associated with impaired microvascular cardiac perfusion and heart failure. In response to LVH and cardiac ischemia, early outgrowth pro-angiogenic cellS(EPCs) mobilize from the bone marrow to facilitate angiogenesis and endothelial repair. In the general population, EPC number and function correlate inversely with cardiovascular risk. In end-stage renal disease (ESRD), EPC number and function are generally reduced. OBJECTIVES: To test whether left ventricular abnormalities retain their potent ability to promote EPC reparative responses in the setting of ESRD. DESIGN: Cross-sectional study. SETTING: St. Michael's Hospital, Toronto, Ontario, Canada. PATIENTS: 47 prevalent chronic dialysis recipients. MEASUREMENTS: (1) circulating CD34(+) and CD133(+) EPC number, (2) cultured EPC migratory ability, in vitro differentiation potential, and apoptosis rate, and (3) cardiac magnetic resonance-measured LV mass, volume and ejection fraction. METHODS: Bivariate correlation analysis was performed with Spearman's rho test. RESULTS: Of the 47 patients (mean age: 54 ± 13 years), the mean delivered urea reduction was 74 ± 10 %. Mean LV mass was 123 ± 38 g. Circulating CD34(+) and CD133(+) EPCs represented 0.14 % (IQR: 0.05 - 0.29 %) and 0.05 % (IQR: 0.01 - 0.10 %) of peripheral blood mononuclear cells. There were no significant correlations between any EPC parameter and measures of LV mass or ejection fraction. LIMITATIONS: Lack of a non-ESRD control population, and the inability to measure all parameters of EPC function due to limitations in blood sampling. Our inability to measure cardiac VEGF expression prevented an assessment of changes in cardiac EPC mobilization signals. CONCLUSIONS: These data suggest that in ESRD, the reparative EPC response to cardiac hypertrophy may be blunted. Further investigation of the effects of uremia on EPC physiology and its relationship to cardiac injury are required.


CONTEXTE: L'hypertrophie ventriculaire gauche (HVG), qui est associée à la perfusion cardiaque microvasculaire alterée et à l'insuffisance cardiaque, n'est pas rare chez les patients qui reçoivent une dialyse chronique. En réponse à l'HVG et à l'ischémie cardiaque, les cellules proangiogéniques à croissance précoce (CPCP) se mobilisent au sein de la moelle osseuse afin de faciliter l'angiogenèse et la réparation endothéliale. Dans l'ensemble de la population, le nombre et l'activité des CPCP sont inversement proportionnels au risque cardiovasculaire. Dans le cas d'insuffisance rénale chronique terminale (IRT), le nombre et l'activité des CPCP sont généralement réduits. OBJECTIFS: Vérifier si les anomalies du ventricule gauche demeurent aussi efficaces à promouvoir les actions réparatrices des CPCP dans le contexte de l'IRT. CONTEXTE: St. Michael's Hospital, à Toronto, en Ontario, au Canada. PARTICIPANTS: 47 cas prévalents de patients qui reçoivent une dialyse chronique. MESURES: (1) le nombre de CPCP CD34+ et CD133+ en circulation, (2) l'habileté migratoire des CPCP, le potentiel de différentiation in vitro, le taux d'apoptose, et (3) la mesure de la masse ventriculaire gauche par imagerie cardiaque à résonnance magnétique. MÉTHODES: L'analyse de la corrélation simple a été effectuée au moyen du coefficient de corrélation des rangs de Spearman. RÉSULTATS: On a observé une réduction de 74 ± 10 % en moyenne parmi les 47 participants (moyenne d'âge : 54 ± 13 ans). La masse ventriculaire gauche était de 123 ± 38 g en moyenne. Les CPCP CD34+ et CD133+ en circulation représentaient 0,14 % (IQR : 0,05­0,29 %) et 0,05 % (IQR : 0,01­0,10 %) des cellules mononuclées de sang périphérique. On n'a observé aucune corrélation substantielle entre les paramètres relatifs aux CPCP et les mesures de la masse ventriculaire gauche, ou la fraction d'éjection. LIMITES DE L'ÉTUDE: L'absence d'une population témoin non atteinte d'IRT, de même que l'inhabilité de mesurer tous les paramètres de l'activité des CPCP, en raison des limites de l'échantillon sanguin. Notre incapacité à mesurer l'expression du facteur de croissance endothéliale vasculaire nous a empêchés d'effectuer l'analyse des modifications dans les signaux cardiaques de mobilisation des CPCP. CONCLUSIONS: Ces données suggèrent que dans le cas d'une IRT, la réponse réparatrice des CPCP à l'hypertrophie cardiaque peut être atténuée. Des observations plus poussées sur les effets de l'urémie sur la physiologie des CPCP et sur le lien avec les lésions cardiaques seraient nécessaires.

7.
Cardiovasc Res ; 101(3): 423-33, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24403316

RESUMO

AIMS: The aim of this study was to investigate anti-apoptotic gene therapy using ultrasound-mediated plasmid delivery of survivin, an inhibitor of apoptosis protein, to prevent apoptosis and to attenuate left ventricular (LV) systolic dysfunction in a model of heart failure induced by doxorubicin. METHODS AND RESULTS: Effect of survivin transduction was investigated in vitro in rat cardiomyoblasts. After survivin transduction, survivin protein was detected in cell culture supernate confirming secretion of extracellular survivin. Under doxorubicin stimulation, survivin-transduced cells had significantly reduced apoptosis; however, incubation with survivin-conditioned media also showed reduced apoptosis that was absent with null-conditioned media. Doxorubicin-induced cardiomyopathy was established in Fischer rats. Subsets of animals underwent ultrasound-mediated survivin gene delivery or empty vector gene delivery at Week 3. Control rats received doxorubicin alone. Animals were studied using PCR, immunohistochemistry, echocardiography, and invasive haemodynamic studies out to Week 6. By Week 6, LV % fractional shortening by echocardiography and systolic function by pressure-volume loops were greater in survivin treated when compared with control- and empty-treated animals. There was reduced apoptosis by TUNEL and caspase activity in survivin-treated animals compared with control and empty treated at Week 4, with reduced interstitial fibrosis at Week 6. CONCLUSION: Survivin gene therapy can attenuate the progression of LV systolic dysfunction in doxorubicin cardiomyopathy. This effect can be attributed to decreased myocyte apoptosis and prevention of maladaptive LV remodelling, by both direct myocyte transfection and potentially by paracrine mechanisms.


Assuntos
Apoptose/efeitos dos fármacos , Doxorrubicina/toxicidade , Fibrose/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Repressoras/metabolismo , Disfunção Ventricular Esquerda/terapia , Animais , Apoptose/fisiologia , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/genética , Linhagem Celular , Terapia Genética/métodos , Proteínas Inibidoras de Apoptose/genética , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Miocárdio/metabolismo , Ratos , Proteínas Repressoras/genética , Survivina , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo
8.
Can J Cardiol ; 30(11): 1444-51, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25249499

RESUMO

BACKGROUND: Perlecan is a heparan sulfate proteoglycan (HSPG) constituent of the extracellular matrix with roles in cell growth, differentiation, and angiogenesis. The role of the HS side chains in regulating in vivo angiogenesis after hind-limb ischemia is unknown. METHODS: Heparan sulfate (HS)-deficient perlecan (Hspg2(Δ3/Δ3)) mice (n = 35), containing normal perlecan core protein but deficient in HS side chains, and wild-type (n = 33) littermates underwent surgical induction of hind-limb ischemia. Laser Doppler perfusion imaging (LDPI) and contrast-enhanced ultrasonography (CEU) provided serial assessment of hind-limb perfusion. Harvested muscles underwent immunostaining for endothelial cell density (CD31), real-time reverse transcription polymerase chain reaction RT-PCR for vascular endothelial growth factor (VEGF) mRNA expression and western blot analysis for VEGF and fibroblast growth factor (FGF)2 protein expression at days 2 and 28. RESULTS: Serial LDPI showed significantly greater perfusion recovery in ischemic limbs of wild-type compared with Hspg2(Δ3/Δ3) mice. CEU showed that normalized microvascular perfusion was increased in wild-type compared with Hspg2(Δ3/Δ3) mice at day 28 (0.67 ± 0.12 vs 0.26 ± 0.08; P = 0.001). CD31-positive cell counts were significantly higher in wild-type compared with Hspg2(Δ3/Δ3) mice on day 28 (122 ± 30 cells vs 84 ± 34 cells per high-power field [HPF]; P < 0.05). Endogenous VEGF mRNA expression (P < 0.05) and VEGF protein expression (P < 0.002) were significantly decreased in the ischemic limbs of Hspg2(Δ3/Δ3) mice compared with wild-type mice at day 2 and day 28, respectively. FGF2 protein expression showed no significant differences. CONCLUSIONS: These results suggest that the HS side chains in perlecan are important mediators of the angiogenic response to ischemia through a mechanism that involves upregulation of VEGF expression.


Assuntos
Proteoglicanas de Heparan Sulfato/fisiologia , Membro Posterior/irrigação sanguínea , Isquemia/metabolismo , Neovascularização Patológica/metabolismo , Animais , Western Blotting , Proliferação de Células , Modelos Animais de Doenças , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica/fisiologia , Imuno-Histoquímica , Isquemia/complicações , Isquemia/patologia , Fluxometria por Laser-Doppler , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/etiologia , Neovascularização Patológica/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Fator A de Crescimento do Endotélio Vascular/genética
9.
Mol Ther Nucleic Acids ; 2: e94, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23695537

RESUMO

Ultrasound-targeted microbubble destruction (UTMD) can be used to deliver silencing gene therapy to tumors. We hypothesized that UTMD would be effective in suppressing angiogenesis within tumors, and that modulation of the ultrasound pulsing intervals (PI) during UTMD would affect the magnitude of target knockdown. We performed UTMD of vascular endothelial growth factor receptor-2 (VEGFR2) short hairpin (sh)RNA plasmid in an heterotopic mammary adenocarcinoma model in rats, evaluating PIs of 2, 5, 10, and 20 seconds. We demonstrated that UTMD with a PI of 10 seconds resulted in the greatest knockdown of VEGFR2 by PCR, immunostaining, western blotting, smaller tumor volumes and perfused areas, and lower tumor microvascular blood volume (MBV) and flow by contrast-enhanced ultrasound (CEU) compared with UTMD-treated tumors at 2, 5, and 20 seconds, control tumors, tumors treated with intravenous shRNA plasmid and scrambled plasmid. CEU perfusion assessment using the therapeutic probe demonstrated that tumors were fully replenished with microbubbles within 10 seconds, but incompletely replenished at PI-2 and PI-5 seconds. In conclusion, for anti-VEGFR2 cancer gene therapy by UTMD, PI of 10 seconds results in higher target knockdown and a greater anti-angiogenic effect. Complete replenishment of tumor vasculature with silencing gene-bearing microbubbles in between destructive pulses of UTMD is required to maximize the efficacy of anti-angiogenic cancer gene therapy.Molecular Therapy - Nucleic Acids (2013) 2, e94; doi:10.1038/mtna.2013.20; published online 21 May 2013.

10.
J Am Coll Cardiol ; 59(14): 1320-8, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22464261

RESUMO

OBJECTIVES: The aim of this study was to compare temporally separated vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-1 delivery with concomitant delivery or single VEGF delivery, for therapeutic angiogenesis in chronic ischemia. BACKGROUND: Single gene delivery of VEGF results in immature neovessels that ultimately regress. Endogenously, VEGF acts early to initiate angiogenesis, whereas Ang-1 acts later to induce vessel maturation. Timing VEGF and Ang-1 gene delivery to mimic endogenous angiogenesis might be more effective for sustained neovascularization. METHODS: Unilateral hindlimb ischemia was induced in 170 rats. Ultrasound-mediated gene delivery was performed with cationic microbubbles and plasmid deoxyribonucleic acid. Groups included VEGF at 2 weeks, VEGF/Ang-1 at 2 weeks, VEGF at 2 weeks with Ang-1 at 4 weeks, and untreated control subjects. At 2, 4, and 8 weeks after ligation, blood flow and flow reserve (FR) were assessed by contrast-enhanced ultrasound. Vascular density, organization, and supporting cell coverage were assessed by fluorescent microangiography and immunohistochemistry. RESULTS: In untreated control subjects, blood flow, FR, and vessel density remained reduced. The VEGF delivery improved flow and vessel density at 4 weeks; however, FR remained low, supporting cell coverage was poor, and flow and vessel density regressed by 8 weeks. The VEGF/Ang-1 co-delivery marginally increased flow and vessel density; however, FR and supporting cell coverage improved. After temporally separated VEGF and Ang-1 delivery, blood flow, vessel density, and FR increased and were sustained, with improved pericyte coverage at 8 weeks. CONCLUSIONS: In conclusion, temporally separated VEGF and Ang-1 gene therapy results in sustained and functional neovascularization.


Assuntos
Angiopoietina-1/genética , Angiopoietina-1/uso terapêutico , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Neovascularização Fisiológica/efeitos dos fármacos , Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Doença Crônica , DNA/genética , DNA/uso terapêutico , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Masculino , Plasmídeos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Fluxo Sanguíneo Regional , Fatores de Crescimento do Endotélio Vascular/genética
11.
Clin J Am Soc Nephrol ; 6(6): 1345-53, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21597025

RESUMO

BACKGROUND AND OBJECTIVES: Angiogenesis is a key response to tissue ischemia that may be impaired by uremia. Although early-outgrowth endothelial progenitor-like cells promote angiogenesis in the setting of normal renal function, cells from uremic patients are dysfunctional. When compared with conventional hemodialysis, it was hypothesized that nocturnal hemodialysis would improve the in vivo angiogenic activity of these cells in a well described model of ischemic vascular disease. DESIGN, SETTING, PARTICIPANTS, AND MEASUREMENTS: Early-outgrowth endothelial progenitor-like cells were cultured from healthy controls (n = 5) and age- and gender-matched conventional hemodialysis (12 h/wk, n = 10) and nocturnal hemodialysis (30 to 50 h/wk, n = 9) patients. Cells (5 × 10(5)) or saline were injected into the ischemic hindlimb of athymic nude rats 1 day after left common iliac artery ligation. RESULTS: Although conventional dialysis cell injection had no effect versus saline, nocturnal hemodialysis and healthy control cell injection significantly improved ischemic hindlimb perfusion and capillary density. Nocturnal hemodialysis cell injection was also associated with significant increases in endogenous angiopoietin 1 expression in the ischemic hindlimb compared with saline and conventional dialysis cell injection. CONCLUSIONS: In contrast to a conventional dialytic regimen, nocturnal hemodialysis is associated with a significantly improved ability of early-outgrowth endothelial progenitor-like cells to promote angiogenesis and thus restore perfusion in a model of ischemic vascular disease.


Assuntos
Células Endoteliais/patologia , Hemodiálise no Domicílio/métodos , Isquemia/fisiopatologia , Falência Renal Crônica/terapia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Células-Tronco/patologia , Adulto , Análise de Variância , Angiopoietina-1/metabolismo , Animais , Velocidade do Fluxo Sanguíneo , Capilares/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Membro Posterior , Humanos , Isquemia/diagnóstico por imagem , Isquemia/metabolismo , Isquemia/patologia , Isquemia/cirurgia , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Masculino , Pessoa de Meia-Idade , Ontário , Fenótipo , Ratos , Ratos Nus , Fluxo Sanguíneo Regional , Transplante de Células-Tronco , Células-Tronco/metabolismo , Fatores de Tempo , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA