Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-750317

RESUMO

@#Objective    To investigate how to establish stable mice cervical heart transplantation model. Methods    Totally, 40 male C57 mice with the age of 6-8 weeks and weight of 19-24 g were randomly divided into recipients and donors (n=20 in each group). Mice cervical heart transplantation model was established by connecting the ascending aorta of donors to the right cervical common artery of recipients through end to side anastmosis and the pulmonary artery of donors to the right external jugular vein of recipients through end to end anastmosis. Results    More than 95% recipients survived after surgery. Cold ischemia time was 15±5 min, warm ischemia time 23±6 min, and the whole operation took about 55±15 min. The recipients survived more than 30 d with functional heart grafts. Histologically, there was no difference between the heart graft one month after the transplantion and the normal heart. Conclusion    Cervical heart transplantation of mice model is reliable and feasible, which is easy to monitor the survival condition of heart graft by visual examination and palpation, which will benefit the basic research in transplantation field.

2.
J Exp Med ; 210(11): 2205-21, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-24127489

RESUMO

Using meta-analysis of eight independent transplant datasets (236 graft biopsy samples) from four organs, we identified a common rejection module (CRM) consisting of 11 genes that were significantly overexpressed in acute rejection (AR) across all transplanted organs. The CRM genes could diagnose AR with high specificity and sensitivity in three additional independent cohorts (794 samples). In another two independent cohorts (151 renal transplant biopsies), the CRM genes correlated with the extent of graft injury and predicted future injury to a graft using protocol biopsies. Inferred drug mechanisms from the literature suggested that two FDA-approved drugs (atorvastatin and dasatinib), approved for nontransplant indications, could regulate specific CRM genes and reduce the number of graft-infiltrating cells during AR. We treated mice with HLA-mismatched mouse cardiac transplant with atorvastatin and dasatinib and showed reduction of the CRM genes, significant reduction of graft-infiltrating cells, and extended graft survival. We further validated the beneficial effect of atorvastatin on graft survival by retrospective analysis of electronic medical records of a single-center cohort of 2,515 renal transplant patients followed for up to 22 yr. In conclusion, we identified a CRM in transplantation that provides new opportunities for diagnosis, drug repositioning, and rational drug design.


Assuntos
Rejeição de Enxerto/genética , Especificidade de Órgãos , Transplante/métodos , Aloenxertos , Animais , Atorvastatina , Biópsia , Estudos de Coortes , Dasatinibe , Bases de Dados como Assunto , Modelos Animais de Doenças , Aprovação de Drogas , Registros Eletrônicos de Saúde , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/genética , Rejeição de Enxerto/tratamento farmacológico , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/genética , Transplante de Coração , Ácidos Heptanoicos/farmacologia , Ácidos Heptanoicos/uso terapêutico , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Transplante de Rim , Metanálise como Assunto , Camundongos , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Pirróis/farmacologia , Pirróis/uso terapêutico , Reprodutibilidade dos Testes , Estudos Retrospectivos , Tiazóis/farmacologia , Tiazóis/uso terapêutico , Estados Unidos , United States Food and Drug Administration
3.
Stem Cells ; 31(11): 2354-63, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24038578

RESUMO

RATIONALE: Human embryonic stem cell (hESC) derivatives are attractive candidates for therapeutic use. The engraftment and survival of hESC derivatives as xenografts or allografts require effective immunosuppression to prevent immune cell infiltration and graft destruction. OBJECTIVE: To test the hypothesis that a short-course, dual-agent regimen of two costimulation-adhesion blockade agents can induce better engraftment of hESC derivatives compared to current immunosuppressive agents. METHODS AND RESULTS: We transduced hESCs with a double fusion reporter gene construct expressing firefly luciferase (Fluc) and enhanced green fluorescent protein, and differentiated these cells to endothelial cells (hESC-ECs). Reporter gene expression enabled longitudinal assessment of cell engraftment by bioluminescence imaging. Costimulation-adhesion therapy resulted in superior hESC-EC and mouse EC engraftment compared to cyclosporine therapy in a hind limb model. Costimulation-adhesion therapy also promoted robust hESC-EC and hESC-derived cardiomyocyte survival in an ischemic myocardial injury model. Improved hESC-EC engraftment had a cardioprotective effect after myocardial injury, as assessed by magnetic resonance imaging. Mechanistically, costimulation-adhesion therapy is associated with systemic and intragraft upregulation of T-cell immunoglobulin and mucin domain 3 (TIM3) and a reduced proinflammatory cytokine profile. CONCLUSIONS: Costimulation-adhesion therapy is a superior alternative to current clinical immunosuppressive strategies for preventing the post-transplant rejection of hESC derivatives. By extending the window for cellular engraftment, costimulation-adhesion therapy enhances functional preservation following ischemic injury. This regimen may function through a TIM3-dependent mechanism.


Assuntos
Anticorpos Monoclonais/farmacologia , Ciclosporina/farmacologia , Células-Tronco Embrionárias/transplante , Rejeição de Enxerto/prevenção & controle , Imunoconjugados/farmacologia , Imunossupressores/farmacologia , Prednisona/farmacologia , Abatacepte , Animais , Cardiotônicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Células Endoteliais/imunologia , Células Endoteliais/transplante , Rejeição de Enxerto/imunologia , Humanos , Tolerância Imunológica , Terapia de Imunossupressão/métodos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/cirurgia , Distribuição Aleatória
4.
PLoS One ; 8(8): e73580, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951359

RESUMO

Transgenic mouse with a stably integrated reporter gene(s) can be a valuable resource for obtaining uniformly labeled stem cells, tissues, and organs for various applications. We have generated a transgenic mouse model that ubiquitously expresses a tri-fusion reporter gene (fluc2-tdTomato-ttk) driven by a constitutive chicken ß-actin promoter. This "Tri-Modality Reporter Mouse" system allows one to isolate most cells from this donor mouse and image them for bioluminescent (fluc2), fluorescent (tdTomato), and positron emission tomography (PET) (ttk) modalities. Transgenic colonies with different levels of tri-fusion reporter gene expression showed a linear correlation between all three-reporter proteins (R(2)=0.89 for TdTomato vs Fluc, R(2)=0.94 for Fluc vs TTK, R(2)=0.89 for TdTomato vs TTK) in vitro from tissue lysates and in vivo by optical and PET imaging. Mesenchymal stem cells (MSCs) isolated from this transgenics showed high level of reporter gene expression, which linearly correlated with the cell numbers (R(2)=0.99 for bioluminescence imaging (BLI)). Both BLI (R(2)=0.93) and micro-PET (R(2)=0.94) imaging of the subcutaneous implants of Tri-Modality Reporter Mouse derived MSCs in nude mice showed linear correlation with the cell numbers and across different imaging modalities (R(2)=0.97). Serial imaging of MSCs transplanted to mice with acute myocardial infarction (MI) by intramyocardial injection exhibited significantly higher signals in MI heart at days 2, 3, 4, and 7 (p<0.01). MSCs transplanted to the ischemic hindlimb of nude mice showed significantly higher BLI and PET signals in the first 2 weeks that dropped by 4(th) week due to poor cell survival. However, laser Doppler perfusion imaging revealed that blood circulation in the ischemic limb was significantly improved in the MSCs transplantation group compared with the control group. In summary, this mouse can be used as a source of donor cells and organs in various research areas such as stem cell research, tissue engineering research, and organ transplantation.


Assuntos
Rastreamento de Células/métodos , Efeito Fundador , Genes Reporter , Transplante de Células-Tronco Mesenquimais , Camundongos Transgênicos/genética , Actinas/genética , Actinas/metabolismo , Animais , Galinhas/genética , Regulação da Expressão Gênica , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Isquemia/patologia , Isquemia/terapia , Medições Luminescentes , Solanum lycopersicum/química , Solanum lycopersicum/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Tomografia por Emissão de Pósitrons , Regiões Promotoras Genéticas
5.
Proc Natl Acad Sci U S A ; 110(31): 12643-8, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23858471

RESUMO

Organs are composites of tissue types with diverse developmental origins, and they rely on distinct stem and progenitor cells to meet physiological demands for cellular production and homeostasis. How diverse stem cell activity is coordinated within organs is not well understood. Here we describe a lineage-restricted, self-renewing common skeletal progenitor (bone, cartilage, stromal progenitor; BCSP) isolated from limb bones and bone marrow tissue of fetal, neonatal, and adult mice. The BCSP clonally produces chondrocytes (cartilage-forming) and osteogenic (bone-forming) cells and at least three subsets of stromal cells that exhibit differential expression of cell surface markers, including CD105 (or endoglin), Thy1 [or CD90 (cluster of differentiation 90)], and 6C3 [ENPEP glutamyl aminopeptidase (aminopeptidase A)]. These three stromal subsets exhibit differential capacities to support hematopoietic (blood-forming) stem and progenitor cells. Although the 6C3-expressing subset demonstrates functional stem cell niche activity by maintaining primitive hematopoietic stem cell (HSC) renewal in vitro, the other stromal populations promote HSC differentiation to more committed lines of hematopoiesis, such as the B-cell lineage. Gene expression analysis and microscopic studies further reveal a microenvironment in which CD105-, Thy1-, and 6C3-expressing marrow stroma collaborate to provide cytokine signaling to HSCs and more committed hematopoietic progenitors. As a result, within the context of bone as a blood-forming organ, the BCSP plays a critical role in supporting hematopoiesis through its generation of diverse osteogenic and hematopoietic-promoting stroma, including HSC supportive 6C3(+) niche cells.


Assuntos
Osso e Ossos/metabolismo , Cartilagem/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Osso e Ossos/citologia , Cartilagem/citologia , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Transgênicos , Células Estromais/citologia , Células Estromais/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(9): 3405-10, 2013 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-23391730

RESUMO

A goal of regenerative medicine is to identify cardiovascular progenitors from human ES cells (hESCs) that can functionally integrate into the human heart. Previous studies to evaluate the developmental potential of candidate hESC-derived progenitors have delivered these cells into murine and porcine cardiac tissue, with inconclusive evidence regarding the capacity of these human cells to physiologically engraft in xenotransplantation assays. Further, the potential of hESC-derived cardiovascular lineage cells to functionally couple to human myocardium remains untested and unknown. Here, we have prospectively identified a population of hESC-derived ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells that give rise to cardiomyocytes, endothelial cells, and vascular smooth muscle cells in vitro at a clonal level. We observed rare clusters of ROR2(+) cells and diffuse expression of KDR and PDGFRα in first-trimester human fetal hearts. We then developed an in vivo transplantation model by transplanting second-trimester human fetal heart tissues s.c. into the ear pinna of a SCID mouse. ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells were delivered into these functioning fetal heart tissues: in contrast to traditional murine heart models for cell transplantation, we show structural and functional integration of hESC-derived cardiovascular progenitors into human heart.


Assuntos
Células-Tronco Embrionárias/citologia , Feto/citologia , Coração/embriologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/transplante , Transplante de Células-Tronco , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem da Célula , Separação Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/citologia , Feto/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/citologia , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Linha Primitiva/citologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Sobrevivência de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
PLoS One ; 8(2): e56657, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437201

RESUMO

Despite advanced immunosuppression, redundancy in the molecular diversity of acute rejection (AR) often results in incomplete resolution of the injury response. We present a bioinformatics based approach for identification of these redundant molecular pathways in AR and a drug repositioning approach to suppress these using FDA approved drugs currently available for non-transplant indications. Two independent microarray data-sets from human renal allograft biopsies (n = 101) from patients on majorly Th1/IFN-y immune response targeted immunosuppression, with and without AR, were profiled. Using gene-set analysis across 3305 biological pathways, significant enrichment was found for the IL17 pathway in AR in both data-sets. Recent evidence suggests IL17 pathway as an important escape mechanism when Th1/IFN-y mediated responses are suppressed. As current immunosuppressions do not specifically target the IL17 axis, 7200 molecular compounds were interrogated for FDA approved drugs with specific inhibition of this axis. A combined IL17/IFN-y suppressive role was predicted for the antilipidemic drug Fenofibrate. To assess the immunregulatory action of Fenofibrate, we conducted in-vitro treatment of anti-CD3/CD28 stimulated human peripheral blood cells (PBMC), and, as predicted, Fenofibrate reduced IL17 and IFN-γ gene expression in stimulated PMBC. In-vivo Fenofibrate treatment of an experimental rodent model of cardiac AR reduced infiltration of total leukocytes, reduced expression of IL17/IFN-y and their pathway related genes in allografts and recipients' spleens, and extended graft survival by 21 days (p<0.007). In conclusion, this study provides important proof of concept that meta-analyses of genomic data and drug databases can provide new insights into the redundancy of the rejection response and presents an economic methodology to reposition FDA approved drugs in organ transplantation.


Assuntos
Rejeição de Enxerto , Interleucina-17/metabolismo , Transplante de Rim/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Biologia Computacional , Feminino , Fenofibrato/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Transplante de Coração/métodos , Humanos , Terapia de Imunossupressão , Interferon gama/metabolismo , Interleucina-17/imunologia , Transplante de Rim/métodos , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Análise Serial de Tecidos , Transplante Homólogo , Adulto Jovem
8.
Circ Res ; 111(7): 882-93, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22821929

RESUMO

RATIONALE: Induced pluripotent stem cells (iPSCs) hold great promise for the development of patient-specific therapies for cardiovascular disease. However, clinical translation will require preclinical optimization and validation of large-animal iPSC models. OBJECTIVE: To successfully derive endothelial cells from porcine iPSCs and demonstrate their potential utility for the treatment of myocardial ischemia. METHODS AND RESULTS: Porcine adipose stromal cells were reprogrammed to generate porcine iPSCs (piPSCs). Immunohistochemistry, quantitative PCR, microarray hybridization, and angiogenic assays confirmed that piPSC-derived endothelial cells (piPSC-ECs) shared similar morphological and functional properties as endothelial cells isolated from the autologous pig aorta. To demonstrate their therapeutic potential, piPSC-ECs were transplanted into mice with myocardial infarction. Compared with control, animals transplanted with piPSC-ECs showed significant functional improvement measured by echocardiography (fractional shortening at week 4: 27.2±1.3% versus 22.3±1.1%; P<0.001) and MRI (ejection fraction at week 4: 45.8±1.3% versus 42.3±0.9%; P<0.05). Quantitative protein assays and microfluidic single-cell PCR profiling showed that piPSC-ECs released proangiogenic and antiapoptotic factors in the ischemic microenvironment, which promoted neovascularization and cardiomyocyte survival, respectively. Release of paracrine factors varied significantly among subpopulations of transplanted cells, suggesting that transplantation of specific cell populations may result in greater functional recovery. CONCLUSIONS: In summary, this is the first study to successfully differentiate piPSCs-ECs from piPSCs and demonstrate that transplantation of piPSC-ECs improved cardiac function after myocardial infarction via paracrine activation. Further development of these large animal iPSC models will yield significant insights into their therapeutic potential and accelerate the clinical translation of autologous iPSC-based therapy.


Assuntos
Transplante de Células , Endotélio Vascular/citologia , Endotélio Vascular/transplante , Coração/fisiopatologia , Técnicas Analíticas Microfluídicas , Infarto do Miocárdio/terapia , Comunicação Parácrina/fisiologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Ecocardiografia , Endotélio Vascular/fisiologia , Feminino , Imageamento por Ressonância Magnética , Camundongos , Camundongos SCID , Modelos Animais , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Neovascularização Fisiológica , Células-Tronco Pluripotentes/fisiologia , Suínos , Porco Miniatura
9.
Circ Cardiovasc Imaging ; 5(4): 481-90, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22565608

RESUMO

BACKGROUND: Human cardiac progenitor cells have demonstrated great potential for myocardial repair in small and large animals, but robust methods for longitudinal assessment of their engraftment in humans is not yet readily available. In this study, we sought to optimize and evaluate the use of positron emission tomography (PET) reporter gene imaging for monitoring human cardiac progenitor cell (hCPC) transplantation in a mouse model of myocardial infarction. METHODS AND RESULTS: hCPCs were isolated and expanded from human myocardial samples and stably transduced with herpes simplex virus thymidine kinase (TK) PET reporter gene. Thymidine kinase-expressing hCPCs were characterized in vitro and transplanted into murine myocardial infarction models (n=57). Cardiac echocardiographic, magnetic resonance imaging and pressure-volume loop analyses revealed improvement in left ventricular contractile function 2 weeks after transplant (hCPC versus phosphate-buffered saline, P<0.03). Noninvasive PET imaging was used to track hCPC fate over a 4-week time period, demonstrating a substantial decline in surviving cells. Importantly, early cell engraftment as assessed by PET was found to predict subsequent functional improvement, implying a "dose-effect" relationship. We isolated the transplanted cells from recipient myocardium by laser capture microdissection for in vivo transcriptome analysis. Our results provide direct evidence that hCPCs augment cardiac function after their transplantation into ischemic myocardium through paracrine secretion of growth factors. CONCLUSIONS: PET reporter gene imaging can provide important diagnostic and prognostic information regarding the ultimate success of hCPC treatment for myocardial infarction.


Assuntos
Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Tomografia por Emissão de Pósitrons/métodos , Transplante de Células-Tronco , Análise de Variância , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Ecocardiografia , Técnicas de Transferência de Genes , Genes Reporter/genética , Terapia Genética , Guanina/análogos & derivados , Humanos , Imuno-Histoquímica , Microdissecção e Captura a Laser , Modelos Lineares , Imageamento por Ressonância Magnética , Camundongos , Camundongos SCID , Contração Miocárdica/fisiologia , Infarto do Miocárdio/fisiopatologia , Comunicação Parácrina/fisiologia , Fenótipo , Recuperação de Função Fisiológica , Timidina Quinase/genética , Timidina Quinase/metabolismo , Proteínas Virais/metabolismo
10.
Ann Thorac Surg ; 94(2): 542-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22560321

RESUMO

BACKGROUND: Although αß T cells are known to participate in the development of acute cardiac allograft rejection, the role of γδ T cells remains poorly understood. We hypothesized that γδ T cells contribute to acute allograft rejection thru interleukin (IL)-17 production. METHODS: Donor hearts from FVB mice (H-2q) were heterotopically transplanted into C57BL/6-wild type (WT) and γδ T cell-deficient (TCRδ-/-) recipient mice (H-2b). Overall graft survival was monitored. Graft infiltrating cell profile, including γδ T cell subtype, cytokine expression, and myeloperoxidase activity were measured by flow cytometry, TaqMan (Applied Biosystems, Carlsbad, CA) polymerase chain reaction, and myeloperoxidase assay, respectively, on postoperative days 3 and 6. RESULTS: Graft survival was prolonged in TCRδ-/- recipients compared with WT controls. Graft infiltrating cells, including CD45+, CD4+, CD8+, and Gr1+ cells were significantly decreased in TCRδ-/- recipients compared with WT. Donor hearts transplanted into TCRδ-/- recipients had reduced IL-17 and IL-6 messenger RNA expression. Corroborating the gene expression, intracellular cytokine staining showed decreased IL-17 producing cells in TCRδ-/- recipients. Finally, Vγ1+ and Vγ4+ T cells did not produce IL-17, although both represent 20% to 30% total graft infiltrating γδ T cells. CONCLUSIONS: The γδ T cells promote acute cardiac allograft rejection, presumably by producing IL-17. The γδ T cell depletion may prove beneficial in prolonging allograft survival by suppressing IL-17 production.


Assuntos
Rejeição de Enxerto/imunologia , Transplante de Coração , Interleucina-17/imunologia , Linfócitos T/imunologia , Animais , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Antígenos de Linfócitos T gama-delta
11.
J Heart Lung Transplant ; 31(1): 73-84, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21497519

RESUMO

BACKGROUND: Dendritic cells are professional antigen presenting cells that perform antigen processing and antigen presentation functions and rely on the proper functioning and distribution of the endoplasmic reticulum (ER) and Golgi apparatus and of vesicular trafficking pathways. We previously developed a model system to study the mechanisms governing inhibition of chronic rejection of heart allografts. METHODS: Heterotopic cardiac transplants were placed intra-abdominally and the major histocompatibility class (MHC) class I allochimeric molecule, [α1h1/u]-RT1.Aa, which contains donor-type (Wistar Furth, WF; RT1u) immunogenic epitopes displayed on recipient-type (ACI, RT1a) sequences, was delivered by portal vein to the recipients of heterotopic hearts. Dendritic cells were isolated from the recipient bone marrow at 1 and 3 days after transplantation and were immunostained or processed for Western blotting with anti-RhoB, translationally controlled tumor protein (TCTP), Sprouty-related (Spred1) protein, ER, and Golgi antibodies. RESULTS: Western blotting analyses showed the downregulation of RhoB GTPase, TCTP, and Spred1 in dendritic cells isolated from allochimeric molecule-treated rats. Immunostaining showed that in these cells, Spred 1 was shifted to the base of cellular processes, Rho B formed nonvesicular band in the cell equator, and TCTP was highly enriched in the cell nucleus. The Golgi apparatus was drastically reduced in size and formed a tiny nonvesicular aggregate, and the ER partially lost vesicular appearance. CONCLUSIONS: The function of allochimeric molecule in the abrogation of heart allograft rejection may rely on the downregulation of RhoB pathway components that regulate the structure and function of the ER/Golgi/vesicular trafficking pathways involved in antigen processing and presentation by dendritic cells.


Assuntos
Biomarcadores Tumorais/imunologia , Células Dendríticas/imunologia , Rejeição de Enxerto/metabolismo , Transplante de Coração/imunologia , Antígenos de Histocompatibilidade/imunologia , Proteínas Repressoras/imunologia , Tolerância ao Transplante/imunologia , Animais , Biomarcadores Tumorais/metabolismo , Western Blotting , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Antígenos de Histocompatibilidade/metabolismo , Tolerância Imunológica , Imunoglobulina E , Sistema de Sinalização das MAP Quinases , Masculino , Ratos , Ratos Endogâmicos WF , Proteínas Repressoras/metabolismo , Transplante Homólogo , Proteína Tumoral 1 Controlada por Tradução
12.
J Heart Lung Transplant ; 30(12): 1409-17, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22055099

RESUMO

BACKGROUND: IL-16 promotes the recruitment of various cells expressing CD4, a receptor for IL-16. The precise role of IL-16 in transplant rejection remains unknown; therefore, the present study investigated the contribution of IL-16 to the development of chronic rejection in heart transplants. METHODS: C-H-2(bm12)KhEg (H-2(bm12)) donor hearts were transplanted into (1) IL-16-deficient (IL-16(-/-)) C57BL/6J or (b) wild type (WT) control recipients (MHC class II mismatch). Grafts were harvested at 52 days, parenchymal rejection was assessed by the ISHLT grading system, and CAV was examined morphometrically. Graft infiltrating cells were detected 10 and 52 days after transplantation. Intragraft cytokine and chemokine profiles were assessed. To confirm the role of IL-16 in CAV development, C-H-2(bm12)KhEg (H-2(bm12)) donor hearts were transplanted into C57BL/6J WT recipients treated with (1) anti-IL-16-neutralization monoclonal antibody or (b) control immunoglobulin G. Grafts were harvested at 52 days, and CAV was quantified morphometrically. Graft-infiltrating cells were examined histologically. RESULTS: Parenchymal rejection and CAV was significantly attenuated in donor hearts transplanted into IL-16(-/-) recipient mice compared with WT controls. Donor hearts transplanted into IL-16(-/-) recipients had a significant reduction in coronary artery luminal occlusion, intima-to-media ratio, and percentage of diseased vessels. CAV was associated with decreased donor organ inflammation, as well as donor organ cytokine (IL-1ß and IL-6) and chemokine (MCP-1 and KC) protein expression. Intimal proliferation and inflammatory cell infiltration were significantly reduced in hearts transplanted into recipients treated with an IL-16-neutralization antibody. CONCLUSIONS: IL-16-deficiency reduced graft inflammatory cell recruitment, and allograft inflammatory cytokine and chemokine production. Therefore, IL-16 neutralization may provide a potential target for novel therapeutic treatment for cardiac allograft rejection.


Assuntos
Doença da Artéria Coronariana/prevenção & controle , Doença da Artéria Coronariana/fisiopatologia , Rejeição de Enxerto/prevenção & controle , Rejeição de Enxerto/fisiopatologia , Transplante de Coração/fisiologia , Interleucina-16/deficiência , Animais , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Quimiocinas/metabolismo , Doença da Artéria Coronariana/complicações , Citocinas/metabolismo , Modelos Animais de Doenças , Rejeição de Enxerto/etiologia , Interleucina-16/genética , Interleucina-16/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transplante Homólogo , Túnica Íntima/citologia , Túnica Íntima/efeitos dos fármacos
13.
Circulation ; 124(11 Suppl): S187-96, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21911812

RESUMO

BACKGROUND: Interleukin-17 (IL-17), which is predominantly produced by T helper 17 cells distinct from T helper 1 or T helper 2 cells, participates in the pathogenesis of infectious, autoimmune, and allergic disorders. However, the precise role in allograft rejection remains uncertain. In the present study, we investigated the role of IL-17 in acute allograft rejection using IL-17-deficient mice. METHODS AND RESULTS: Donor hearts from FVB mice were heterotopically transplanted into either C57BL/6J-IL-17-deficient (IL-17(-/-)) or -wild-type mice. Allograft survival was significantly prolonged in IL-17(-/-) recipient mice due to reduced local inflammation accompanied by decreased inflammatory cell recruitment and cytokine/chemokine expression. IL-17(-/-) recipient mice exhibited decreased IL-6 production and reciprocally enhanced regulatory T cell expansion, suggesting a contribution of regulatory T cells to prolonged allograft survival. Indeed, allografts transplanted into anti-CD25 mAb-treated IL-17(-/-) recipient mice (regulatory T cell-depleted) developed acute rejection similar to wild-type recipient mice. Surprisingly, we found that gamma delta T cells rather than CD4(+) and CD8(+) T cells were key IL-17 producers in the allografts. In support, equivalent allograft rejection was observed in Rag-2(-/-) recipient mice engrafted with either wild-type or IL-17(-/-) CD4(+) and CD8(+) T cells. Finally, hearts transplanted into gamma delta T cell-deficient mice resulted in decreased allograft rejection compared with wild-type controls. CONCLUSIONS: During heart transplantation, (1) IL-17 is crucial for acceleration of acute rejection; (2) IL-17-deficiency enhances regulatory T cell expansion; and (3) gamma delta T cells rather than CD4(+) and CD8(+) T cells are a potential source of IL-17. IL-17 neutralization may provide a potential target for novel therapeutic treatment for cardiac allograft rejection.


Assuntos
Proliferação de Células , Rejeição de Enxerto/fisiopatologia , Transplante de Coração/fisiologia , Interleucina-17/fisiologia , Linfócitos T Reguladores/patologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Células Cultivadas , Rejeição de Enxerto/patologia , Transplante de Coração/patologia , Interleucina-17/deficiência , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T Reguladores/metabolismo , Células Th17/metabolismo , Células Th17/patologia , Transplante Homólogo
14.
Circulation ; 124(11 Suppl): S27-34, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21911815

RESUMO

BACKGROUND: Although stem cell therapy has provided a promising treatment for myocardial infarction, the low survival of the transplanted cells in the infarcted myocardium is possibly a primary reason for failure of long-term improvement. Therefore, the development of novel prosurvival strategies to boost stem cell survival will be of significant benefit to this field. METHODS AND RESULTS: Cardiac progenitor cells (CPCs) were isolated from transgenic mice, which constitutively express firefly luciferase and green fluorescent protein. The CPCs were transduced with individual lentivirus carrying the precursor of miR-21, miR-24, and miR-221, a cocktail of these 3 microRNA precursors, or green fluorescent protein as a control. After challenge in serum free medium, CPCs treated with the 3 microRNA cocktail showed significantly higher viability compared with untreated CPCs. After intramuscular and intramyocardial injections, in vivo bioluminescence imaging showed that microRNA cocktail-treated CPCs survived significantly longer after transplantation. After left anterior descending artery ligation, microRNA cocktail-treated CPCs boost the therapeutic efficacy in terms of functional recovery. Histological analysis confirmed increased myocardial wall thickness and CPC engraftment in the myocardium with the microRNA cocktail. Finally, we used bioinformatics analysis and experimental validation assays to show that Bim, a critical apoptotic activator, is an important target gene of the microRNA cocktail, which collectively can bind to the 3'UTR region of Bim and suppress its expression. CONCLUSIONS: We have demonstrated that a microRNA prosurvival cocktail (miR-21, miR-24, and miR-221) can improve the engraftment of transplanted cardiac progenitor cells and therapeutic efficacy for treatment of ischemic heart disease.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Sobrevivência de Enxerto/fisiologia , MicroRNAs/genética , MicroRNAs/uso terapêutico , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Injeções Intramusculares , Lentivirus , Camundongos , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Células-Tronco/fisiologia , Transdução Genética , Resultado do Tratamento
15.
Circulation ; 124(11 Suppl): S46-54, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21911818

RESUMO

BACKGROUND: Under normoxic conditions, hypoxia-inducible factor (HIF)-1α is rapidly degraded by 2 hydroxylases: prolyl hydroxylase (PHD) and factor-inhibiting HIF-1 (FIH). Because HIF-1α mediates the cardioprotective response to ischemic injury, its upregulation may be an effective therapeutic option for ischemic heart failure. METHODS AND RESULTS: PHD and FIH were cloned from mouse embryonic stem cells. The best candidate short hairpin (sh) sequences for inhibiting PHD isoenzyme 2 and FIH were inserted into novel, nonviral, minicircle vectors. In vitro studies after cell transfection of mouse C2C12 myoblasts, HL-1 atrial myocytes, and c-kit(+) cardiac progenitor cells demonstrated higher expression of angiogenesis factors in the double-knockdown group compared with the single-knockdown and short hairpin scramble control groups. To confirm in vitro data, shRNA minicircle vectors were injected intramyocardially after left anterior descending coronary artery ligation in adult FVB mice (n=60). Functional studies using MRI, echocardiography, and pressure-volume loops showed greater improvement in cardiac function in the double-knockdown group. To assess mechanisms of this functional recovery, we performed a cell trafficking experiment, which demonstrated significantly greater recruitment of bone marrow cells to the ischemic myocardium in the double-knockdown group. Fluorescence-activated cell sorting showed significantly higher activation of endogenous c-kit(+) cardiac progenitor cells. Immunostaining showed increased neovascularization and decreased apoptosis in areas of injured myocardium. Finally, western blots and laser-capture microdissection analysis confirmed upregulation of HIF-1α protein and angiogenesis genes, respectively. CONCLUSIONS: We demonstrated that HIF-1α upregulation by double knockdown of PHD and FIH synergistically increases stem cell mobilization and myocardial angiogenesis, leading to improved cardiac function.


Assuntos
Células-Tronco Embrionárias/transplante , Técnicas de Silenciamento de Genes , Terapia Genética/métodos , Oxigenases de Função Mista/genética , Infarto do Miocárdio/terapia , Neovascularização Fisiológica/fisiologia , Pró-Colágeno-Prolina Dioxigenase/genética , Animais , Apoptose/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos , Oxigenases de Função Mista/metabolismo , Modelos Animais , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Transplante de Células-Tronco , Resultado do Tratamento
16.
Transpl Immunol ; 23(4): 185-93, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20619345

RESUMO

Proper actin cytoskeleton architecture and dynamics are indispensable for events in the immunological response such as T cell migration, redistribution of T cell receptors, and interaction with antigen presenting cells. Thus, T cell activation, downstream signaling events and effector functions are all actin-dependent. Actin cytoskeleton architecture and dynamics are regulated by proteins belonging to the superfamily of small GTP-binding proteins, such as RhoA GTPase. We previously showed that the administration of an MHC class I allochimeric molecule [alpha1h1/u]-RT1.Aa, which contains donor-type (Wistar Furth, WF; RT1u) immunogenic epitopes displayed on recipient-type (ACI, RT1a) sequences, to the ACI recipient of heterotopic WF heart resulted in the restriction of the TCR repertoire, inhibition of T cell infiltration into the heterotopic cardiac allografts, abrogation of acute and chronic rejection, and induction of indefinite survival of the allograft. Here we show that the allochimeric molecule treatment caused downregulation of RhoA GTPase in T cells. This resulted in dramatic changes in the distribution of actin and the actin-binding protein, Hip55, in these cells, which in turn, inhibited T cell infiltration into the graft. This indicates that the immunosuppressive activity of the allochimeric molecule is achieved via downregulation of the RhoA pathway and disruption of the proper organization of T cell actin cytoskeleton to inhibit T cell functions such as motility and/or TCR signaling events.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Rejeição de Enxerto/tratamento farmacológico , Transplante de Coração , Proteínas dos Microfilamentos/metabolismo , Linfócitos T/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Regulação para Baixo , Rejeição de Enxerto/imunologia , Antígenos de Histocompatibilidade Classe I/administração & dosagem , Proteínas dos Microfilamentos/genética , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/administração & dosagem , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Proteína rhoA de Ligação ao GTP/genética , Domínios de Homologia de src/genética
17.
Genesis ; 48(1): 8-19, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19882666

RESUMO

The MHC class I allochimeric protein containing donor-type epitopes on recipient-type heavy chains induces indefinite survival of heterotopic cardiac allografts in rats. We analyzed gene expression profile of heart allograft tissue. Mutated peptide [alpha1h1/u]-RT1.Aa that contains donor-type (Wistar Furth, WF; RT1u) immunogenic epitopes displayed on recipient-type (ACI, RT1a) was delivered into ACI recipients of WF hearts at the time of transplantation in addition to a 3 days course of oral cyclosporine. Microarray analysis was performed using Affymetrix Rat 230 2.0 Microarray. Allochimeric molecule treatment caused upregulation of genes involved in structural integrity of heart muscle, downregulation of IL-1beta a key modulator of the immune response, and downregulation of partitioning defective six homolog gamma PAR6, which is involved in T cell polarity, motility, and ability to scan dendritic cells (DC). These indicate that the immunosuppressive function of allochimeric molecule and/or the establishment of allograft tolerance depend on the induction of genes responsible for the heart tissue integrity, the suppression of cytokine pathway(s), and possibly the impairment of T cells mobility and their DC scanning ability. These novel findings may have important clinical implications for inhibition of chronic rejection in transplant recipients.


Assuntos
Perfilação da Expressão Gênica , Transplante de Coração , Antígenos de Histocompatibilidade/imunologia , Tolerância ao Transplante/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Ciclosporina/farmacologia , Expressão Gênica/efeitos dos fármacos , Imunossupressores/farmacologia , Interleucina-1beta/genética , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Endogâmicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tolerância ao Transplante/imunologia
18.
PLoS One ; 4(12): e8020, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19956540

RESUMO

BACKGROUND: The allochimeric MHC class I molecule [alpha1h1/u]-RT1.Aa that contains donor-type (Wistar Furth, WF; RT1u) epitopes displayed on recipient-type (ACI, RT1a) administered in conjunction with sub-therapeutic dose of cyclosporine (CsA) induces indefinite survival of heterotopic cardiac allografts in rat model. In vascularized transplantation models, the spleen contributes to graft rejection by generating alloantigen reactive T cells. The immune response in allograft rejection involves a cascade of molecular events leading to the formation of immunological synapses between T cells and the antigen-presenting cells. METHODOLOGY/PRINCIPAL FINDINGS: To elucidate the molecular pathways involved in the immunosuppressive function of allochimeric molecule we performed microarray and quantitative RTPCR analyses of gene expression profile of splenic T cells from untreated, CsA treated, and allochimeric molecule + subtherapeutic dose of CsA treated animals at day 1, 3 and 7 of post transplantation. Allochimeric molecule treatment caused down regulation of genes involved in actin filament polymerization (RhoA and Rac1), cell adhesion (Catna1, Vcam and CD9), vacuolar transport (RhoB, Cln8 and ATP6v1b2), and MAPK pathway (Spred1 and Dusp6) involved in tubulin cytoskeleton reorganization and interaction between actin and microtubule cytoskeleton. All these genes are involved in T cell polarity and motility, i.e., their ability to move, scan and to form functional immunological synapse with antigen presenting cells (APCs). CONCLUSIONS: These results indicate that the immunosuppressive function of allochimeric molecule may depend on the impairment of T cells' movement and scanning ability, and possibly also the formation of immunological synapse. We believe that these novel findings may have important clinical implications for organ transplantation.


Assuntos
Movimento Celular/genética , Polaridade Celular/genética , Regulação para Baixo/genética , Transplante de Coração/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Linfócitos T/citologia , Tolerância ao Transplante/genética , Animais , Movimento Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Perfilação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/imunologia , Peptídeos/farmacologia , Análise de Componente Principal , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Fatores de Tempo , Tolerância ao Transplante/efeitos dos fármacos
19.
J Huazhong Univ Sci Technolog Med Sci ; 27(5): 564-70, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18060637

RESUMO

In order to provide us new clues to induce some endogenous protective molecular mechanisms, the changes in gene expression profile induced by ischemia-reperfusion in pulmonary tissues of rats were investigated and the dynamic mechanism of pulmonary ischemia-reperfusion injury was elucidated. Thirty male Wistar rats were randomly divided into 6 groups: 5 ischemia-reperfusion (I/R) groups (I/R 0-h, I/R 1-h, I/R 3-h, I/R 6-h, I/R 24-h) and control group (n=5 in each). An in situ ischemia-reperfusion lung injury rat model was established by occluded hilus of lung. The RatRef-12 Expression Beadchip (22 226 gene probes per array) was used to analyze the pattern of gene expression in all groups. The results showed that 648, 340, 711, 1279 and 641 genes were differentially expressed in I/R 0-, 1-, 3-, 6-and 24-h groups respectively. The differentially expressed genes were classified as following 7 functional categories: cytokine, adhesion molecule, growth factor and apoptosis-related factor, oxidation and antioxidation molecule, metabolic enzyme, ion channel and aquaporin, signal transduction molecule. It was suggested that gene chip technology was an effective and quick method for screening differentially expressed genes. Many differentially expressed genes with different functions interacted each other to result in pulmonary ischemia-reperfusion injury.


Assuntos
Perfilação da Expressão Gênica , Pulmão/irrigação sanguínea , Traumatismo por Reperfusão/genética , Animais , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar
20.
J Huazhong Univ Sci Technolog Med Sci ; 26(1): 99-100, 136, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16711018

RESUMO

In order to establish more simple and effective rat orthotopic lung transplantation models, 20 rats were divided into donor and recipient groups. Rat lung transplantation models were established by using improved cuff technique. All the 10 operations were accomplished successfully. The mean operative time of recipients was 45 +/- 4 min. The survival time was over 30 days after lung transplantation. The checks of X-ray were almost normal. There was no significant difference in the blood gas analysis before and after clipping the right hilum (P > .05). This method is more simple, applicable and requires less time.


Assuntos
Transplante de Pulmão , Modelos Animais , Animais , Feminino , Transplante de Pulmão/métodos , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...