Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
PLoS Pathog ; 19(1): e1010351, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36689486

RESUMO

CMV, a ubiquitous herpesvirus, elicits an extraordinarily large T cell response that is sustained or increases over time, a phenomenon termed 'memory inflation.' Remarkably, even latent, non-productive infection can drive memory inflation. Despite intense research on this phenomenon, the infected cell type(s) involved are unknown. To identify the responsible cell type(s), we designed a Cre-lox murine CMV (MCMV) system, where a spread-deficient (ΔgL) virus expresses recombinant SIINFEKL only in Cre+ host cells. We found that latent infection of endothelial cells (ECs), but not dendritic cells (DCs) or hepatocytes, was sufficient to drive CD8 T cell memory inflation. Infection of Lyve-1-Cre and Prox1-CreERT2 mice revealed that amongst EC subsets, infection of lymphatic ECs was sufficient. Genetic ablation of ß2m on lymphatic ECs did not prevent inflation, suggesting another unidentified cell type can also present antigen to CD8 T cells during latency. This novel system definitively shows that antigen presentation by lymphatic ECs drives robust CD8 T cell memory inflation.


Assuntos
Infecções por Citomegalovirus , Infecção Latente , Muromegalovirus , Animais , Camundongos , Células Endoteliais , Linfócitos T CD8-Positivos , Antígenos , Memória Imunológica
3.
Nature ; 502(7469): 100-4, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24025770

RESUMO

Established infections with the human and simian immunodeficiency viruses (HIV and SIV, respectively) are thought to be permanent with even the most effective immune responses and antiretroviral therapies only able to control, but not clear, these infections. Whether the residual virus that maintains these infections is vulnerable to clearance is a question of central importance to the future management of millions of HIV-infected individuals. We recently reported that approximately 50% of rhesus macaques (RM; Macaca mulatta) vaccinated with SIV protein-expressing rhesus cytomegalovirus (RhCMV/SIV) vectors manifest durable, aviraemic control of infection with the highly pathogenic strain SIVmac239 (ref. 5). Here we show that regardless of the route of challenge, RhCMV/SIV vector-elicited immune responses control SIVmac239 after demonstrable lymphatic and haematogenous viral dissemination, and that replication-competent SIV persists in several sites for weeks to months. Over time, however, protected RM lost signs of SIV infection, showing a consistent lack of measurable plasma- or tissue-associated virus using ultrasensitive assays, and a loss of T-cell reactivity to SIV determinants not in the vaccine. Extensive ultrasensitive quantitative PCR and quantitative PCR with reverse transcription analyses of tissues from RhCMV/SIV vector-protected RM necropsied 69-172 weeks after challenge did not detect SIV RNA or DNA sequences above background levels, and replication-competent SIV was not detected in these RM by extensive co-culture analysis of tissues or by adoptive transfer of 60 million haematolymphoid cells to naive RM. These data provide compelling evidence for progressive clearance of a pathogenic lentiviral infection, and suggest that some lentiviral reservoirs may be susceptible to the continuous effector memory T-cell-mediated immune surveillance elicited and maintained by cytomegalovirus vectors.


Assuntos
Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Animais , Citomegalovirus/genética , Citomegalovirus/imunologia , Feminino , Macaca mulatta , Masculino , Dados de Sequência Molecular , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Fatores de Tempo , Vacinas Atenuadas/imunologia , Carga Viral , Replicação Viral/fisiologia
4.
Mol Ther ; 24(8): 1444-55, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27434584

RESUMO

Cytomegalovirus is an attractive cancer vaccine platform because it induces strong, functional CD8(+) T-cell responses that accumulate over time and migrate into most tissues. To explore this, we used murine cytomegalovirus expressing a modified gp100 melanoma antigen. Therapeutic vaccination by the intraperitoneal and intradermal routes induced tumor infiltrating gp100-specific CD8(+) T-cells, but provided minimal benefit for subcutaneous lesions. In contrast, intratumoral infection of established tumor nodules greatly inhibited tumor growth and improved overall survival in a CD8(+) T-cell-dependent manner, even in mice previously infected with murine cytomegalovirus. Although murine cytomegalovirus could infect and kill B16F0s in vitro, infection was restricted to tumor-associated macrophages in vivo. Surprisingly, the presence of a tumor antigen in the virus only slightly increased the efficacy of intratumoral infection and tumor-specific CD8(+) T-cells in the tumor remained dysfunctional. Importantly, combining intratumoral murine cytomegalovirus infection with anti-PD-L1 therapy was synergistic, resulting in tumor clearance from over half of the mice and subsequent protection against tumor challenge. Thus, while a murine cytomegalovirus-based vaccine was poorly effective against established subcutaneous tumors, direct infection of tumor nodules unexpectedly delayed tumor growth and synergized with immune checkpoint blockade to promote tumor clearance and long-term protection.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Imunidade , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Muromegalovirus/fisiologia , Animais , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Terapia Combinada , Expressão Gênica , Ordem dos Genes , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Imunoterapia , Macrófagos/imunologia , Macrófagos/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Resultado do Tratamento , Carga Tumoral , Vacinação , Antígeno gp100 de Melanoma/genética
5.
Nucleic Acids Res ; 41(4): 2644-58, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23303783

RESUMO

The RNA-binding protein AUF1 binds AU-rich elements in 3'-untranslated regions to regulate mRNA degradation and/or translation. Many of these mRNAs are predicted microRNA targets as well. An emerging theme in post-transcriptional control of gene expression is that RNA-binding proteins and microRNAs co-regulate mRNAs. Recent experiments and bioinformatic analyses suggest this type of co-regulation may be widespread across the transcriptome. Here, we identified mRNA targets of AUF1 from a complex pool of cellular mRNAs and examined a subset of these mRNAs to explore the links between RNA binding and mRNA degradation for both AUF1 and Argonaute 2 (AGO2), which is an essential effector of microRNA-induced gene silencing. Depending on the specific mRNA examined, AUF1 and AGO2 binding is proportional/cooperative, reciprocal/competitive or independent. For most mRNAs in which AUF1 affects their decay rates, mRNA degradation requires AGO2. Thus, AUF1 and AGO2 present mRNA-specific allosteric binding relationships for co-regulation of mRNA degradation.


Assuntos
Proteínas Argonautas/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas , Células HeLa , Ribonucleoproteína Nuclear Heterogênea D0 , Humanos , Células K562
6.
J Exp Med ; 204(7): 1519-24, 2007 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-17562817

RESUMO

Antigen-induced immune suppression, like T cell activation, requires antigen-presenting cells (APCs); however, the role of APCs in mediating these opposing effects is not well understood, especially in vivo. We report that genetic inactivation of CD11b, which is a CD18 subfamily of integrin receptors that is highly expressed on APCs, abolishes orally induced peripheral immune tolerance (oral tolerance) without compromising APC maturation or antigen-specific immune activation. The defective oral tolerance in CD11b(-/-) mice can be restored by adoptive transfer of wild-type APCs. CD11b deficiency leads to enhanced interleukin (IL) 6 production by APCs, which subsequently promotes preferential differentiation of naive T cells to T helper 17 (Th17) cells, which are a T cell lineage characterized by their production of IL-17. Consequently, antigen feeding and immunization of CD11b(-/-) mice results in significant production of IL-17 within the draining lymph nodes that interferes with the establishment of oral tolerance. Together, we conclude that CD11b facilitates oral tolerance by suppressing Th17 immune differentiation.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antígeno CD11b/imunologia , Diferenciação Celular/imunologia , Tolerância Imunológica , Linfócitos T/imunologia , Animais , Antígenos/imunologia , Antígeno CD11b/genética , Divisão Celular , Citocinas/metabolismo , Hipersensibilidade Tardia , Interleucina-17/imunologia , Camundongos , Camundongos Knockout
7.
Biochem Biophys Res Commun ; 437(2): 287-91, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-23811402

RESUMO

Cytomegalovirus (CMV) induces strong and long-lasting immune responses, which make it an attractive candidate for a cancer vaccine vector. In this study, we tested whether a tumor antigen expressed in CMV can induce a strong anti-tumor effect. We expressed an unmodified melanoma antigen, mouse tyrosinase-related protein 2 (TRP2), in mouse cytomegalovirus (MCMV). Prophylactic vaccination of the mice with a single dose of MCMV-TRP2 induced rejection of B16 melanoma challenge; therapeutic vaccination with MCMV-TRP2 prolonged the survival of the mice challenged with B16 cells. Additionally, vaccination with MCMV-TRP2 five months before tumor challenge still induced tumor rejection, which indicated that the vaccine induced long-term protection. Furthermore, MCMV-TRP2 protected mice against B16 melanoma challenge regardless of the pre-existing CMV infection. We found that vaccination with MCMV-TRP2 induced long-lasting TRP2 specific antibodies but not CD8 T cells. In addition, depletion of CD4 and CD8 T cells did not compromise the antitumor effect by MCMV-TRP2; while in B cell deficient (µMT) mice, the vaccine lost its antitumor effect. These results indicate that antibodies, not T cells, are important in mediating the antitumor effect during the effector phase by the vaccine. We also made a spread deficient MCMV-TRP2 lacking the essential glycoprotein gL, which showed a similar antitumor effect. In conclusion, our study indicates that tumor antigen (TRP2) expressed in MCMV induces a strong and long-lasting anti-melanoma effect through an antibody-dependent mechanism. Our findings demonstrate that CMV might be a promising vector for the development of cancer vaccines.


Assuntos
Vacinas Anticâncer/uso terapêutico , Oxirredutases Intramoleculares/genética , Melanoma Experimental/prevenção & controle , Animais , Vacinas Anticâncer/genética , Feminino , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL
8.
Mol Cancer ; 9: 176, 2010 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-20604947

RESUMO

BACKGROUND: Components of the microenvironment such as bone marrow stromal cells (BMSCs) are well known to support multiple myeloma (MM) disease progression and resistance to chemotherapy including the proteasome inhibitor bortezomib. However, functional distinctions between BMSCs in MM patients and those in disease-free marrow are not completely understood. We and other investigators have recently reported that NF-kappaB activity in primary MM cells is largely resistant to the proteasome inhibitor bortezomib, and that further enhancement of NF-kappaB by BMSCs is similarly resistant to bortezomib and may mediate resistance to this therapy. The mediating factor(s) of this bortezomib-resistant NF-kappaB activity is induced by BMSCs is not currently understood. RESULTS: Here we report that BMSCs specifically derived from MM patients are capable of further activating bortezomib-resistant NF-kappaB activity in MM cells. This induced activity is mediated by soluble proteinaceous factors secreted by MM BMSCs. Among the multiple factors evaluated, interleukin-8 was secreted by BMSCs from MM patients at significantly higher levels compared to those from non-MM sources, and we found that IL-8 contributes to BMSC-induced NF-kappaB activity. CONCLUSIONS: BMSCs from MM patients uniquely enhance constitutive NF-kappaB activity in MM cells via a proteinaceous secreted factor in part in conjunction with IL-8. Since NF-kappaB is known to potentiate MM cell survival and confer resistance to drugs including bortezomib, further identification of the NF-kappaB activating factors produced specifically by MM-derived BMSCs may provide a novel biomarker and/or drug target for the treatment of this commonly fatal disease.


Assuntos
Antineoplásicos/uso terapêutico , Células da Medula Óssea/patologia , Ácidos Borônicos/uso terapêutico , Mieloma Múltiplo/patologia , NF-kappa B/metabolismo , Pirazinas/uso terapêutico , Células Estromais/patologia , Bortezomib , Humanos , Interleucina-8/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo
9.
Stem Cells ; 27(4): 942-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19353522

RESUMO

Mesenchymal stem cells (MSCs) are potent immunoregulators and have shown clinical utility in suppressing immunity. MSC function is modulated by cytokines, since inflammatory cytokines, such as interferon-gamma (IFNgamma) concomitant with tumor necrosis factor-alpha (TNFalpha), induce their immunoregulatory capability. Here, we show that IFNgamma and TNFalpha act synergistically to induce high levels of expression of interleukin-6 (IL-6) and several other immune-related molecules in MSCs in vitro. We further found that, while either IFNgamma or TNFalpha alone induced minor expression of C/EBPbeta in MSCs, this transcription factor was dramatically upregulated when these cytokines were added together. A causal relationship between C/EBPbeta upregulation and IL-6 expression was demonstrated by small interfering RNA knockdown of C/EBPbeta. C/EBPbeta knockdown also inhibited the synergistic expression of CXCL1, inducible nitric oxide synthase, and CCL5 in response to concomitant IFNgamma and TNFalpha. We conclude that C/EBPbeta is a key transcription factor in synergistic gene upregulation by IFNgamma and TNFalpha. Importantly, C/EBPbeta similarly mediated synergistic gene induction in response to IFNgamma accompanied by IL-1beta or lipopolysaccharide, suggesting that synergy between IFNgamma and other stimuli share C/EBPbeta as common mechanism. Furthermore, while STAT1 is critical in IFNgamma signaling, we found that STAT1 knockdown in MSCs did not affect C/EBPbeta expression or the synergistic induction of IL-6 and CXCL1 by IFNgamma and TNFalpha. Thus, C/EBPbeta is not regulated by STAT1. These results demonstrate the importance of cytokine interactions in MSC immunobiology, a better understanding of which will allow improved clinical application of these cells.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação da Expressão Gênica/fisiologia , Interferon gama/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Western Blotting , Células da Medula Óssea/metabolismo , Quimiocina CCL5/metabolismo , Quimiocina CXCL1/metabolismo , Expressão Gênica , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/metabolismo , Regulação para Cima
10.
J Clin Invest ; 116(9): 2434-41, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16906223

RESUMO

IFN-gamma is an important Th1 proinflammatory cytokine and has a paradoxical effect on EAE in which disease susceptibility is unexpectedly heightened in IFN-gamma-deficient mice. In this study, we provide what we believe is new evidence indicating that IFN-gamma is critically required for the conversion of CD4+ CD25- T cells to CD4+ Tregs during EAE. In our study, the added severity of EAE in IFN-gamma knockout mice was directly associated with altered encephalitogenic T cell responses, which correlated with reduced frequency and function of CD4+ CD25+ Foxp3+ Tregs when compared with those of WT mice. It was demonstrated in both human and mouse systems that in vitro IFN-gamma treatment of CD4+ CD25- T cells led to conversion of CD4+ Tregs as characterized by increased expression of Foxp3 and enhanced regulatory function. Mouse CD4+ CD25- T cells, when treated in vitro with IFN-gamma, acquired marked regulatory properties as evidenced by suppression of EAE by adoptive transfer. These findings have important implications for the understanding of the complex role of IFN-gamma in both induction and self regulation of inflammatory processes.


Assuntos
Antígenos CD4/imunologia , Encefalomielite Autoimune Experimental/imunologia , Fatores de Transcrição Forkhead/genética , Interferon gama/fisiologia , Receptores de Interleucina-2/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Encéfalo/imunologia , Interferon gama/deficiência , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Proteínas da Mielina , Glicoproteína Associada a Mielina/genética , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/química , Reação em Cadeia da Polimerase , Medula Espinal/imunologia
11.
PLoS One ; 14(1): e0210252, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30673723

RESUMO

The development of a sterilizing vaccine against malaria remains one of the highest priorities for global health research. While sporozoite vaccines targeting the pre-erythrocytic stage show great promise, it has not been possible to maintain efficacy long-term, likely due to an inability of these vaccines to maintain effector memory T cell responses in the liver. Vaccines based on human cytomegalovirus (HCMV) might overcome this limitation since vectors based on rhesus CMV (RhCMV), the homologous virus in rhesus macaques (RM), elicit and indefinitely maintain high frequency, non-exhausted effector memory T cells in extralymphoid tissues, including the liver. Moreover, RhCMV strain 68-1 elicits CD8+ T cells broadly recognizing unconventional epitopes exclusively restricted by MHC-II and MHC-E. To evaluate the potential of these unique immune responses to protect against malaria, we expressed four Plasmodium knowlesi (Pk) antigens (CSP, AMA1, SSP2/TRAP, MSP1c) in RhCMV 68-1 or in Rh189-deleted 68-1, which additionally elicits canonical MHC-Ia-restricted CD8+ T cells. Upon inoculation of RM with either of these Pk Ag expressing RhCMV vaccines, we obtained T cell responses to each of the four Pk antigens. Upon challenge with Pk sporozoites we observed a delayed appearance of blood stage parasites in vaccinated RM consistent with a 75-80% reduction of parasite release from the liver. Moreover, the Rh189-deleted RhCMV/Pk vectors elicited sterile protection in one RM. Once in the blood, parasite growth was not affected. In contrast to T cell responses induced by Pk infection, RhCMV vectors maintained sustained T cell responses to all four malaria antigens in the liver post-challenge. The delayed appearance of blood stage parasites is thus likely due to a T cell-mediated inhibition of liver stage parasite development. As such, this vaccine approach can be used to efficiently test new T cell antigens, improve current vaccines targeting the liver stage and complement vaccines targeting erythrocytic antigens.


Assuntos
Antígenos de Protozoários/imunologia , Citomegalovirus/genética , Vacinas Antimaláricas/imunologia , Malária/imunologia , Parasitemia/imunologia , Plasmodium knowlesi/imunologia , Esporozoítos/imunologia , Animais , Anopheles/imunologia , Anopheles/parasitologia , Feminino , Vetores Genéticos/administração & dosagem , Memória Imunológica , Fígado/imunologia , Fígado/parasitologia , Macaca mulatta , Malária/sangue , Malária/parasitologia , Malária/prevenção & controle , Masculino , Parasitemia/sangue , Parasitemia/parasitologia , Parasitemia/prevenção & controle , Plasmodium knowlesi/genética , Proteínas de Protozoários/imunologia , Linfócitos T/imunologia , Linfócitos T/parasitologia
12.
J Clin Invest ; 115(4): 1060-7, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15761492

RESUMO

Osteopontin (OPN) is an extracellular matrix protein of pleiotropic properties and has been recently recognized as a potential inflammatory cytokine. In this study, we demonstrate, for the first time to our knowledge, that overexpression of OPN in synovial T cells is associated with local inflammatory milieu and that OPN acts as an important mediator in amplification and perpetuation of rheumatoid synovitis. The study revealed that mRNA expression of OPN was highly elevated in CD4(+) synovial T cells derived from patients with RA, which correlated with increased OPN concentrations in synovial fluid (SF). The pattern of OPN overexpression was confined to rheumatoid synovium and correlated with coexpression of selected OPN receptors in synovial T cells, including integrins alphav and beta1 and CD44. RA-derived SF stimulated the expression of OPN in T cells, which was attributable to IL-10 present in SF and abrogated by anti-IL-10 antibody. Among the more than 300 autoimmune and inflammatory response genes examined, OPN selectively induced the expression of proinflammatory cytokines and chemokines known to promote migration and recruitment of inflammatory cells. Furthermore, it was evident that OPN activated transcription factor NF-kappaB in mononuclear cells. The study has important implications for understanding the role of OPN in rheumatoid synovitis and other inflammatory conditions.


Assuntos
Citocinas/imunologia , Sialoglicoproteínas/fisiologia , Sinovite/imunologia , Adulto , Idoso , Artrite Reumatoide/imunologia , Citocinas/sangue , Citocinas/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Interleucina-10/sangue , Interleucina-10/genética , Interleucina-10/imunologia , Masculino , Pessoa de Meia-Idade , NF-kappa B/genética , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Osteopontina , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Sialoglicoproteínas/genética , Sialoglicoproteínas/imunologia , Sinovite/patologia , Subpopulações de Linfócitos T , Linfócitos T/imunologia
13.
Biochem Biophys Res Commun ; 375(4): 666-70, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18755151

RESUMO

Bone marrow stromal cells (BMSCs) have been shown to promote the growth and survival of a wide variety of tumors. However, in the present study, we found that BMSCs induced apoptosis of lymphoma cells in the presence of INFgamma and TNF. IFNgamma and TNF dramatically induced the expression of inducible nitric oxide synthase (iNOS) by BMSCs in culture, and BMSCs generated from iNOS knockout mice did not induce apoptosis of lymphoma cells in the presence of IFNgamma and TNF. In addition, we found that IFNgamma and TNF also increased IL-6 expression by BMSCs, and anti-IL-6 further increased the killing of tumor cells by BMSCs. Taken together, our findings indicate that BMSCs induce apoptosis of lymphoma cells in the presence of IFNgamma and TNF, and that the proapoptotic effect of BMSCs is mediated by nitric oxide. Our findings suggest a possibility to harness this proapoptotic feature of BMSCs for the development of novel therapeutic strategy to eliminate tumor cells, especially tumor cells in bone marrow.


Assuntos
Apoptose , Células da Medula Óssea/efeitos dos fármacos , Interferon gama/farmacologia , Linfoma/terapia , Óxido Nítrico/biossíntese , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células da Medula Óssea/enzimologia , Linhagem Celular Tumoral , Linfoma/patologia , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Células Estromais/efeitos dos fármacos , Células Estromais/enzimologia
14.
Nat Med ; 24(2): 130-143, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29334373

RESUMO

Despite widespread use of the bacille Calmette-Guérin (BCG) vaccine, tuberculosis (TB) remains a leading cause of global mortality from a single infectious agent (Mycobacterium tuberculosis or Mtb). Here, over two independent Mtb challenge studies, we demonstrate that subcutaneous vaccination of rhesus macaques (RMs) with rhesus cytomegalovirus vectors encoding Mtb antigen inserts (hereafter referred to as RhCMV/TB)-which elicit and maintain highly effector-differentiated, circulating and tissue-resident Mtb-specific CD4+ and CD8+ memory T cell responses-can reduce the overall (pulmonary and extrapulmonary) extent of Mtb infection and disease by 68%, as compared to that in unvaccinated controls, after intrabronchial challenge with the Erdman strain of Mtb at ∼1 year after the first vaccination. Fourteen of 34 RhCMV/TB-vaccinated RMs (41%) across both studies showed no TB disease by computed tomography scans or at necropsy after challenge (as compared to 0 of 17 unvaccinated controls), and ten of these RMs were Mtb-culture-negative for all tissues, an exceptional long-term vaccine effect in the RM challenge model with the Erdman strain of Mtb. These results suggest that complete vaccine-mediated immune control of highly pathogenic Mtb is possible if immune effector responses can intercept Mtb infection at its earliest stages.


Assuntos
Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Vacina BCG/imunologia , Citomegalovirus/imunologia , Macaca mulatta/imunologia
15.
Cell Res ; 16(2): 126-33, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16474424

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is an important hematopoietic growth factor and immune modulator. GM-CSF also has profound effects on the functional activities of various circulating leukocytes. It is produced by a variety of cell types including T cells, macrophages, endothelial cells and fibroblasts upon receiving immune stimuli. Although GM-CSF is produced locally, it can act in a paracrine fashion to recruit circulating neutrophils, monocytes and lymphocytes to enhance their functions in host defense. Recent intensive investigations are centered on the application of GM-CSF as an immune adjuvant for its ability to increase dendritic cell (DC) maturation and function as well as macrophage activity. It is used clinically to treat neutropenia in cancer patients undergoing chemotherapy, in AIDS patients during therapy, and in patients after bone marrow transplantation. Interestingly, the hematopoietic system of GM-CSF-deficient mice appears to be normal; the most significant changes are in some specific T cell responses. Although molecular cloning of GM-CSF was carried out using cDNA library of T cells and it is well known that the T cells produce GM-CSF after activation, there is a lack of systematic investigation of this cytokine in production by T cells and its effect on T cell function. In this article, we will focus mainly on the immunobiology of GM-CSF in T cells.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Ativação Linfocitária , Linfócitos T/metabolismo , Animais , Vacinas Anticâncer , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Inflamação/imunologia , Fenótipo
16.
Chin Med J (Engl) ; 116(4): 524-8, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12875715

RESUMO

OBJECTIVES: To obtain prokaryotic expressed IA-2 recombinant protein and to identify its immunological activity. METHODS: The complimentary DNA (cDNA) coding for the intracytoplasmic part of IA-2 (IA-2ic) was amplified from human fetal brain RNA, and was subcloned into the PinPoint Xa-1 T vector to construct recombinant expression plasmid, and was then expressed in E. coli JM109 cells as a fusion protein with a biotinylated peptide sequence at the aminoterminus. The biotinylated fusion protein was then purified by affinity chromatography and was subsequently dialyzed. Finally, its immunogenicity was evaluated by enzyme linked immunosorbent assay (ELISA). RESULTS: The purified IA-2ic fusion protein resolved on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) as a single Coomassie brilliant blue stained band with a molecular weight of 59 kDa and its immunogenicity was confirmed by ELISA. CONCLUSIONS: E. coli expressed IA-2ic fusion protein has immunological activity. It can be used for detection of IA-2 autoantibodies (IA-2A) and for further studies on type 1 diabetes in future.


Assuntos
Autoantígenos/biossíntese , Diabetes Mellitus Tipo 1/imunologia , Proteínas de Membrana/biossíntese , Proteínas Tirosina Fosfatases/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Animais , DNA Complementar/análise , Escherichia coli/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Plasmídeos , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/isolamento & purificação , Coelhos , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/isolamento & purificação
17.
Chin Med J (Engl) ; 116(4): 529-33, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12875716

RESUMO

OBJECTIVES: To identify differences in gene expression in renal and visceral adipose tissue in type 2 diabetic rats using cDNA representational difference analysis (RDA) and to explore the molecular pathogenesis of type 2 diabetes and its chronic vascular complications. METHODS: A rat model of type 2 diabetes was generated by administration of a high fat and calorie diet combined with a low dose of streptozocin (STZ) injected into the tail vein. The difference bands were generated by cDNA representational difference analysis (cDNA RDA). The final difference products were ligated into the pUC-18 vector and sequenced. A bioformatics analysis was performed on the obtained expressed sequence tags (ESTs), and then the expression levels of known and novel genes were verified by semi-quantitative reverse transcription-PCR (RT-PCR). At the same time, full-length cDNA of a novel gene was cloned in silico. RESULTS: The type 2 diabetic rats in this experiment experienced hyperglycemia, lipidemia, lower insulin sensitivity and normal body weight. We obtained 9 novel ESTs and 2 novel genes from renal tissue of rats and 6 novel ESTs and 1 known gene, the rat lipoprotein lipase (LPL) gene from their visceral adipose tissue. The 2 novel genes (RS91 and RS2) from the renal tissue were both very similar to serine (or cysteine) proteinase inhibitor, clade F and eukaryotic translation initiation factor 3 and subunit 5 (EIF-3 epsilon). The expression of both novel genes and the LPL gene were upregulated in renal and visceral adipose tissue of type 2 diabetic and fat-enriched rats. Full-length cDNA of the novel gene RS91 was cloned in silico. CONCLUSIONS: (1) The rat model of type 2 diabetes generated in this study was ideal because the disease in the animals closely mimicked type 2 diabetic patients. (2) cDNA RDA is a flexible, inexpensive, more accurate, sensitive and highly effective technique for identifying differences in gene expression. (3) Six novel ESTs and 1 known gene were obtained from rat visceral adipose tissue. The LPL gene was upregulated in adipose tissue of type 2 diabetic and fat-enriched rats, a result possibly related to the diabetic animals' high fat and calorie diet, lipidemia, insulin resistance (RI) and molecular pathogenesis. (4) Nine novel ESTs and 2 novel genes were obtained from the renal tissue of rat. We believe the 2 novel genes to be the serine proteinase inhibitors clade F and EIF-3 epsilon in rats. The upregulation of the 2 novel genes in renal tissue of type 2 diabetic rats and may have been related to their renal impairment.


Assuntos
Tecido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Perfilação da Expressão Gênica , Rim/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Clonagem Molecular , Etiquetas de Sequências Expressas , Masculino , Plasmídeos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vísceras
18.
J Clin Invest ; 124(5): 1928-44, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24691437

RESUMO

The most abundantly produced virion protein in human cytomegalovirus (HCMV) is the immunodominant phosphoprotein 65 (pp65), which is frequently included in CMV vaccines. Although it is nonessential for in vitro CMV growth, pp65 displays immunomodulatory functions that support a potential role in primary and/or persistent infection. To determine the contribution of pp65 to CMV infection and immunity, we generated a rhesus CMV lacking both pp65 orthologs (RhCMVΔpp65ab). While deletion of pp65ab slightly reduced growth in vitro and increased defective particle formation, the protein composition of secreted virions was largely unchanged. Interestingly, pp65 was not required for primary and persistent infection in animals. Immune responses induced by RhCMVΔpp65ab did not prevent reinfection with rhesus CMV; however, reinfection with RhCMVΔUS2-11, which lacks viral-encoded MHC-I antigen presentation inhibitors, was prevented. Unexpectedly, induction of pp65b-specific T cells alone did not protect against RhCMVΔUS2-11 challenge, suggesting that T cells targeting multiple CMV antigens are required for protection. However, pp65-specific immunity was crucial for controlling viral dissemination during primary infection, as indicated by the marked increase of RhCMVΔpp65ab genome copies in CMV-naive, but not CMV-immune, animals. Our data provide rationale for inclusion of pp65 into CMV vaccines but also demonstrate that pp65-induced T cell responses alone do not recapitulate the protective effect of natural infection.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Fosfoproteínas/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Apresentação de Antígeno/imunologia , Linhagem Celular , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/patologia , Vacinas contra Citomegalovirus/genética , Vacinas contra Citomegalovirus/imunologia , Deleção de Genes , Humanos , Macaca mulatta , Camundongos , Fosfoproteínas/genética , Linfócitos T/imunologia , Linfócitos T/patologia , Proteínas da Matriz Viral/genética
19.
Science ; 340(6135): 1237874, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23704576

RESUMO

CD8(+) T cell responses focus on a small fraction of pathogen- or vaccine-encoded peptides, and for some pathogens, these restricted recognition hierarchies limit the effectiveness of antipathogen immunity. We found that simian immunodeficiency virus (SIV) protein-expressing rhesus cytomegalovirus (RhCMV) vectors elicit SIV-specific CD8(+) T cells that recognize unusual, diverse, and highly promiscuous epitopes, including dominant responses to epitopes restricted by class II major histocompatibility complex (MHC) molecules. Induction of canonical SIV epitope-specific CD8(+) T cell responses is suppressed by the RhCMV-encoded Rh189 gene (corresponding to human CMV US11), and the promiscuous MHC class I- and class II-restricted CD8(+) T cell responses occur only in the absence of the Rh157.5, Rh157.4, and Rh157.6 (human CMV UL128, UL130, and UL131) genes. Thus, CMV vectors can be genetically programmed to achieve distinct patterns of CD8(+) T cell epitope recognition.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/imunologia , Epitopos de Linfócito T/imunologia , Vetores Genéticos/imunologia , Vacinas contra a SAIDS/imunologia , Animais , Citocinas/imunologia , Citomegalovirus/genética , Feminino , Vetores Genéticos/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Macaca mulatta , Masculino , Glicoproteínas de Membrana/genética , Vacinas contra a SAIDS/administração & dosagem , Proteínas do Envelope Viral/genética
20.
J Immunol ; 181(1): 190-6, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18566384

RESUMO

IL-17-secreting CD4+ T cells (Th17 cells) play a critical role in immune responses to certain infections and in the development of many autoimmune disorders. The mechanisms controlling homeostasis in this cell population are largely unknown. In this study, we show that murine Th17 cells undergo rapid apoptosis in vitro upon restimulation through the TCR. This activation-induced cell death (AICD), a common mechanism for elimination of activated T cells, required the Fas and FasL interaction: Fas was stably expressed, while FasL was up-regulated upon TCR reactivation of Th17 cells; Ab ligation of Fas induced Th17 cell death; and AICD was completely absent in Th17 cells differentiated from gld/gld CD4+ T cells. Thus, the Fas/FasL pathway is essential in regulating the AICD of Th17 cells. Interestingly, IFN-gamma, a cytokine previously found to be important for the AICD of T cells, did not affect Th17 cell apoptosis. Furthermore, Th17 cells derived from mice deficient in IFN-gamma receptor 1 (IFN-gammaR1-/-) underwent AICD similar to wild-type cells. Thus, AICD of Th17 cells occurs via the Fas pathway, but is independent of IFN-gamma.


Assuntos
Apoptose/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Receptor fas/metabolismo , Animais , Células Cultivadas , Proteína Ligante Fas/metabolismo , Interferon gama/metabolismo , Camundongos , Camundongos Transgênicos , Receptor Cross-Talk/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/metabolismo , Células Th2 , Fatores de Tempo , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA