Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Transgenic Res ; 24(1): 31-41, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25048992

RESUMO

Lentiviral technology has been recently proposed to generate transgenic farm animals more efficiently and easier than traditional techniques. The objective was to evaluate several parameters of lambs obtained by lentiviral transgenesis in comparison with non-transgenic counterparts. In vitro produced embryos were microinjected (TG group) at two-cell stage with a lentiviral construct containing enhanced green fluorescent protein (eGFP) gene, while embryos produced by in vitro fertilization (IVF group) or intrauterine insemination (IUI group) were not microinjected. Microinjection technique efficiently generated eight-cell transgenic embryos (97.4%; 114/117). Development rate on day 5 after fertilization was similar for TG (39.3%, 46/117) and IVF embryos (39.6%, 44/111). Pregnancy rate was detected in 50.0% (6/12) of recipient ewes with TG embryos, in 46.7% (7/15) with IVF embryos, and in 65.0% (13/20) of IUI ewes (P = NS). Nine lambs were born in TG group, six lambs in IVF group, and 16 lambs in IUI group. All TG lambs (9/9) were GFP positive to real-time PCR and eight (88.9%) showed a strong and evident GFP expression in mucosae, eyes and keratin tissues. Fetal growth monitored every 15 day by ultrasonography did not show significant differences. Transgenic lambs neither differ in morphometric variables in comparison with non transgenic IVF lambs within 3 months after birth. Transmission of the transgene to the progeny was observed in green fluorescent embryos produced by IVF using semen from the TG founder lambs. In conclusion, this study demonstrates the high efficiency of lentiviral technology to produce transgenic sheep, with no clinic differences in comparison with non transgenic lambs.


Assuntos
Desenvolvimento Embrionário/genética , Desenvolvimento Fetal/genética , Proteínas de Fluorescência Verde/genética , Lentivirus/genética , Animais , Animais Geneticamente Modificados/genética , Feminino , Fertilização in vitro , Vetores Genéticos , Gravidez , Ovinos
2.
Am J Transplant ; 14(5): 1109-19, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24612827

RESUMO

Xenogenic fetal neuroblasts are considered as a potential source of transplantable cells for the treatment of neurodegenerative diseases, but immunological barriers limit their use in the clinic. While considerable work has been performed to decipher the role of the cellular immune response in the rejection of intracerebral xenotransplants, there is much still to learn about the humoral reaction. To this end, the IgG response to the transplantation of fetal porcine neural cells (PNC) into the rat brain was analyzed. Rat sera did not contain preformed antibodies against PNC, but elicited anti-porcine IgG was clearly detected in the host blood once the graft was rejected. Only the IgG1 and IgG2a subclasses were up-regulated, suggesting a T-helper 2 immune response. The main target of these elicited IgG antibodies was porcine neurons, as determined by double labeling in vitro and in vivo. Complement and anti-porcine IgG were present in the rejecting grafts, suggesting an active role of the host humoral response in graft rejection. This hypothesis was confirmed by the prolonged survival of fetal porcine neurons in the striatum of immunoglobulin-deficient rats. These data suggest that the prolonged survival of intracerebral xenotransplants relies on the control of both cell-mediated and humoral immune responses.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Córtex Cerebral/imunologia , Rejeição de Enxerto/imunologia , Imunoglobulina G/imunologia , Neurônios/imunologia , Transplante Heterólogo , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/cirurgia , Citometria de Fluxo , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto , Técnicas Imunoenzimáticas , Neurônios/citologia , Neurônios/transplante , Ratos , Ratos Endogâmicos Lew , Suínos
3.
Am J Transplant ; 14(5): 1021-1031, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24731243

RESUMO

The administration of autologous (recipient-derived) tolerogenic dendritic cells (ATDCs) is under clinical evaluation. However, the molecular mechanisms by which these cells prolong graft survival in a donor-specific manner is unknown. Here, we tested mouse ATDCs for their therapeutic potential in a skin transplantation model. ATDC injection in combination with anti-CD3 treatment induced the accumulation of CD8(+) CD11c(+) T cells and significantly prolonged allograft survival. TMEM176B is an intracellular protein expressed in ATDCs and initially identified in allograft tolerance. We show that Tmem176b(-/-) ATDCs completely failed to trigger both phenomena but recovered their effect when loaded with donor peptides before injection. These results strongly suggested that ATDCs require TMEM176B to cross-present antigens in a tolerogenic fashion. In agreement with this, Tmem176b(-/-) ATDCs specifically failed to cross-present male antigens or ovalbumin to CD8(+) T cells. Finally, we observed that a Tmem176b-dependent cation current controls phagosomal pH, a critical parameter in cross-presentation. Thus, ATDCs require TMEM176B to cross-present donor antigens to induce donor-specific CD8(+) CD11c(+) T cells with regulatory properties and prolong graft survival.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Apresentação de Antígeno/imunologia , Complexo CD3/imunologia , Células Dendríticas/imunologia , Sobrevivência de Enxerto/fisiologia , Proteínas de Membrana/fisiologia , Transplante de Pele , Aloenxertos , Animais , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada , Eletrofisiologia , Endocitose/fisiologia , Feminino , Citometria de Fluxo , Tolerância Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/fisiologia
4.
Am J Transplant ; 11(10): 2036-45, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21794083

RESUMO

Innovative therapeutic strategies are needed to diminish the impact of harmful immunosuppression in transplantation. Dendritic cell (DC)-based therapy is a promising approach for induction of antigen-specific tolerance. Using a heart allograft model in rats, we analyzed the immunoregulatory mechanisms by which injection of autologous tolerogenic DCs (ATDCs) plus suboptimal immunosuppression promotes indefinite graft survival. Surprisingly, we determined that Interferon-gamma (IFNG), a cytokine expected to be propathogenic, was threefold increased in the spleen of tolerant rats. Importantly, its blockade led to allograft rejection [Mean Survival Time (MST) = 25.6 ± 4 days], showing that IFNG plays a critical role in immunoregulatory mechanisms triggered by ATDCs. IFNG was expressed by TCRαß(+) CD3(+) CD4(-) CD8(-) NKRP1(-) cells (double negative T cells, DNT), which accumulated in the spleen of tolerant rats. Interestingly, ATDCs specifically induced IFNG production by DNT cells. ATDCs expressed the cytokinic chain Epstein-Barr virus-induced gene 3 (EBI3), an IL-12 family member. EBI3 blockade or knock-down through siRNA completely abolished IFNG expression in DNT cells. Finally, EBI3 blockade in vivo led to allograft rejection (MST = 36.8 ± 19.7 days), demonstrating for the first time a role for EBI3 in transplantation tolerance. Taken together our results have important implications in the rationalization of DC-based therapy in transplantation as well as in the patient immunomonitoring follow-up.


Assuntos
Transplante de Células , Células Dendríticas/citologia , Herpesvirus Humano 4/metabolismo , Interferon gama/metabolismo , Proteínas Virais/metabolismo , Animais , Teste de Cultura Mista de Linfócitos , Microscopia Confocal , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Transplante Homólogo
5.
J Exp Med ; 167(2): 452-72, 1988 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-2831292

RESUMO

We report evidence that FcR(CD16) on human NK cells are signal-transducing molecules that, upon ligand binding, induce transcription of genes encoding surface activation molecules [IL-2-R(CD25)] and cytokines (IFN-gamma and TNF) relevant to NK cell biology and functions. Homogeneous NK and T cell populations purified from short-term bulk cultures of PBMC with irradiated B lymphoblastoid cell lines were cultured in the presence of FcR ligands (particulate immune complexes or immobilized anti-CD16 antibodies) alone or with rIL-2. Upon 18 h of stimulation, NK cells express Tac, TfR, and 4F2 antigens and produce IFN-gamma and TNF; both effects are synergistically enhanced in the presence of rIL-2, which is itself ineffective. Treatment of NK cells with FcR(CD16) ligands induces accumulation of mRNA for IFN-gamma and TNF and, to a lesser extent, IL-2-R with fast kinetics also in the absence of de novo protein synthesis. rIL-2 and FcR(CD16) ligands synergize to induce mRNA accumulation. mRNA accumulation and transcription of TNF and IFN-gamma genes induced by FcR(CD16) ligands are greater than those induced by rIL-2, and the reverse is true for IL-2-R. The two stimuli do not synergize at the transcriptional level. These observations indicate that the mechanisms through which FcR(CD16) ligands and rIL-2 induce NK cell activation are, in part, distinct. Both operate at the transcriptional level, although other mechanisms are probably induced by the FcR ligand stimulus per se or in combination with other lymphokines and synergize at a post-transcriptional or translational level to enhance NK cell activation.


Assuntos
Células Matadoras Naturais/metabolismo , Linfocinas/genética , Receptores Fc/fisiologia , Receptores Imunológicos/genética , Transcrição Gênica , Animais , Antígenos de Superfície/biossíntese , Produtos Biológicos/biossíntese , Produtos Biológicos/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Reagentes de Ligações Cruzadas , Citocinas , Sinergismo Farmacológico , Humanos , Interleucina-2/farmacologia , Células Matadoras Naturais/imunologia , Cinética , Camundongos , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Receptores Imunológicos/metabolismo , Receptores de Interleucina-2 , Proteínas Recombinantes/farmacologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral
6.
J Exp Med ; 169(2): 549-67, 1989 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-2536067

RESUMO

In this study, we present evidence that interaction of Fc gamma R(CD16) with ligands (immune complexes or anti-CD16 antibodies) induces a rapid rise in [Ca2+]i and fast production of both inositol 1,4,5 triphosphate (IP3) and IP4 in homogeneous NK cell preparations. Part of the initial [Ca2+]i rise observed upon stimulation of NK cells with either anti-CD16 antibodies alone or after their crosslinking at the cell membrane depends on Ca2+ mobilization from intracellular stores, but sustained [Ca2+]i levels are maintained, after the initial spike, through influx of extracellular Ca2+. The [Ca2+]i rise is mediated, at least in part, by increases in IP3 after receptor-induced hydrolysis of membrane polyphosphoinositides (PPI). The role of extracellular Ca2+ in Fc gamma R(CD16)-dependent induction of lymphokine gene expression has been tested by evaluating production, mRNA accumulation and transcription of IFN-gamma and TNF in NK cells stimulated with Fc gamma R(CD16) ligands and/or rIL-2 in the presence of EGTA. Under these conditions, accumulation and transcription of both IFN-gamma and TNF mRNA induced by CD16 ligands, but not that induced by rIL-2, is completely abolished and neither cytokine can be detected at significant levels in the supernatant fluids of cells so treated. These data confirm that NK cell activation by specific ligands occurs through mechanisms distinct from those induced by IL-2, and indicate that extracellular Ca2+ represents a stringent requirement for cytokine production induced in NK cells through specific (Fc gamma R) stimulation. Our data also indicate that the [Ca2+]i rise induced upon Fc gamma R(CD16) crosslinking, though necessary, is not sufficient per se to induce activation of lymphokine genes, compatible with the hypothesis that Fc gamma R(CD16) crosslinking generates additional transducing signals that synergize with IL-2 to maximally activate NK cells.


Assuntos
Antígenos de Diferenciação/fisiologia , Cálcio/fisiologia , Células Matadoras Naturais/fisiologia , Ativação Linfocitária , Linfocinas/genética , Fosfatidilinositóis/fisiologia , Receptores Fc/fisiologia , Éteres/farmacologia , Regulação da Expressão Gênica , Humanos , Técnicas In Vitro , Interferon gama/genética , Ionomicina , Ligantes , RNA Mensageiro/genética , Receptores de IgG , Receptores de Interleucina-2/fisiologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética
7.
J Exp Med ; 169(2): 569-83, 1989 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-2521357

RESUMO

We have analyzed the ability of highly purified preparations of human NK cells to produce CSF. NK cells, purified by negative selection from 10-d cultures of PBMC incubated with irradiated B-lymphoblastoid cell lines, were stimulated with rIL-2, FcR(CD16) ligands (particulate immune complexes or anti-CD16 antibodies bound to Sepharose), a combination of CD16 ligands and rIL-2, or the phorbol diester phorbol dibutyrate (PDBu) together with the Ca2+ ionophore A23187. Both rIL-2 and CD16 ligands induce accumulation of GM-CSF mRNA in NK cells and the combined effect of the two stimuli is synergistic. Maximal accumulation of GM-CSF mRNA is observed after PDBu/A23187 stimulation. The participation of contaminant T cells in the observed expression of the GM-CSF gene is excluded because CD16 ligands do not stimulate T cells and CD3 ligands, powerful stimulators of T cells, are inactive on NK cells. Accumulation of CSF-1 mRNA is observed only in NK cells stimulated with both CD16 ligands and rIL-2, whereas accumulation of IL-3 mRNA is observed only in NK cells stimulated with PDBu/A23187. Transcripts of the G-CSF, IL-1 alpha, and IL-1 beta genes were never detected in NK cells in these experiments. The kinetics of accumulation of GM-CSF and CSF-1 mRNA in NK cells stimulated with CD16 ligands and rIL-2 peaked at 2-4 h and was slower than that of TNF and IFN-gamma mRNA, which peak at 1 h. GM-CSF was precipitated from the supernatant fluids of NK cells stimulated with PDBu/A23187 and its biological activity was demonstrated by the ability of the supernatants to sustain proliferation of the TALL-101 cell line or CML blasts. Biological activity of IL-3 and CSF-1 was demonstrable in supernatant fluids of NK cells stimulated with PDBu/A23187 and CD16 ligands/rIL-2, respectively.


Assuntos
Fatores Estimuladores de Colônias/biossíntese , Substâncias de Crescimento/biossíntese , Interleucina-3/biossíntese , Células Matadoras Naturais/fisiologia , Antígenos de Diferenciação/fisiologia , Antígenos de Diferenciação de Linfócitos T/fisiologia , Bioensaio , Northern Blotting , Complexo CD3 , Calcimicina/farmacologia , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Humanos , Interleucina-1/biossíntese , Interleucina-2/farmacologia , Ligantes , Ésteres de Forbol/farmacologia , Testes de Precipitina , RNA Mensageiro/genética , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores Fc/fisiologia , Receptores de IgG
8.
FASEB J ; 23(9): 3070-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19420134

RESUMO

Clinical translation of dendritic cell (DC)-based cell therapy requires preclinical studies in nonhuman primates (NHPs). The aim of this work was to establish the in vitro conditions for generation of NHP tolerogenic DCs (Tol-DCs), as well as to analyze the molecular mechanisms by which these cells could control an immune response. Two populations of NHP bone marrow-derived DCs (BMDCs) were obtained: adherent and nonadherent. Although both populations displayed a quite similar phenotype, they were very different functionally. We characterized the adherent BMDCs as Tol-DCs that were poor stimulators of T cells and actively inhibited T-cell proliferation, whereas the nonadherent population displayed immunogenic properties in vitro. Interestingly, the anti-inflammatory and immunosuppressive enzyme heme oxygenase-1 (HO-1) was up-regulated in Tol-DCs, compared to the immunogenic BMDCs. We demonstrated that HO-1 mediates the immunosuppressive properties of Tol-DCs in vitro (in NHPs and rats) and that HO-1 is involved in the in vivo tolerogenic effect of Tol-DCs in a rat model of allotransplantation. In conclusion, here we characterized the in vitro generation of NHP Tol-DCs. Furthermore, we showed for the first time that HO-1 plays a role in the active inhibition of T-cell responses by rat and NHP Tol-DCs.


Assuntos
Células Dendríticas/imunologia , Heme Oxigenase-1/genética , Tolerância Imunológica/imunologia , Linfócitos T/imunologia , Animais , Células da Medula Óssea , Adesão Celular , Transplante de Células , Células Cultivadas , Células Dendríticas/enzimologia , Células Dendríticas/transplante , Primatas , Ratos , Transplante Homólogo , Regulação para Cima/genética
9.
Transgenic Res ; 19(5): 745-63, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20094912

RESUMO

Adoptive cell transfer studies in regenerative research and identification of genetically modified cells after gene therapy in vivo require unequivocally identifying and tracking the donor cells in the host tissues, ideally over several days or for up to several months. The use of reporter genes allows identifying the transferred cells but unfortunately most are immunogenic to wild-type hosts and thus trigger rejection in few days. The availability of transgenic animals from the same strain that would express either high levels of the transgene to identify the cells or low levels but that would be tolerant to the transgene would allow performing long-term analysis of labelled cells. Herein, using lentiviral vectors we develop two new lines of GFP-expressing transgenic rats displaying different levels and patterns of GFP-expression. The "high-expresser" line (GFP(high)) displayed high expression in most tissues, including adult neurons and neural precursors, mesenchymal stem cells and in all leukocytes subtypes analysed, including myeloid and plasmacytoid dendritic cells, cells that have not or only poorly characterized in previous GFP-transgenic rats. These GFP(high)-transgenic rats could be useful for transplantation and immunological studies using GFP-positive cells/tissue. The "low-expresser" line expressed very low levels of GFP only in the liver and in less than 5% of lymphoid cells. We demonstrate these animals did not develop detectable humoral and cellular immune responses against both transferred GFP-positive splenocytes and lentivirus-mediated GFP gene transfer. Thus, these GFP-transgenic rats represent useful tools for regenerative medicine and gene therapy.


Assuntos
Genes Reporter , Terapia Genética , Proteínas de Fluorescência Verde/genética , Ratos Transgênicos/genética , Medicina Regenerativa , Transferência Adotiva , Animais , Diferenciação Celular , Regulação da Expressão Gênica , Genes Sintéticos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/biossíntese , Lentivirus/genética , Leucócitos/metabolismo , Fígado/metabolismo , Linfócitos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neurônios/metabolismo , Especificidade de Órgãos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/biossíntese
10.
Sci Rep ; 10(1): 5995, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32265471

RESUMO

Different mutations of the OTOF gene, encoding for otoferlin protein expressed in the cochlear inner hair cells, induces a form of deafness that is the major cause of nonsyndromic recessive auditory neuropathy spectrum disorder in humans. We report the generation of the first large animal model of OTOF mutations using the CRISPR system associated with different Cas9 components (mRNA or protein) assisted by single strand oligodeoxynucleotides (ssODN) to induce homology-directed repair (HDR). Zygote microinjection was performed with two sgRNA targeting exon 5 and 6 associated to Cas9 mRNA or protein (RNP) at different concentrations in a mix with an ssODN template targeting HDR in exon 5 containing two STOP sequences. A total of 73 lambs were born, 13 showing indel mutations (17.8%), 8 of which (61.5%) had knock-in mutations by HDR. Higher concentrations of Cas9-RNP induced targeted mutations more effectively, but negatively affected embryo survival and pregnancy rate. This study reports by the first time the generation of OTOF disrupted sheep, which may allow better understanding and development of new therapies for human deafness related to genetic disorders. These results support the use of CRISPR/Cas system assisted by ssODN as an effective tool for gene editing in livestock.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Proteínas de Membrana/genética , Oligodesoxirribonucleotídeos/genética , Ovinos/genética , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Feminino , Masculino , Microinjeções , Mutação , Reparo de DNA por Recombinação , Ovinos/embriologia
11.
Gene Ther ; 15(18): 1247-56, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18762806

RESUMO

In this review, we summarize the work published over the last 2 years using genetic modifications of animals in the field of xenotransplantation. Genetic engineering of the donor has become a powerful tool in xenotransplantation, both for the inactivation of one particular porcine gene and for the addition of human genes with the goal of overcoming xenogeneic barriers. We summarize the work relative to the knockout of the alpha1,3-galactosyltransferase gene, followed by genetic engineering aimed at reducing the humoral and cellular immune response, complement activation and coagulation. Finally, we report on the genetic modification of pigs to reduce porcine endogenous retrovirus infection risk in the xenogeneic context.


Assuntos
Engenharia Genética , Imunologia de Transplantes , Transplante Heterólogo/métodos , Animais , Animais Geneticamente Modificados , Rejeição de Enxerto/imunologia , Humanos , Suínos , Transplante Heterólogo/imunologia
12.
Nat Commun ; 9(1): 1133, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29556040

RESUMO

In genome editing with CRISPR-Cas9, transgene integration often remains challenging. Here, we present an approach for increasing the efficiency of transgene integration by homology-dependent repair (HDR). CtIP, a key protein in early steps of homologous recombination, is fused to Cas9 and stimulates transgene integration by HDR at the human AAVS1 safe harbor locus. A minimal N-terminal fragment of CtIP, designated HE for HDR enhancer, is sufficient to stimulate HDR and this depends on CDK phosphorylation sites and the multimerization domain essential for CtIP activity in homologous recombination. HDR stimulation by Cas9-HE, however, depends on the guide RNA used, a limitation that may be overcome by testing multiple guides to the locus of interest. The Cas9-HE fusion is simple to use and allows obtaining twofold or more efficient transgene integration than that with Cas9 in several experimental systems, including human cell lines, iPS cells, and rat zygotes.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Nucleares/metabolismo , Reparo de DNA por Recombinação , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Endodesoxirribonucleases , Elementos Facilitadores Genéticos , Feminino , Células HCT116 , Células HEK293 , Recombinação Homóloga , Humanos , Mutação INDEL , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Oócitos/metabolismo , Fosforilação , Multimerização Proteica , RNA Guia de Cinetoplastídeos/genética , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transgenes , Integração Viral/genética , Zigoto/metabolismo
13.
J Clin Invest ; 102(11): 1920-6, 1998 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9835616

RESUMO

Donor-specific (DST) or nonspecific blood transfusions administered before transplantation can enhance survival of vascularized allografts both in humans and animals but the immunological mechanisms of this effect remain unclear. We have analyzed the expression and the role of endogenous TGF-beta1 in a model of heart allograft tolerance, induced by pregraft DST in adult rats. We reported previously that this tolerance occurs despite a strong infiltration of leukocytes into the graft that are unable to produce both Th1- and Th2-related cytokines in vivo. Allografts from DST-treated rats express high levels of TGF-beta1 mRNA and active protein. This phenomenon is correlated with the rapid infiltration of leukocytes producing high amounts of TGF-beta1. TGF-beta1-producing cells are virtually absent among early infiltrating cells in rejected grafts but are found at a later time point. The induction of allograft tolerance in vivo is abrogated by administration of neutralizing anti-TGF-beta mAb. Moreover, overexpression of active TGF- beta1 in heart allografts using a recombinant adenovirus leads to prolonged graft survival in unmodified recipients. Taken together, our results identify TGF-beta as a critical cytokine involved in the suppression of allograft rejection induced by DST and suggest that TGF-beta-producing regulatory cells are also involved in allograft tolerance.


Assuntos
Transfusão de Sangue , Facilitação Imunológica de Enxerto , Sobrevivência de Enxerto/fisiologia , Transplante de Coração , Fator de Crescimento Transformador beta/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Rejeição de Enxerto/imunologia , Leucócitos/metabolismo , Teste de Cultura Mista de Linfócitos , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos BUF , Ratos Endogâmicos Lew , Baço/imunologia , Doadores de Tecidos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/imunologia , Transplante Homólogo
14.
Theriogenology ; 86(1): 160-9, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27155732

RESUMO

Genetically engineered sheep and goats represent useful models applied to proof of concepts, large-scale production of novel products or processes, and improvement of animal traits, which is of interest in biomedicine, biopharma, and livestock. This disruptive biotechnology arose in the 80s by injecting DNA fragments into the pronucleus of zygote-staged embryos. Pronuclear microinjection set the transgenic concept into people's mind but was characterized by inefficient and often frustrating results mostly because of uncontrolled and/or random integration and unpredictable transgene expression. Somatic cell nuclear transfer launched the second wave in the late 90s, solving several weaknesses of the previous technique by making feasible the transfer of a genetically modified and fully characterized cell into an enucleated oocyte, capable of cell reprogramming to generate genetically engineered animals. Important advances were also achieved during the 2000s with the arrival of new techniques like the lentivirus system, transposons, RNA interference, site-specific recombinases, and sperm-mediated transgenesis. We are now living the irruption of the third technological wave in which genome edition is possible by using endonucleases, particularly the CRISPR/Cas system. Sheep and goats were recently produced by CRISPR/Cas9, and for sure, cattle will be reported soon. We will see new genetically engineered farm animals produced by homologous recombination, multiple gene editing in one-step generation and conditional modifications, among other advancements. In the following decade, genome edition will continue expanding our technical possibilities, which will contribute to the advancement of science, the development of clinical or commercial applications, and the improvement of people's life quality around the world.


Assuntos
Engenharia Genética/veterinária , Cabras/genética , Ovinos/genética , Animais , Animais Geneticamente Modificados , Engenharia Genética/métodos , Técnicas de Reprodução Assistida/veterinária
15.
Mucosal Immunol ; 9(2): 539-49, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26329427

RESUMO

Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel diseases (IBDs), are characterized by high levels of IL-22 production. Rodent studies revealed that this cytokine is protective during colitis but whether this is true in IBDs is unclear. We show here that levels of the soluble inhibitor of IL-22, interleukin 22-binding protein (IL-22BP), are significantly enhanced during IBDs owing to increased numbers of IL-22BP-producing eosinophils, that we unexpectedly identify as the most abundant source of IL-22BP protein in human gut. In addition, using IL-22BP-deficient rats, we confirm that endogenous IL-22BP is effective at blocking protective actions of IL-22 during acute colitis. In conclusion, our study provides new important insights regarding the biology of IL-22 and IL-22BP in the gut and indicates that protective actions of IL-22 are likely to be suboptimal in IBDs thus making IL-22BP a new relevant therapeutic target.


Assuntos
Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Eosinófilos/imunologia , Interleucinas/imunologia , Receptores de Interleucina/imunologia , Adulto , Animais , Estudos de Casos e Controles , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Colite/patologia , Colite Ulcerativa/genética , Colite Ulcerativa/patologia , Colo/imunologia , Colo/patologia , Doença de Crohn/genética , Doença de Crohn/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Eosinófilos/metabolismo , Eosinófilos/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Interleucinas/genética , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores de Interleucina/deficiência , Receptores de Interleucina/genética , Transdução de Sinais , Interleucina 22
16.
J Leukoc Biol ; 53(4): 390-8, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8482919

RESUMO

We analyzed the phenotypic changes associated with monocyte activation and differentiation using a newly developed monoclonal antibody (B148.4). Among peripheral blood leukocytes, the antigen recognized by this antibody is expressed on monocytes and granulocytes at high and low density, respectively. Antigen expression is lost during in vitro differentiation of monocytes and is absent on tissue macrophages, indicating that expression of this antigen is related to monocyte differentiation. Only 1 alpha,25-dihydroxyvitamin D3 and phorbol diesters, of several inducers tested, up-regulate B148.4 antigen expression on cells (monocytes and certain myeloid cell lines) that constitutively bear the antigen, without, however, allowing its maintenance during monocytic differentiation or inducing it on negative cells. By immunoprecipitation from B148.4+ U937 cells, the antigen is a complex of a major 116-kd and two minor 38- and 46-kd molecules. Analysis of eight different tissues reveals that the antigen is shared with endothelial cells. Biochemical characteristics, cellular distribution, and expression pattern on monocytes, myeloid cell lines, and AML cells upon culture with different stimuli indicate that B148.4 is a novel monocyte differentiation antigen.


Assuntos
Antígenos de Diferenciação/análise , Monócitos/fisiologia , Anticorpos Monoclonais , Antígenos de Diferenciação/imunologia , Calcitriol/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Linfócitos/citologia , Linfócitos/fisiologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/fisiologia , Células Tumorais Cultivadas
17.
PLoS One ; 10(8): e0136690, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26305800

RESUMO

While CRISPR/Cas9 technology has proven to be a valuable system to generate gene-targeted modified animals in several species, this tool has been scarcely reported in farm animals. Myostatin is encoded by MSTN gene involved in the inhibition of muscle differentiation and growth. We determined the efficiency of the CRISPR/Cas9 system to edit MSTN in sheep and generate knock-out (KO) animals with the aim to promote muscle development and body growth. We generated CRISPR/Cas9 mRNAs specific for ovine MSTN and microinjected them into the cytoplasm of ovine zygotes. When embryo development of CRISPR/Cas9 microinjected zygotes (n = 216) was compared with buffer injected embryos (n = 183) and non microinjected embryos (n = 173), cleavage rate was lower for both microinjected groups (P<0.05) and neither was affected by CRISPR/Cas9 content in the injected medium. Embryo development to blastocyst was not affected by microinjection and was similar among the experimental groups. From 20 embryos analyzed by Sanger sequencing, ten were mutant (heterozygous or mosaic; 50% efficiency). To obtain live MSTN KO lambs, 53 blastocysts produced after zygote CRISPR/Cas9 microinjection were transferred to 29 recipient females resulting in 65.5% (19/29) of pregnant ewes and 41.5% (22/53) of newborns. From 22 born lambs analyzed by T7EI and Sanger sequencing, ten showed indel mutations at MSTN gene. Eight showed mutations in both alleles and five of them were homozygous for indels generating out-of frame mutations that resulted in premature stop codons. Western blot analysis of homozygous KO founders confirmed the absence of myostatin, showing heavier body weight than wild type counterparts. In conclusion, our results demonstrate that CRISPR/Cas9 system was a very efficient tool to generate gene KO sheep. This technology is quick and easy to perform and less expensive than previous techniques, and can be applied to obtain genetically modified animal models of interest for biomedicine and livestock.


Assuntos
Animais Geneticamente Modificados , Sistemas CRISPR-Cas , Miostatina/genética , Animais , Feminino , Técnicas de Inativação de Genes , Microinjeções , Gravidez , Carneiro Doméstico/genética , Zigoto
18.
Hum Gene Ther ; 8(13): 1625-34, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9322095

RESUMO

The in vitro purification of pancreatic islets offers an opportunity for their modification by ex vivo gene transfer. We investigated the efficiency and functional consequences of adenovirus-mediated gene transfer into adult murine pancreatic islets with a recombinant adenovirus encoding for the beta-galactosidase (beta-Gal) reporter gene. At 10(6) pfu/islet, almost all of the islets were transduced, but maximal transduction was obtained at 10(7) pfu/islet. Histochemical analysis of frozen islet sections showed that transduced cells were only located at the periphery of islets. Transduced islets showed normal insulin secretion in vitro, and were able to normalize in vivo the glycemia of streptozotocin-induced diabetes in syngeneic and allogeneic mice. beta-Gal expression in transduced islets was observed for at least 6 weeks in naive normal recipients and in immunodeficient mice, but was shortened in mice preimmunized to adenovirus. Nevertheless, islets maintained normal control of glycemia in all mice. An early leukocyte infiltrate was observed in syngeneic grafts of transduced islets, but no acceleration in rejection of fully MHC-incompatible islet grafts occurred. In summary, adenovirus-mediated gene transfer in adult mouse islets, although sparing most of the beta-cells, was highly efficient and did not impair insulin secretion by islets. The immune response to the adenovirus and/or to the transgene might be only partially responsible for the decreased expression over time of the transduced gene. Accordingly, adenovirus-mediated gene transfer might allow efficient expression of vectorized sequences with potential immunosuppressive effects in the islet microenvironment.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Ilhotas Pancreáticas/metabolismo , Adenoviridae/imunologia , Animais , Feminino , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/imunologia , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Transdução Genética
19.
Hum Gene Ther ; 9(12): 1755-68, 1998 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-9721086

RESUMO

Recombinant adenoviruses can be used for in vivo gene transfer with great efficiency. However, the duration of transgene expression and the possibility of readministering the virus are severely limited by the host anti-adenovirus immune response, which is controlled mainly by cytokine networks. Adenoviruses encoding IL-4 (AdIL-4) or IL-10 (AdIL-10) were administered to rats through the portal vein and the anti-adenovirus immune response was studied. As compared with administering adenoviruses without transgene (Addl324) or with the lacZ gene (AdlacZ), AdIL-4, but not AdIL-10, resulted in a significant increase in leukocytes in the liver, with a predominance of macrophages that peaked on days 7 and 14 after gene transfer and gradually returned to normal by day 28. AdIL-4 induced a significant increase in both neutralizing and ELISA-detected anti-adenovirus antibodies, whereas AdIL-10 caused an increase in ELISA-detected antibodies alone. Anti-adenovirus antibodies were predominantly of Th1-dependent immunoglobulin subclasses in rats receiving Addl324, AdlacZ, or AdIL-10, whereas animals receiving AdIL-4 showed a predominance of Th2-dependent immunoglobulin subclasses. Type 1 (IFN-gamma) and type 2 (IL-5) cytokines were increased only in livers from rats receiving AdIL-4. Rats receiving AdIL-4 showed increased anti-adenovirus cytotoxic T lymphocyte activity and CD8+ cell depletion prevented leukocyte infiltration in the liver. These results show that IL-4 increases local and systemic immune responses against recombinant adenoviruses.


Assuntos
Adenoviridae/imunologia , Interleucina-10/genética , Interleucina-4/genética , Adenoviridae/genética , Animais , Anticorpos Antivirais/biossíntese , Sequência de Bases , Primers do DNA , Imuno-Histoquímica , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Óperon Lac , Leucócitos/citologia , Fígado/citologia , Fígado/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Transgenes
20.
Curr Gene Ther ; 2(1): 9-21, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12108976

RESUMO

Transplantation offers a unique opportunity for gene transfer into allografts before grafting. After organ retrieval, the cold ischemic period renders organs available for manipulation and gene transfer. Local expression of protective or immunomodulatory molecules within the graft environment offers a better local bioavailability of bioreagents and potentially less systemic side effects. Protection against ischemia-reperfusion injury, acute and/or chronic rejection without significant side effects would be a major breakthrough in transplant research. However, protocols of transfection adapted to the transplant setting and control of gene expression must be clearly evaluated before going to clinical trials. The first part of this review deals with gene transfer techniques into the allograft, emphasizing particular transplant conditions that are encountered and that must be respected when designing protocols for gene transfer experiments. The second part deals with specific therapeutic strategies to protect and prolong allograft survival.


Assuntos
Técnicas de Transferência de Genes , Engenharia Genética/métodos , Transplante Homólogo/métodos , Adenoviridae/genética , Antioxidantes/metabolismo , Cátions , DNA/metabolismo , Dependovirus/genética , Vetores Genéticos , Lentivirus/genética , Leucócitos/metabolismo , Metabolismo dos Lipídeos , Vírus Sendai/genética , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA