Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Xenotransplantation ; 25(2): e12379, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29250828

RESUMO

BACKGROUND: CD4+CD25Hi FoxP3+ T (Treg) cells are a small subset of CD4+ T cells that have been shown to exhibit immunoregulatory function. Although the absolute number of Treg cells in peripheral blood lymphocytes (PBL) is very small, they play an important role in suppressing immune reactivity. Several studies have demonstrated that the number of Treg cells, rather than their intrinsic suppressive capacity, may contribute to determining the long-term fate of transplanted grafts. In this study, we analyzed Treg cells in PBL of long-term baboon recipients who have received genetically modified cardiac xenografts from pig donors. METHODS: Heterotopic cardiac xenotransplantation was performed on baboons using hearts obtained from GTKO.hCD46 (n = 8) and GTKO.hCD46.TBM (n = 5) genetically modified pigs. Modified immunosuppression regimen included antithymocyte globulin (ATG), anti-CD20, mycophenolate mofetil (MMF), cobra venom factor (CVF), and costimulation blockade (anti-CD154/anti-CD40 monoclonal antibody). FACS analysis was performed on PBLs labeled with anti-human CD4, CD25, and FoxP3 monoclonal antibodies (mAb) to analyze the percentage of Treg cells in six baboons that survived longer than 2 months (range: 42-945 days) after receiving a pig cardiac xenograft. RESULTS: Total WBC count was low due to immunosuppression in baboons who received cardiac xenograft from GTKO.hCD46 and GTKO.hCD46.hTBM donor pigs. However, absolute numbers of CD4+CD25Hi FoxP3 Treg cells in PBLs of long-term xenograft cardiac xenograft surviving baboon recipients were found to be increased (15.13 ± 1.50 vs 7.38 ± 2.92; P < .018) as compared to naïve or pre-transplant baboons. Xenograft rejection in these animals was correlated with decreased numbers of regulatory T cells. CONCLUSION: Our results suggest that regulatory T (Treg) cells may contribute to preventing or delaying xenograft rejection by controlling the activation and expansion of donor-reactive T cells, thereby masking the antidonor immune response, leading to long-term survival of cardiac xenografts.


Assuntos
Transplante de Coração , Xenoenxertos/imunologia , Linfócitos T Reguladores/imunologia , Tempo , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Forkhead/imunologia , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Transplante de Coração/métodos , Tolerância Imunológica , Terapia de Imunossupressão/métodos , Imunossupressores/farmacologia , Papio , Suínos , Transplante Heterólogo/métodos
2.
Exp Cell Res ; 323(1): 56-65, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24583397

RESUMO

Studying the proliferative ability of human bone marrow derived mesenchymal stem cells in hypoxic conditions can help us achieve the effective regeneration of ischemic injured myocardium. Cardiac-type fatty acid binding protein (FABP3) is a specific biomarker of muscle and heart tissue injury. This protein is purported to be involved in early myocardial development, adult myocardial tissue repair and responsible for the modulation of cell growth and proliferation. We have investigated the role of FABP3 in human bone marrow derived mesenchymal stem cells under ischemic conditions. MSCs from 12 donors were cultured either in standard normoxic or modified hypoxic conditions, and the differential expression of FABP3 was tested by quantitative (RT)PCR and western blot. We also established stable FABP3 expression in MSCs and searched for variation in cellular proliferation and differentiation bioprocesses affected by hypoxic conditions. We identified: (1) the FABP3 differential expression pattern in the MSCs under hypoxic conditions; (2) over-expression of FABP3 inhibited the growth and proliferation of the MSCs; however, improved their survival in low oxygen environments; (3) the cell growth factors and positive cell cycle regulation genes, such as PCNA, APC, CCNB1, CCNB2 and CDC6 were all down-regulated; while the key negative cell cycle regulation genes TP53, BRCA1, CASP3 and CDKN1A were significantly up-regulated in the cells with FABP3 overexpression. Our data suggested that FABP3 was up-regulated under hypoxia; also negatively regulated the cell metabolic process and the mitotic cell cycle. Overexpression of FABP3 inhibited cell growth and proliferation via negative regulation of the cell cycle and down-regulation of cell growth factors, but enhances cell survival in hypoxic or ischemic conditions.


Assuntos
Hipóxia Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/fisiologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Células-Tronco Mesenquimais/metabolismo , Adolescente , Adulto , Biomarcadores/metabolismo , Células da Medula Óssea/metabolismo , Ciclo Celular/genética , Diferenciação Celular , Células Cultivadas , Regulação para Baixo , Proteína 3 Ligante de Ácido Graxo , Proteínas de Ligação a Ácido Graxo/biossíntese , Proteínas de Ligação a Ácido Graxo/genética , Feminino , Células HeLa , Traumatismos Cardíacos/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Isquemia/genética , Isquemia/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , Adulto Jovem
3.
J Magn Reson Imaging ; 40(3): 691-7, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24123370

RESUMO

PURPOSE: To investigate MR lymphangiography in mice and primates with intradermal Gadofosveset and human serum albumin. Gadofosveset is a US FDA approved small molecule Gadolinium (Gd) chelate (957 Da) which reversibly binds serum albumin and temporally behaves as a macromolecule. As the structure of albumin varies among species, the affinity of Gadofosveset is optimized for human albumin. In this study, Gadofosveset premixed with 10% human serum albumin (HSA) was injected intradermally in mice and monkeys, and then MR lymphangiography was performed on a 3.0 Tesla clinical scanner. MATERIALS AND METHODS: Twenty microliters of each agent was injected intradermally at both sides of the front and back paws using a 30-gauge needle into female athymic nude mice (6-8 weeks old, n = 3 mice in each group). The performance of Gadofosveset-HSA was compared with Gd-labeled dendrimers (G4: 6 nm, G6: 10 nm) or Gd-DTPA. The target-to-muscle ratio (TMR = target signal intensity (SI)/muscle SI) was calculated at each time point. The TMRs were compared with a one-way analysis of variance followed by a Bonferroni multiple comparison test. RESULTS: Images taken as early as 2.5 min after intradermal (id) injection depicted enhanced lymph nodes using Gadofosveset-HSA (2.41 ± 0.20). Up to 7.5 min after injection, TMRs of Gadofosveset-HSA were greater than those of dendrimers (G4 or G6-Gd-DTPA: 2.24 ± 0.10, 2.12 ± 0.11, respectively). By 15 min postinjection, TMRs of Gadofosveset-HSA (2.18 ± 0.19) were comparable to Gd-labeled dendrimers (G4-Gd-DTPA: 2.37 ± 0.15, G6-Gd-DTPA: 2.25 ± 0.18). Gadofosveset-HSA and Gd labeled dendrimers resulted in satisfactory MR lymphography in mice and monkeys. CONCLUSION: Because both Gadofosveset and HSA are approved for human use and Gadofosveset clears rapidly through the kidneys, this method has advantages over Gd-dendrimers and could be used for visualizing lymphatic drainage and detecting lymph nodes.


Assuntos
Gadolínio/administração & dosagem , Linfonodos/anatomia & histologia , Imageamento por Ressonância Magnética/métodos , Compostos Organometálicos/administração & dosagem , Albumina Sérica/administração & dosagem , Animais , Meios de Contraste/administração & dosagem , Dendrímeros/administração & dosagem , Gadolínio DTPA/administração & dosagem , Humanos , Injeções Intradérmicas , Camundongos , Camundongos Nus , Saimiri , Razão Sinal-Ruído
4.
Xenotransplantation ; 21(1): 35-45, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24164510

RESUMO

BACKGROUND: Recently, we have shown that an immunosuppression regimen including costimulation blockade via anti-CD154 antibody significantly prolongs the cardiac xenograft survival in a GTKO.hCD46Tg pig-to-baboon heterotopic xenotransplantation model. Unfortunately, many coagulation disorders were observed with the use of anti-CD154 antibody, and recipient survival was markedly reduced by these complications. MATERIAL AND METHODS: In this experiment, we replaced anti-CD154 antibody with a more clinically acceptable anti-CD40 antibody while keeping the rest of the immunosuppressive regimen and the donor pig genetics the same. This was carried out to evaluate the antibody's role in xenograft survival and prevention of coagulopathies. Two available clones of anti-CD40 antibody were tested. One mouse anti-human CD40 antibody, (clone 3A8), activated B lymphocytes in vitro and only modestly suppressed antibody production in vivo. Whereas a recombinant mouse non-human primate chimeric raised against macaque CD40, (clone 2C10R4), blocked B-cell activation in vitro and completely blocked antibody production in vivo. RESULTS: The thrombotic complications seen with anti-CD154 antibody were effectively avoided but the graft survival, although extended, was not as prolonged as observed with anti-CD154 antibody treatment. The longest survival for the 3A8 antibody group was 27 days, and the longest graft survival in the 2C10R4 antibody group was 146 days. All of the grafts except two rejected and were explanted. Only two recipient baboons had to be euthanized due to unrelated complications, and the rest of the baboons remained healthy throughout the graft survival period or after graft explantation. In contrast to our anti-CD 154 antibody-treated baboons, the non-Gal antibody levels started to rise after B cells made their appearance around 8 weeks post-transplantation. CONCLUSIONS: Anti-CD40 antibody at the current dose does not induce any coagulopathies but while effective, had reduced efficacy to induce similar long-term graft survival as with anti-CD154 antibody perhaps due to ineffective control of B-cell function and antibody production at the present dose. More experiments are required to determine antibody affinity and effective dose for inducing long-term cardiac xenograft survival.


Assuntos
Anticorpos/imunologia , Antígenos CD40/imunologia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Animais , Animais Geneticamente Modificados , Formação de Anticorpos , Linfócitos B/imunologia , Ligante de CD40/imunologia , Rejeição de Enxerto/prevenção & controle , Xenoenxertos , Imunossupressores/farmacologia , Papio , Sus scrofa , Suínos , Transplante Heterólogo/métodos
5.
Blood Adv ; 4(24): 6148-6156, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33351110

RESUMO

Intrabone (IB) injection of umbilical cord blood has been proposed as a potential mechanism to improve transplant engraftment and prevent graft failure. However, conventional IB techniques produce low retention of transplanted cells in the marrow. To overcome this barrier, we developed an optimized IB (OIB) injection method using low-volume, computer-controlled slow infusion that promotes cellular retention in the marrow. Here, we compare engraftment of CD34+ cells transplanted in a myeloablative rhesus macaque (RM) model using the OIB method compared with IV delivery. RM CD34+ cells obtained by apheresis were split equally for transduction with lentiviral vectors encoding either green fluorescent protein or yellow fluorescent protein reporters. Following conditioning, one marked autologous population of CD34+ cells was injected directly IB using the OIB method and the other was injected via slow IV push into the same animal (n = 3). Daily flow cytometry of blood quantified the proportion of engrafting cells deriving from each source. Marrow retention was examined using positron emission tomography/computed tomography imaging of 89Zirconium (89Zr)-oxine-labeled CD34+ cells. CD34+ cells injected via the OIB method were retained in the marrow and engrafted in all 3 animals. However, OIB-transplanted progenitor cells did not engraft any faster than those delivered IV and contributed significantly less to hematopoiesis than IV-delivered cells at all time points. Rigorous testing of our OIB delivery system in a competitive RM myeloablative transplant model showed no engraftment advantage over conventional IV infusion. Given the increased complexity and potential risks of IB vs IV approaches, our data do not support IB transplantation as a strategy to improve hematopoietic engraftment.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Animais , Antígenos CD34 , Macaca mulatta , Radioisótopos , Zircônio
6.
Biochem Biophys Res Commun ; 390(3): 902-7, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19852944

RESUMO

Cell-based therapies have been employed with conflicting results. Whether direct injection of ex-vivo expanded autologous marrow stromal cells (MSCs) would improve the function of ischemic myocardium and enhance angiogenesis is not well defined. In a porcine model of chronic ischemia, MSCs were isolated and cultured for 4 weeks. Sixteen animals were random divided into two groups to receive either direct intramyocardial injection of autologous MSCs, or equal volumes and injections sites of saline. Cine MRI and epicardial echocardiography were performed just prior to the injections and again 6 weeks later at the time of sacrifice at which point tissue was also analyzed. Myocardial function as assessed by regional wall thickening (as measured by dobutamine stress echocardiograms) demonstrated a 40.9% improvement after cell treatment of the ischemic zone (p=0.016) whereas the saline treated animals only had a 3.7% change (p=0.82) compared to baseline. The left ventricular ejection fractions of MSC group showed 19.5% improvement from baseline 35.9+/-3.8% to 42.9+/-5.8% (p=0.049). Increased vascularity was found in the MSC group compared to controls (0.80+/-0.30 vs 0.50+/-0.19 capillary/myocyte ratio, p=0.018). Direct injection of autologous MSCs promotes angiogenesis and enhances the functional improvements following chronic myocardial ischemia. This suggests that the angiogenesis engendered by cell treatment may be physiologically meaningful by improving the contractility of ischemic myocardium.


Assuntos
Transplante de Células-Tronco Mesenquimais , Isquemia Miocárdica/cirurgia , Função Ventricular , Animais , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Injeções , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/patologia , Células Estromais/transplante , Suínos
7.
Invest Radiol ; 43(1): 7-15, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18097272

RESUMO

OBJECTIVES: To determine whether in vivo T2-weighted cardiac magnetic resonance imaging (MRI) delineates the area at risk (AAR) in 2-day-old nonreperfused myocardial infarction (MI). AAR was defined as the size of the perfusion defect on day 0. MI and the residual ischemic viable border zone comprise the AAR. MATERIALS AND METHODS: Fourteen dogs with permanent coronary artery occlusion were imaged on day 0 and day 2. The size of the AAR as measured by first-pass magnetic resonance perfusion on day 0 was compared with retrospectively determined AAR using day 2 T2-weighted MRI. Triphenyltetrazolium chloride staining was used to measure infarct size. Microspheres were used to detect residual perfusion. RESULTS: Hyperintense zones on day 2 T2-weighted magnetic resonance images accurately depicted the AAR as measured by first-pass perfusion on day 0 (38.9 +/- 3.0 vs. 36.3% +/- 3.3% of left ventricular, P = 0.07). Good correlation (R = 0.91) and Bland-Altman agreement was observed between the AAR measurements and the corresponding T2-weighted hyperintense regions. Both measures of AAR were larger than the infarcted zone (25.6% +/- 2.5% of left ventricular area; P < 0.001). CONCLUSIONS: Hyperintense regions visualized with in vivo T2-weighted cardiac MRI allow determination of the AAR 2 days postinfarction in nonreperfused MI.


Assuntos
Aumento da Imagem/métodos , Interpretação de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Infarto do Miocárdio/complicações , Infarto do Miocárdio/diagnóstico , Medição de Risco/métodos , Animais , Cães , Reperfusão , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
8.
Circulation ; 113(15): 1865-70, 2006 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-16606793

RESUMO

BACKGROUND: The aim of this study was to determine whether edema imaging by T2-weighted cardiac magnetic resonance (CMR) imaging could retrospectively delineate the area at risk in reperfused myocardial infarction. We hypothesized that the size of the area at risk during a transient occlusion would be similar to the T2-weighted hyperintense region observed 2 days later, that the T2-weighted hyperintense myocardium would show partial functional recovery after 2 months, and that the T2 abnormality would resolve over 2 months. METHODS AND RESULTS: Seventeen dogs underwent a 90-minute coronary artery occlusion, followed by reperfusion. The area at risk, as measured with microspheres (9 animals), was comparable to the size of the hyperintense zone on T2-weighted images 2 days later (43.4+/-3.3% versus 43.0+/-3.4% of the left ventricle; P=NS), and the 2 measures correlated (R=0.84). The infarcted zone was significantly smaller (23.1+/-3.7; both P<0.001). To test whether the hyperintense myocardium would exhibit partial functional recovery over time, 8 animals were imaged on day 2 and 2 months later. Systolic strain was mapped with displacement encoding with stimulated echoes. Edema, as detected by a hyperintense zone on T2-weighted images, resolved, and regional radial systolic strain partially improved from 4.9+/-0.7 to 13.1+/-1.5 (P=0.001) over 2 months. CONCLUSIONS: These findings are consistent with the premise that the T2 abnormality depicts the area at risk, a zone of reversibly and irreversibly injured myocardium associated with reperfused subendocardial infarctions. The persistence of postischemic edema allows T2-weighted CMR to delineate the area at risk 2 days after reperfused myocardial infarction.


Assuntos
Ecocardiografia/métodos , Imageamento por Ressonância Magnética , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/terapia , Reperfusão Miocárdica , Animais , Cães , Edema Cardíaco/diagnóstico , Coração/fisiopatologia , Microesferas , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Recuperação de Função Fisiológica , Reprodutibilidade dos Testes , Fatores de Risco , Fatores de Tempo
9.
J Clin Invest ; 112(3): 367-78, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12897204

RESUMO

Hepatic lipase (HL) has a well-established role in lipoprotein metabolism. However, its role in atherosclerosis is poorly understood. Here we demonstrate that HL deficiency raises the proatherogenic apoB-containing lipoprotein levels in plasma but reduces atherosclerosis in lecithin cholesterol acyltransferase (LCAT) transgenic (Tg) mice, similar to results previously observed with HL-deficient apoE-KO mice. These findings suggest that HL has functions that modify atherogenic risk that are separate from its role in lipoprotein metabolism. We used bone marrow transplantation (BMT) to generate apoE-KO and apoE-KO x HL-KO mice, as well as LCAT-Tg and LCAT-Tg x HL-KO mice, chimeric for macrophage HL gene expression. Using in situ RNA hybridization, we demonstrated localized production of HL by donor macrophages in the artery wall. We found that expression of HL by macrophages enhances early aortic lesion formation in both apoE-KO and LCAT-Tg mice, without changing the plasma lipid profile, lipoprotein lipid composition, or HL and lipoprotein lipase activities. HL does, however, enhance oxidized LDL uptake by peritoneal macrophages. These combined data demonstrate that macrophage-derived HL significantly contributes to early aortic lesion formation in two independent mouse models and identify a novel mechanism, separable from the role of HL in plasma lipoprotein metabolism, by which HL modulates atherogenic risk in vivo.


Assuntos
Apolipoproteínas E/deficiência , Arteriosclerose/etiologia , Lipase/fisiologia , Macrófagos Peritoneais/enzimologia , Fosfatidilcolina-Esterol O-Aciltransferase/fisiologia , Animais , Apolipoproteínas E/genética , Arteriosclerose/genética , Arteriosclerose/fisiopatologia , Transplante de Medula Óssea , Feminino , Expressão Gênica , Lipase/deficiência , Lipase/genética , Lipídeos/sangue , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fosfatidilcolina-Esterol O-Aciltransferase/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco
10.
Nat Commun ; 7: 11138, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27045379

RESUMO

Preventing xenograft rejection is one of the greatest challenges of transplantation medicine. Here, we describe a reproducible, long-term survival of cardiac xenografts from alpha 1-3 galactosyltransferase gene knockout pigs, which express human complement regulatory protein CD46 and human thrombomodulin (GTKO.hCD46.hTBM), that were transplanted into baboons. Our immunomodulatory drug regimen includes induction with anti-thymocyte globulin and αCD20 antibody, followed by maintenance with mycophenolate mofetil and an intensively dosed αCD40 (2C10R4) antibody. Median (298 days) and longest (945 days) graft survival in five consecutive recipients using this regimen is significantly prolonged over our recently established survival benchmarks (180 and 500 days, respectively). Remarkably, the reduction of αCD40 antibody dose on day 100 or after 1 year resulted in recrudescence of anti-pig antibody and graft failure. In conclusion, genetic modifications (GTKO.hCD46.hTBM) combined with the treatment regimen tested here consistently prevent humoral rejection and systemic coagulation pathway dysregulation, sustaining long-term cardiac xenograft survival beyond 900 days.


Assuntos
Anticorpos/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Coração , Fatores Imunológicos/farmacologia , Imunoterapia/métodos , Animais , Animais Geneticamente Modificados , Soro Antilinfocitário/farmacologia , Antígenos CD40/antagonistas & inibidores , Antígenos CD40/genética , Antígenos CD40/imunologia , Feminino , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Expressão Gênica , Humanos , Masculino , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/imunologia , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacologia , Papio , Rituximab/farmacologia , Suínos , Trombomodulina/genética , Trombomodulina/imunologia , Transgenes , Transplante Heterólogo
11.
Circulation ; 110(5): 534-9, 2004 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-15277332

RESUMO

BACKGROUND: Delayed ischemic preconditioning promotes cardioprotection via genomic reprogramming. We hypothesize that molecular regulation of mitochondrial energetics is integral to this cardioprotective program. METHODS AND RESULTS: Preconditioning was induced by use of 3 episodes of 3-minute coronary artery occlusion separated by 5 minutes of reperfusion. Twenty-four hours later, infarct size was reduced by 58% after preconditioning compared with sham-operated controls (P<0.001). Cardiac mitochondria were isolated from sham and preconditioned rat hearts. Mitochondrial respiration and ATP production were similar between the groups; however, preconditioned mitochondria exhibit modest hyperpolarization of the inner mitochondrial membrane potential (> or =22% versus control, P<0.001). After 35-minute anoxia and reoxygenation, preconditioned mitochondria demonstrated a 191+/-12% improvement in ADP-sensitive respiration (P=0.002) with preservation of electron-transfer-chain (ETC) activity versus controls. This augmented mitochondrial recovery was eradicated when preconditioning was abolished by the antioxidant 2-mercaptopropionyl glycine (2-MPG). These biochemical modulations appear to be regulated at the genomic level in that the expression of genes encoding rate-controlling complexes in the ETC was significantly upregulated in preconditioned myocardium, with a concordant induction of steady-state protein levels of cytochrome oxidase, cytochrome c, and adenine nucleotide translocase-1. 2-MPG abolished preconditioning induction of these transcripts. Moreover, transcripts of nuclear regulatory peptides known to orchestrate mitochondrial biogenesis, nuclear respiratory factor-1 and peroxisome-proliferator-activated receptor gamma coactivator 1alpha, were significantly induced in preconditioned myocardium. CONCLUSIONS: Delayed preconditioned mitochondria display increased tolerance against anoxia-reoxygenation in association with modifications in mitochondrial bioenergetics, with concordant genomic induction of a mitochondrial energetic gene regulatory program. This program appears to be mediated by reactive oxygen species signaling.


Assuntos
Complexo de Proteínas da Cadeia de Transporte de Elétrons/biossíntese , Regulação da Expressão Gênica , Precondicionamento Isquêmico , Mitocôndrias Cardíacas/metabolismo , Infarto do Miocárdio/genética , Traumatismo por Reperfusão Miocárdica/genética , Tiopronina/uso terapêutico , Trifosfato de Adenosina/biossíntese , Animais , Hipóxia Celular , Esquema de Medicação , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Metabolismo Energético , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Fenótipo , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio , Fatores de Tempo , Tiopronina/administração & dosagem , Tiopronina/farmacologia
12.
Acad Radiol ; 22(5): 646-52, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25683498

RESUMO

RATIONALE AND OBJECTIVES: To determine if intraprostatic injection of gadofosveset trisodium mixed with human serum albumin (HSA) can identify sentinel lymph nodes (LNs) draining the prostate on magnetic resonance imaging (MRI) in a canine model. MATERIALS AND METHODS: Three male canines weighing between 25.7 and 41.3 kg were anesthetized, placed in a 3-T MRI, and a needle was placed transrectally into one side of the prostate using a commercially available intrarectal needle guide. Gadofosveset trisodium premixed with 10% HSA was then administered at doses ranging from 0.1 to 2.5 mL. T1W MRI was performed immediately after injection, and two readers evaluated images for visualization of LNs draining the prostate. RESULTS: Intraprostatic injection of 0.2 mL gadofosveset trisodium premixed with HSA identified the draining periprostatic LNs in all cases. Delayed images demonstrated upper echelon nodes in the pelvis and the abdomen. Higher volume injections resulted in excessive periprostatic extravasation, whereas lower volume injections resulted in suboptimal visualization of LNs. CONCLUSIONS: We demonstrate that gadofosveset trisodium (premixed with 10% HSA) injected intraprostatically at 0.2 mL visualized LNs draining the prostate. This approach can be readily adapted for clinical applications such as sentinel LN imaging in prostate cancer patients before surgery.


Assuntos
Meios de Contraste/administração & dosagem , Gadolínio/administração & dosagem , Metástase Linfática/patologia , Imageamento por Ressonância Magnética/métodos , Compostos Organometálicos/administração & dosagem , Próstata/patologia , Animais , Modelos Animais de Doenças , Cães , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Albumina Sérica/administração & dosagem
13.
J Am Assoc Lab Anim Sci ; 53(4): 387-91, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25199095

RESUMO

Effective pain medication is important for animal stewardship and valid research results. We compared the pharmacokinetic assessments of standard, immediate-release buprenorphine (Bup IR) and a sustained-release buprenorphine formulation (Bup SR Lab) in male C57BL/6J mice, a mouse strain commonly used in biomedical research. We postulated that the administration of Bup SR Lab would achieve a more persistent blood drug concentration (>1 ng/mL) compared with single-dose Bup IR. The study assumed a blood buprenorphine concentration of 1 ng/mL as the minimum that may result in adequate analgesia, as previously reported. The 7 experimental groups included Bup IR (0.03, 0.05, 0.1, and 2 mg/kg), Bup SR Lab (0.3 and 1.2 mg/ kg), and saline placebo (0.7 mL/100 g). Blood sampling occurred at 0.5, 1, 3, 6, 12, 24, 48, and 72 h for evaluation by using a forensic ELISA. Bup IR at 0.03 and 0.05 mg/kg and Bup SR Lab at 0.3 mg/kg failed to obtain maximal blood concentrations (Cmax) above 1 ng/mL. All other doses (0.1 and 2 mg/kg Bup IR and 1.2 mg/kg Bup SR Lab) reached a Cmax above 1 ng/mL within 3 h after injection. In addition, 1.2 mg/kg Bup SR Lab and 2 mg/kg Bup IR provided blood concentrations above 1 ng/mL for up to 12 h, and 0.1 mg/kg Bup IR achieved this criterion for as long as 3 h. In conclusion, Bup SR Lab at 1.2 mg/kg and Bup IR at 0.1 or 2.0 mg/kg achieve or surpass the published threshold for adequate analgesia in mice.


Assuntos
Analgésicos Opioides/farmacocinética , Buprenorfina/farmacocinética , Dor/tratamento farmacológico , Analgésicos Opioides/administração & dosagem , Animais , Buprenorfina/administração & dosagem , Preparações de Ação Retardada , Masculino , Camundongos Endogâmicos C57BL , Medição da Dor
14.
Ann Thorac Surg ; 98(6): 2130-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25443017

RESUMO

BACKGROUND: This study was designed to test the effects of induced pluripotent stem cell (iPSC) in the treatment of chronic myocardial ischemia. METHODS: The reprogramming of passage 3 myocardial fibroblasts was performed by using the lentiviral vector containing 4 human factors: OCT4, SOX2, KLF4, and c-MYC. The iPSC colonies at P12-17 were allogeneically transplanted into ischemic myocardium of 10 swine by direct injection. Cohorts of 2 animals were sacrificed at 2, 4, 6, 8, and 12 weeks after injection. RESULTS: No signs of graft versus host disease were evident at any time points. At 2 weeks, clusters of SSEA-4-positive iPSCs were detected in the injected area. At 4 to 8 weeks, these cells started to proliferate into small spheres surrounded by thin capsules. At 12 weeks the cell clusters still existed, but decreased in size and numbers. The cells inside these masses were homogeneous with no sign of differentiation into any specific lineage. Increased smooth muscle actin or vWF positive cells were found inside and around the iPSC clusters, compared with non-injected areas. By real-time polymerase chain reaction, the levels of VEGF, basic FGF, and ANRT expression were significantly higher in the iPSC-treated myocardium compared with untreated areas. These results suggest that iPSCs contributed to angiogenesis. CONCLUSIONS: Allogeneically transplanted pig iPSCs proliferated despite an ischemic environment in the first 2 months and survived for at least 3 months in immunocompetent hosts. Transplanted iPSCs were also proangiogenic and thus might have beneficial effects on the ischemic heart diseases.


Assuntos
Isquemia Miocárdica/cirurgia , Células-Tronco Pluripotentes/transplante , Transplante de Células-Tronco/métodos , Animais , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/patologia , Fator 4 Semelhante a Kruppel , Isquemia Miocárdica/patologia , Miocárdio/patologia , Suínos , Resultado do Tratamento
15.
J Thorac Cardiovasc Surg ; 148(3): 1131-7; discussiom 1117, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25052825

RESUMO

OBJECTIVES: We sought to investigate if autologous freshly isolated regulatory T cells (Tregs) provide a protective and supportive role when cotransplanted with mesenchymal stem cells (MSCs). METHODS: In a porcine model of chronic ischemia, autologous MSCs were isolated and expanded ex vivo for 4 weeks. Autologous Treg cells were freshly isolated from 100 mL peripheral blood and purified by fluorescence-activated cell sorting. MSCs and Treg cells were then cotransplanted into the chronic ischemic myocardium of Yorkshire pigs by direct intramyocardial injection (1.2 × 10(8) MSCs plus an average of 1.5 million Treg cells in 25 injection sites). Animals were killed 6 weeks postinjection to study the fate of the cells and compare the effect of combined MSCs + Treg cells transplantation versus MSCs alone. RESULTS: The coinjection of MSCs along with Tregs was safe and no deleterious side effects were observed. Six weeks after injection of the cell combination, spherical MSCs clusters with thin layer capsules were found in the injected areas. In animals treated with MSCs only, the MSC clusters were less organized and not encapsulated. Immunofluorescent staining showed CD25+ cells among the CD90+ (MSC marker) cells, suggesting that the injected Treg cells remained present locally, and survived. Factor VIII+ cells were also prevalent suggesting new angiogenesis. We found no evidence that coinjections were associated with the generation of cardiac myocytes. CONCLUSIONS: The cotransplantation of Treg cells with MSCs dramatically increased the MSC survival rate, proliferation, and augmented their role in angiogenesis, which suggests a new way for future clinical application of cell-based therapy.


Assuntos
Proliferação de Células , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Isquemia Miocárdica/cirurgia , Miocárdio/patologia , Linfócitos T Reguladores/transplante , Animais , Forma Celular , Sobrevivência Celular , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/imunologia , Miocárdio/metabolismo , Neovascularização Fisiológica , Sus scrofa , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fatores de Tempo , Transplante Autólogo
16.
J Thorac Cardiovasc Surg ; 148(3): 1106-13; discussion 1113-4, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24998698

RESUMO

OBJECTIVES: Cardiac transplantation and available mechanical alternatives are the only possible solutions for end-stage cardiac disease. Unfortunately, because of the limited supply of human organs, xenotransplantation may be the ideal method to overcome this shortage. We have recently seen significant prolongation of heterotopic cardiac xenograft survival from 3 to 12 months and beyond. METHODS: Hearts from genetically engineered piglets that were alpha 1-3 galactosidase transferase knockout and expressed the human complement regulatory gene, CD46 (groups A-C), and the human thrombomodulin gene (group D) were heterotropically transplanted in baboons treated with antithymocyte globulin, cobra venom factor, anti-CD20 antibody, and costimulation blockade (anti-CD154 antibody [clone 5C8]) in group A, anti-CD40 antibody (clone 3A8; 20 mg/kg) in group B, clone 2C10R4 (25 mg/kg) in group C, or clone 2C10R4 (50 mg/kg) in group D, along with conventional nonspecific immunosuppressive agents. RESULTS: Group A grafts (n = 8) survived for an average of 70 days, with the longest survival of 236 days. Some animals in this group (n = 3) developed microvascular thrombosis due to platelet activation and consumption, which resulted in spontaneous hemorrhage. The median survival time was 21 days in group B (n = 3), 80 days in group C (n = 6), and more than 200 days in group D (n = 5). Three grafts in group D are still contracting well, with the longest ongoing graft survival surpassing the 1-year mark. CONCLUSIONS: Genetically engineered pig hearts (GTKOhTg.hCD46.hTBM) with modified targeted immunosuppression (anti-CD40 monoclonal antibody) achieved long-term cardiac xenograft survival. This potentially paves the way for clinical xenotransplantation if similar survival can be reproduced in an orthotopic transplantation model.


Assuntos
Galactosiltransferases/deficiência , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Transplante de Coração/efeitos adversos , Proteína Cofatora de Membrana/metabolismo , Trombomodulina/metabolismo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Quimioterapia Combinada , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Imunossupressores/farmacologia , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/imunologia , Contração Miocárdica , Papio , Suínos , Trombomodulina/genética , Trombomodulina/imunologia , Fatores de Tempo , Transplante Heterólogo , Função Ventricular Esquerda , Pressão Ventricular
17.
Lymphat Res Biol ; 12(1): 32-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24502282

RESUMO

UNLABELLED: BACKGROUND-RATIONALE: To investigate whether interstitial injection of gadofosveset trisodium (Ablavar®, Lantheus Medical, North Billerica, MA) would be suitable for thoracic duct (TD) imaging in a pig model. METHODS AND RESULTS: Gadofosveset trisodium alone or premixed with 10% human serum albumin (HSA) was administered intradermally in the extremities of pigs at varying doses to visualize the TD by MRI. Two blinded readers evaluated MRIs for TD visibility. The inter-observer variability for all MR imaging sessions was assessed using the Spearman rank correlation test. MR lymphography using gadofosveset trisodium premixed with HSA yielded superior visualization of the TD compared to gadofosveset trisodium alone, with a high inter-observer agreement (correlation coefficient of 0.88 (p=0.00000115)). CONCLUSIONS: We demonstrate that gadofosveset trisodium (premixed with 10%HSA) can be injected intradermally in order to perform MR lymphography of the thoracic duct. Since this agent is already FDA approved for MR imaging, the off-label use of it for imaging of the thoracic duct in humans is feasible, and the approach may prove to be beneficial for patients with TD abnormalities.


Assuntos
Meios de Contraste/administração & dosagem , Gadolínio/administração & dosagem , Linfografia/métodos , Compostos Organometálicos/administração & dosagem , Ducto Torácico , Animais , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Projetos Piloto , Suínos
18.
Ann Thorac Surg ; 91(4): 1206-12, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21353199

RESUMO

BACKGROUND: Marrow stromal cells (MSCs) are reportedly able to improve ventricular function after myocardial infarction through the paracrine effect or regenerating myocytes. However, the evidence to prove that is scant. In this animal study, we employed MSCs isolated from transgenic pigs designed to express enhanced green fluorescent proteins as the donor to study the fate of the cells after allogeneic transplantation. METHODS: Green MSCs prepared from transgenic pigs were allogeneically transplanted into chronic ischemic myocardium of 8 Yorkshire pigs by direct intramyocardial injection (total 1.2 × 10(8) cells in 2.5 mL saline, with 25 injection sites). Cohorts of 2 animals were sacrificed at 1, 2, 4, and 6 weeks, and 3 months after injection to study the fate of the injected cells. RESULTS: Allogeneic injection of the green MSCs is safe; no observable side effects or signs of graft versus host disease were observed. By 4',6-diamidino-2-phenylindole (DAPI) counterstained frozen sections, the green cells were found migrating from the injected area into deeper layers of myocardium over the course of 1 to 6 weeks. By immunofluorescent staining, the green cells were associated with smooth muscle actin or von Willebrand factor positive cells, suggesting that the transplanted cells were contributing to the formation of new vessels. We found no evidence that these cells were associated with the new generation of cardiac myocytes. Three months after injection, clusters of MSCs still can be found in the middle layer of ischemic myocardium; however, no unlimited cell growth was found. CONCLUSIONS: Allogeneic transplantation of green MSCs can be safely used to elucidate the mechanisms of cell-based therapy. The benefits of this therapy appear mainly due to the angiogenesis, not the regeneration, of cardiac myocytes.


Assuntos
Vasos Sanguíneos/citologia , Células da Medula Óssea/citologia , Diferenciação Celular , Isquemia Miocárdica/cirurgia , Células Estromais/citologia , Células Estromais/transplante , Animais , Animais Geneticamente Modificados , Células Cultivadas , Suínos
19.
Stem Cell Res ; 4(2): 117-28, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20172499

RESUMO

This study sought to identify the gene expression patterns of porcine bone marrow-derived MSC in response to hypoxia and to investigate novel specific hypoxic targets that may have a role in determining MSC proliferation/survival and differentiation. MSC from 15 animals were incubated in 1% oxygen and 8% carbon dioxide for 6, 12, and 24 h. RNA samples were isolated and assayed with Affymetrix porcine arrays and quantitative reverse-transcription PCR. Significant gene expression levels among the four groups of normoxia, 6-, 12-, and 24-h hypoxia were identified. The pattern in the 12-h hypoxia group was similar to that of the 24-h group. Of 23,924 probes, 377 and 210 genes were regulated in the 6- and 24-h hypoxia groups, respectively. Functional classification of the hypoxic regulated genes was mainly clustered in cell proliferation and response to stress. However, the major upregulated genes in the 6-h group were activated in cell cycle phases; the genes in the 24-h hypoxia were evenly separated into cell differentiation, apoptosis, and cellular metabolic processes. Twenty-eight genes were upregulated in all hypoxia groups; these genes are considered as hypoxic targets. Our results identified a genome-wide hypoxia-induced gene expression pattern in porcine MSC. This study provides a global view of molecular events in the cells during exposure to hypoxia and revealed a set of novel candidate hypoxic targets.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Hipóxia Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Hipóxia Celular/genética , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos
20.
Metabolism ; 58(4): 568-75, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19303980

RESUMO

Lecithin-cholesterol acyltransferase (LCAT), a key enzyme in high-density lipoprotein (HDL) metabolism, has been proposed to have atheroprotective properties by promoting reverse cholesterol transport. Overexpression of LCAT in various animal models, however, has led to conflicting results on its overall effect on lipoproteins and atherosclerosis. In this study, the effect of overexpression of LCAT in nonhuman primates on lipoprotein metabolism is examined. Human LCAT was expressed with adenovirus in squirrel monkeys (n = 8), resulting on day 4 in a 22-fold increase of LCAT activity (257 +/- 23 vs 5618 +/- 799 nmol mL(-1) h(-1), P < .0001). At its peak, LCAT was found to nearly double the level of HDL cholesterol from baseline (113 +/- 7 vs 260 +/- 24 mg/dL, P < .01). High-density lipoprotein formed after treatment with the adenovirus was larger in size, as assessed by fast protein liquid chromatography (FPLC) analysis. By kinetic studies, it was determined that there was a decrease in apolipoprotein (Apo) A-I resident time (0.373 +/- 0.027 vs 0.685 +/- 0.045 d(-1), P < .0001) and almost a doubling in the ApoA-I synthetic rate (22 +/- 2 vs 41 +/- 3 mg kg(-1) d(-1), P < .0001), but no overall change in ApoA-I levels. In addition, increased expression of LCAT was associated with a 37% reduction of ApoB levels (12 +/- 1 vs 19 +/- 1 mg/dL, P < .05) due to increased low-density lipoprotein catabolism (fractional catabolic rate = 1.7 +/- 0.1 d(-1) in controls vs 4.2 +/- 0.3 d(-1) in LCAT-treated group, P < .05). In summary, overexpression of LCAT in nonhuman primates leads to an antiatherogenic lipoprotein profile by increasing HDL cholesterol and lowering ApoB, thus making LCAT a potential drug target for reducing atherosclerosis.


Assuntos
Adenoviridae/genética , Aterosclerose/genética , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Fosfatidilcolina-Esterol O-Aciltransferase/genética , Animais , Cromatografia Líquida , Humanos , Cinética , Masculino , Fenótipo , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo , Saimiri
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA