Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Environ Manage ; 355: 120504, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38447513

RESUMO

Ammonia-oxidation process directly contribute to soil nitrous oxide (N2O) emissions in agricultural soils. However, taxonomy of the key nitrifiers (within ammonia oxidising bacteria (AOB), archaea (AOA) and complete ammonia oxidisers (comammox Nitrospira)) responsible for substantial N2O emissions in agricultural soils is unknown, as is their regulation by soil biotic and abiotic factors. In this study, cumulative N2O emissions, nitrification rates, abundance and community structure of nitrifiers were investigated in 16 agricultural soils from major crop production regions of China using microcosm experiments with amended nitrogen (N) supplemented or not with a nitrification inhibitor (nitrapyrin). Key nitrifier groups involved in N2O emissions were identified by comparative analyses of the different treatments, combining sequencing and random forest analyses. Soil cumulative N2O emissions significantly increased with soil pH in all agricultural soils. However, they decreased with soil organic carbon (SOC) in alkaline soils. Nitrapyrin significantly inhibited soil cumulative N2O emissions and AOB growth, with a significant inhibition of the AOB Nitrosospira cluster 3a.2 (D11) abundance. One Nitrosospira multiformis-like OTU phylotype (OTU34), which was classified within the AOB Nitrosospira cluster 3a.2 (D11), had the greatest importance on cumulative N2O emissions and its growth significantly depended on soil pH and SOC contents, with higher growth at high pH and low SOC conditions. Collectively, our results demonstrate that alkaline soils with low SOC contents have high N2O emissions, which were mainly driven by AOB Nitrosospira cluster 3a.2 (D11). Nitrapyrin can efficiently reduce nitrification-related N2O emissions by inhibiting the activity of AOB Nitrosospira cluster 3a.2 (D11). This study advances our understanding of key nitrifiers responsible for high N2O emissions in agricultural soils and their controlling factors, and provides vital knowledge for N2O emission mitigation in agricultural ecosystems.


Assuntos
Ecossistema , Solo , Solo/química , Amônia/química , Carbono , Oxirredução , Archaea , Nitrificação , Microbiologia do Solo
2.
Reproduction ; 162(6): 461-472, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34591784

RESUMO

As a multifunctional transcription factor, YY1 regulates the expression of many genes essential for early embryonic development. RTCB is an RNA ligase that plays a role in tRNA maturation and Xbp1 mRNA splicing. YY1 can bind in vitro to the response element in the proximal promoter of Rtcb and regulate Rtcb promoter activity. However, the in vivo regulation and whether these two genes are involved in the mother-fetal dialogue during early pregnancy remain unclear. In this study, we validated that YY1 bound in vivo to the proximal promoter of Rtcb in mouse uterus of early pregnancy. Moreover, via building a variety of animal models, our study suggested that both YY1 and RTCB might play a role in mouse uterus decidualization and embryo implantation during early pregnancy.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Implantação do Embrião , Fatores de Transcrição , Fator de Transcrição YY1/metabolismo , Animais , Decídua/fisiologia , Implantação do Embrião/fisiologia , Feminino , Camundongos , Gravidez , Splicing de RNA , Fatores de Transcrição/genética , Útero
3.
Mol Ther ; 24(9): 1615-26, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27401038

RESUMO

Immunotherapy with CD123-specific T-cell engager proteins or with T cells expressing CD123-specific chimeric antigen receptors is actively being pursued for acute myeloid leukemia. T cells secreting bispecific engager molecules (ENG-T cells) may present a promising alternative to these approaches. To evaluate therapeutic potential, we generated T cells to secrete CD123/CD3-bispecific engager molecules. CD123-ENG T cells recognized primary acute myeloid leukemia (AML) cells and cell lines in an antigen-dependent manner as judged by cytokine production and/or tumor killing, and redirected bystander T cells to AML cells. Infusion of CD123-ENG T cells resulted in regression of AML in xenograft models conferring a significant survival advantage of treated mice in comparison to mice that received control T cells. At high effector to target ratios, CD123-ENG T cells recognized normal hematopoietic stem and progenitor cells (HSPCs) with preferential recognition of HSPCs from cord blood compared to bone marrow. We therefore introduced the CD20 suicide gene that can be targeted in vivo with rituximab into CD123-ENG T cells. The expression of CD20 did not diminish the anti-AML activity of CD123-ENG T cells, but allowed for rituximab-mediated ENG-T cell elimination. Thus, ENG-T cells coexpressing CD20 suicide and CD123 engager molecules may present a promising immunotherapeutic approach for AML.


Assuntos
Imunoterapia , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos CD20/genética , Antígenos CD20/metabolismo , Complexo CD3/genética , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Citocinas/metabolismo , Citotoxicidade Imunológica , Modelos Animais de Doenças , Genes Transgênicos Suicidas/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunoterapia/métodos , Subunidade alfa de Receptor de Interleucina-3/genética , Leucemia Mieloide Aguda/genética , Camundongos , Retroviridae/genética , Rituximab/farmacologia , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Ther ; 23(1): 171-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25142939

RESUMO

Adoptive immunotherapy with antigen-specific T cells has shown promise for the treatment of malignancies. However, infused T cells are unable to redirect resident T cells, limiting potential benefit. While the infusion of bispecific T-cell engagers can redirect resident T cells to tumors, these molecules have a short half-life, and do not self amplify. To overcome these limitations, we generated T cells expressing a secretable T-cell engager specific for CD3 and EphA2, an antigen expressed on a broad range of human tumors (EphA2-ENG T cells). EphA2-ENG T cells were activated and recognized tumor cells in an antigen-dependent manner, redirected bystander T cells to tumor cells, and had potent antitumor activity in glioma and lung cancer severe combined immunodeficiency (SCID) xenograft models associated with a significant survival benefit. This new class of tumor-specific T cells, with the unique ability to redirect bystander T cells, may be a promising alternative to current immunotherapies for cancer.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/terapia , Glioma/terapia , Imunoterapia Adotiva/métodos , Neoplasias Pulmonares/terapia , Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Efeito Espectador/imunologia , Complexo CD3/genética , Complexo CD3/imunologia , Linhagem Celular Tumoral , Expressão Gênica , Vetores Genéticos , Glioma/genética , Glioma/imunologia , Glioma/patologia , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Receptor EphA2/genética , Receptor EphA2/imunologia , Retroviridae/genética , Análise de Sobrevida , Linfócitos T/citologia , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Ther ; 22(1): 102-11, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24135899

RESUMO

Oncolytic vaccinia virus (VV) therapy has shown promise in preclinical models and in clinical studies. However, complete responses have rarely been observed. This lack of efficacy is most likely due to suboptimal virus spread through the tumor resulting in limited tumor cell destruction. We reasoned that redirecting T cells to the tumor has the potential to improve the antitumor activity of oncolytic VVs. We, therefore, constructed a VV encoding a secretory bispecific T-cell engager consisting of two single- chain variable fragments specific for CD3 and the tumor cell surface antigen EphA2 (EphA2-T-cell engager-armed VV (EphA2-TEA-VV)). In vitro, EphA2-TEA-VV's ability to replicate and induce oncolysis was similar to that of unmodified virus. However, only tumor cells infected with EphA2-TEA-VV induced T-cell activation as judged by the secretion of interferon-γ and interleukin-2. In coculture assays, EphA2-TEA-VV not only killed infected tumor cells, but in the presence of T cells, it also induced bystander killing of noninfected tumor cells. In vivo, EphA2-TEA-VV plus T cells had potent antitumor activity in comparison with control VV plus T cells in a lung cancer xenograft model. Thus, arming oncolytic VVs with T-cell engagers may represent a promising approach to improve oncolytic virus therapy.


Assuntos
Vetores Genéticos/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Vírus Oncolíticos/imunologia , Linfócitos T/imunologia , Vaccinia virus/imunologia , Animais , Efeito Espectador/genética , Efeito Espectador/imunologia , Complexo CD3/genética , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Modelos Animais de Doenças , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/genética , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Ativação Linfocitária/imunologia , Neoplasias/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Receptor EphA2/genética , Receptor EphA2/metabolismo , Linfócitos T/metabolismo , Vaccinia virus/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Ther ; 21(8): 1611-20, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23732988

RESUMO

Cancer-associated fibroblasts (CAFs), the principle component of the tumor-associated stroma, form a highly protumorigenic and immunosuppressive microenvironment that mediates therapeutic resistance. Co-targeting CAFs in addition to cancer cells may therefore augment the antitumor response. Fibroblast activation protein-α (FAP), a type 2 dipeptidyl peptidase, is expressed on CAFs in a majority of solid tumors making it an attractive immunotherapeutic target. To target FAP-positive CAFs in the tumor-associated stroma, we genetically modified T cells to express a FAP-specific chimeric antigen receptor (CAR). The resulting FAP-specific T cells recognized and killed FAP-positive target cells as determined by proinflammatory cytokine release and target cell lysis. In an established A549 lung cancer model, adoptive transfer of FAP-specific T cells significantly reduced FAP-positive stromal cells, with a concomitant decrease in tumor growth. Combining these FAP-specific T cells with T cells that targeted the EphA2 antigen on the A549 cancer cells themselves significantly enhanced overall antitumor activity and conferred a survival advantage compared to either alone. Our study underscores the value of co-targeting both CAFs and cancer cells to increase the benefits of T-cell immunotherapy for solid tumors.


Assuntos
Fibroblastos/imunologia , Gelatinases/imunologia , Proteínas de Membrana/imunologia , Neoplasias/imunologia , Receptores de Antígenos/imunologia , Serina Endopeptidases/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Citocinas/biossíntese , Citotoxicidade Imunológica , Modelos Animais de Doenças , Endopeptidases , Fibroblastos/metabolismo , Gelatinases/genética , Gelatinases/metabolismo , Expressão Gênica , Ordem dos Genes , Vetores Genéticos , Humanos , Imunoterapia , Mediadores da Inflamação/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Neoplasias/metabolismo , Neoplasias/mortalidade , Neoplasias/patologia , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Linfócitos T/metabolismo
7.
J Immunol ; 185(7): 4223-32, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20817880

RESUMO

To function optimally as vaccines, dendritic cells (DCs) must actively migrate to lymphoid organs and maintain a viable, mature state for sufficient time to effectively present their Ag to cognate T cells. Unfortunately, mature DCs rapidly lose viability and function after injection, and only a minority leaves the vaccine site and migrates to lymph nodes. We show that all of these functions can be enhanced in DCs by removal of IL-1R-associated kinase M (IRAK-M). We found that IRAK-M is induced in DCs by TLR ligation and that its absence from these cells leads to increased activation of the p38-MAPK and NF-κB pathways, which, in turn, improves DC migration to lymph nodes, increases their longevity, and augments their secretion of Th1-skewing cytokines and chemokines. These biological effects have immunological consequences. IRAK-M(-/-) DCs increase the proliferation and activation of Ag-specific T cells, and a single vaccination with Ag-pulsed, LPS-matured IRAK-M(-/-) DCs eliminates established tumors and prolongs the survival of EG7 or B16.f10 tumor-bearing mice, without discernible induction of autoimmune disease. Thus, manipulation of IRAK-M levels can increase the potency of DC vaccines by enhancing their Ag-presenting function, migration, and longevity.


Assuntos
Vacinas Anticâncer/imunologia , Quimiotaxia de Leucócito/imunologia , Células Dendríticas/imunologia , Quinases Associadas a Receptores de Interleucina-1/imunologia , Animais , Apresentação de Antígeno/imunologia , Western Blotting , Vacinas Anticâncer/metabolismo , Proliferação de Células , Separação Celular , Sobrevivência Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
8.
Mol Ther ; 19(1): 211-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20959814

RESUMO

Although the benefits of adoptive T-cell therapy can be increased by prior lymphodepletion of the recipient, this process usually requires chemotherapy or radiation. Vaccination with antigens to which the transferred T cells respond should be a less toxic means of enhancing their activity, but to date such vaccines have not been effective. We, therefore, determined which characteristics an adenoviral vaccine has to fulfill to optimally activate and expand adoptively transferred antigen-specific T cells in vivo. We evaluated (i) antigen, (ii) flagellin, a Toll-like receptor (TLR) 5 ligand, and (iii) an inhibitor of the antigen-presenting attenuator A20. Vaccination of mice before T-cell transfer with a vaccine that contained all three components dramatically enhanced the effector function of ovalbumin (OVA)-specific T cells as judged by the regression of established B16-OVA tumors compared to one- and two-component vaccines. Immunization with the three-component vaccine induced a strong Th1 environment, which was critical for the observed synergy and proved as effective as cytoxan-induced lymphodepletion in enhancing in vivo T-cell expansion. Thus, the combination of our vaccine with T-cell therapy has the potential to enhance and broaden adoptive cellular immunotherapy.


Assuntos
Adenoviridae/imunologia , Vacinas Anticâncer/imunologia , Imunoterapia Adotiva/métodos , Linfócitos T/imunologia , Células Th1/imunologia , Vacinas Virais/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos/imunologia , Linhagem Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Flagelina/genética , Imunização Secundária/métodos , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia , RNA Interferente Pequeno/genética , Vacinas Sintéticas/genética , Vacinas Sintéticas/metabolismo
10.
Adv Cell Gene Ther ; 4(2)2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33829146

RESUMO

Oncolytic virotherapy has been tested in numerous early phase clinical studies. However, the antitumor activity of oncolytic viruses thus far has been limited. Numerous strategies are being explored to enhance their antitumor activity by activating the adaptive arm of the immune system. We reasoned that it might also be possible to engineer oncolytic viruses to redirect tumor-associated macrophages to tumor cells for therapeutic benefit. We engineered an oncolytic vaccinia virus (VV) to disrupt the CD47/SIRPα interaction by expressing a chimeric molecule that consists of the ectodomain of SIRPα and the Fc domain of IgG4 (SIRPα-Fc-VV). SIRPα-Fc-VV readily replicated in tumor cells and redirected M1 as well as M2 macrophages to tumor cells in vitro. In contrast, control VVs that either encoded YFP (YFP-VV) or SIRPα (SIRPα-VV) did not. In vivo, SIRPα-Fc-VV had greater antitumor activity than YFP-VV and SIRPα-VV in an immune competent osteosarcoma model resulting in a significant survival advantage. Pretreatment with cytoxan further augmented the antitumor activity of SIRPα-Fc-VV. Thus, arming oncolytic viruses with SIRPα-Fc may present a promising strategy to enhance their antitumor activity for the virotherapy of solid tumors.

11.
Biomedicines ; 8(7)2020 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-32664210

RESUMO

Oncolytic viruses (OVs) are potent anti-cancer biologics with a bright future, having substantial evidence of efficacy in patients with cancer. Bi- and tri-specific antibodies targeting tumor antigens and capable of activating T cell receptor signaling have also shown great promise in cancer immunotherapy. In a cutting-edge strategy, investigators have incorporated the two independent anti-cancer modalities, transforming them into bi- or tri-specific T cell engager (BiTE or TriTE)-armed OVs for targeted immunotherapy. Since 2014, multiple research teams have studied this combinatorial strategy, and it showed substantial efficacy in various tumor models. Here, we first provide a brief overview of the current status of oncolytic virotherapy and the use of multi-specific antibodies for cancer immunotherapy. We then summarize progress on BiTE and TriTE antibodies as a novel class of cancer therapeutics in preclinical and clinical studies, followed by a discussion of BiTE- or TriTE-armed OVs for cancer therapy in translational models. In addition, T cell receptor mimics (TCRm) have been developed into BiTEs and are expected to greatly expand the application of BiTEs and BiTE-armed OVs for the effective targeting of intracellular tumor antigens. Future applications of such innovative combination strategies are emerging as precision cancer immunotherapies.

12.
J Clin Invest ; 116(1): 90-100, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16357940

RESUMO

DC-based tumor vaccine research has largely focused on enhancing DC maturation/costimulation and antigen presentation in order to break tolerance against self tumor-associated antigens. DC immunization can activate autoreactive T cells but rarely causes autoimmune pathologies, indicating that self tolerance at the host level is still maintained in the vaccinated hosts. This study in mice reveals a novel regulatory mechanism for the control of self tolerance at the host level by DCs through the restriction of positive cytokine feedback loops by cytokine signaling inhibitor SOCS1. The study further finds the requirement of persistent antigen presentation by DCs for inducing pathological autoimmune responses against normal tissues and tumor, which can be achieved by silencing SOCS1 to unleash the unbridled signaling of IL-12 and the downstream cytokine cascade. However, the use of higher-affinity self peptides, enhancement of DC maturation, and persistent stimulation with cytokines or TLR agonists fail to break tolerance and induce pathological antitumor immunity. Thus, this study indicates the necessity of inhibiting SOCS1, an antigen presentation attenuator, to break self tolerance and induce effective antitumor responses.


Assuntos
Proteínas de Transporte/fisiologia , Células Dendríticas/imunologia , Interleucina-12/genética , Melanoma Experimental/imunologia , Proteínas Repressoras/fisiologia , Tolerância a Antígenos Próprios/fisiologia , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica , Interleucina-12/deficiência , Interleucina-12/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/imunologia , Proteína 1 Supressora da Sinalização de Citocina
13.
JCI Insight ; 3(3)2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29415891

RESUMO

BACKGROUND: DC-based tumor vaccines have had limited clinical success thus far. SOCS1, a key inhibitor of inflammatory cytokine signaling, is an immune checkpoint regulator that limits DC immunopotency. METHODS: We generated a genetically modified DC (gmDC) vaccine to perform immunotherapy. The adenovirus (Ad-siSSF) delivers two tumor-associated antigens (TAAs), survivin and MUC1; secretory bacterial flagellin for DC maturation; and an RNA interference moiety to suppress SOCS1. A 2-stage phase I trial was performed for patients with relapsed acute leukemia after allogenic hematopoietic stem cell transplantation: in stage 1, we compared the safety and efficacy between gmDC treatment (23 patients) and standard donor lymphocyte infusion (25 patients); in stage 2, we tested the efficacy of the gmDC vaccine for 12 acute myeloid leukemia (AML) patients with early molecular relapse. RESULTS: gmDCs elicited potent TAA-specific CTL responses in vitro, and the immunostimulatory activity of gmDC vaccination was demonstrated in rhesus monkeys. A stage 1 study established that this combinatory gmDC vaccine is safe in acute leukemia patients and yielded improved survival rate. In stage 2, we observed a complete remission rate of 83% in 12 relapsed AML patients. Overall, no grade 3 or grade 4 graft-versus-host disease incidence was detected in any of the 35 patients enrolled. CONCLUSIONS: This study, with combinatory modifications in DCs, demonstrates the safety and efficacy of SOCS1-silenced DCs in treating relapsed acute leukemia. TRIAL REGISTRATION: ClinicalTrials.gov NCT01956630. FUNDING: National Institute of Health (R01CA90427); the Key New Drug Development and Manufacturing Program of the "Twelfth Five-Year Plan" of China (2011ZX09102-001-29); and Clinical Application Research of Beijing (Z131107002213148).


Assuntos
Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Leucemia Mieloide Aguda/terapia , Recidiva Local de Neoplasia/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adenoviridae/genética , Adolescente , Adulto , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Engenharia Celular/métodos , Criança , Células Dendríticas/transplante , Feminino , Seguimentos , Vetores Genéticos/genética , Doença Enxerto-Hospedeiro/epidemiologia , Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/mortalidade , Transfusão de Linfócitos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Análise de Sobrevida , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
14.
PLoS Med ; 3(1): e11, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16381597

RESUMO

BACKGROUND: Current efforts to develop HIV vaccines that seek to stimulate immune responses have been disappointing, underscoring the inability of natural immune responses to control HIV-1 infection. Here we tested an alternative strategy to induce anti-HIV immune responses by inhibiting a host's natural immune inhibitor. METHODS AND FINDINGS: We used small interfering RNA (siRNA) to inhibit suppressor of cytokine signaling (SOCS) 1, a key negative regulator of the JAK/STAT pathway, and investigated the effect of this silencing on the ability of dendritic cells (DCs) to induce anti-HIV-1 immunity. We found that SOCS1-silenced DCs broadly induced enhanced HIV-1 envelope (Env)-specific CD8+ cytotoxic T lymphocytes and CD4+ T helper cells, as well as antibody responses, in mice. Importantly, SOCS1-silenced DCs were more resistant to HIV Env-mediated suppression and were capable of inducing memory HIV Env-specific antibody and T cell responses. SOCS1-restricted signaling, as well as production of proinflammatory cytokines such as interleukin-12 by DCs, play a critical role in regulating the anti-HIV immune response. Furthermore, the potency of HIV DNA vaccination is significantly enhanced by coimmunization with SOCS1 siRNA expressor DNA. CONCLUSIONS: This study demonstrates that SOCS1 functions as an antigen presentation attenuator to control both HIV-1-specific humoral and cellular responses. This study represents the first, to our knowledge, attempt to elicit HIV-specific T cell and antibody responses by inhibiting a host's antigen presentation attenuator, which may open a new and alternative avenue to develop effective therapeutic and prophylactic HIV vaccines.


Assuntos
Vacinas contra a AIDS/imunologia , Adjuvantes Imunológicos , Apresentação de Antígeno , Células Dendríticas/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Vacinação/métodos , Transferência Adotiva , Animais , Formação de Anticorpos , Linfócitos B/imunologia , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Infecções por HIV/prevenção & controle , Interleucina-12/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , RNA Interferente Pequeno/metabolismo , Receptores de Interleucina-12/deficiência , Receptores de Interleucina-12/genética , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/imunologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Linfócitos T/imunologia
15.
Leuk Res ; 41: 76-84, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26740053

RESUMO

Hematopoietic stem cell transplantation (HSCT) has been increasingly used as a curative treatment for acute myeloid leukemia (AML). However, relapse rates after HSCT in complete remission (CR) are reported between 30% and 70%. In addition, numerous studies suggested that secondary viral infection from a variety of viruses including Epstein-Barr virus (EBV), adenovirus (Adv), and cytomegalovirus (CMV) are among the most common causes of death post-HSCT. Currently, chimeric antigen receptor (CAR)-based T cells have been developed to treat AML in clinical studies, while virus-specific cytotoxic T cells (VST) have been proven to be able to effectively prevent or treat viral infection after HSCT. Thus it would be desirable to develop T cells with the ability of simultaneously targeting AML relapse and viral infection. In this article, we now describe the generation of VST cells that are engineered to express CAR for a specific AML cell-surface antigen CD123 (CD123-CAR-VST). Using Dendritic cells (DCs) pulsed with EBV, Adv, and CMV peptides as sources of viral antigens, we generated VST from A2 donor peripheral mononuclear cells (PBMC). VST were then transduced with retroviral vector encoding CD123-CAR to generate CD123-CAR-VST. We demonstrated that CD123-CAR-VST recognized EBV, Adv, and CMV epitopes and had HLA-restricted virus-specific cytotoxic effector function against EBV target. In addition, CD123-CAR-VST retained the specificity against CD123-positive AML cell lines such as MOLM13 and THP-1 in vitro. Thus our results suggested that CD123-CAR-VST might be a valuable candidate to simultaneously prevent or treat relapse and viral infection in AML HSCT recipients.


Assuntos
Antineoplásicos/imunologia , Antivirais/imunologia , Imunoterapia Adotiva/métodos , Subunidade alfa de Receptor de Interleucina-3/imunologia , Leucemia Mieloide Aguda/imunologia , Linfócitos T Citotóxicos/imunologia , Antígenos Virais/imunologia , Separação Celular , ELISPOT , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Imunofenotipagem , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão/imunologia , Viroses/imunologia
16.
Sci Rep ; 6: 27130, 2016 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-27255991

RESUMO

T cells expressing chimeric antigen receptors (CARs) or the infusion of bispecific T-cell engagers (BITEs) have shown antitumor activity in humans for CD19-positive malignancies. While BITEs redirect the large reservoir of resident T cells to tumors, CAR T cells rely on significant in vivo expansion to exert antitumor activity. We have shown that it is feasible to modify T cells to secrete solid tumor antigen-specific BITEs, enabling T cells to redirect resident T cells to tumor cells. To adapt this approach to CD19-positive malignancies we now generated T cells expressing secretable, CD19-specific BITEs (CD19-ENG T cells). CD19-ENG T cells recognized tumor cells in an antigen-dependent manner as judged by cytokine production and tumor killing, and redirected bystander T cells to tumor cells. Infusion of CD19-ENG T cells resulted in regression of leukemia or lymphoma in xenograft models and a survival advantage in comparison to control mice. Genetically modified T cells expressing engager molecules may present a promising addition to current CD19-targeted immunotherapies.


Assuntos
Antígenos CD19/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Anticorpos de Cadeia Única/genética , Linfócitos T/transplante , Células A549 , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Humanos , Imunoterapia , Células K562 , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Anticorpos de Cadeia Única/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Sci Rep ; 6: 23010, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26972749

RESUMO

Interleukin 6 (IL-6) has been shown to be an important regulator of cardiac interstitial fibrosis. In this study, we explored the role of interleukin-6 in the development of diabetic cardiomyopathy and the underlying mechanisms. Cardiac function of IL-6 knockout mice was significantly improved and interstitial fibrosis was apparently alleviated in comparison with wildtype (WT) diabetic mice induced by streptozotocin (STZ). Treatment with IL-6 significantly promoted the proliferation and collagen production of cultured cardiac fibroblasts (CFs). High glucose treatment increased collagen production, which were mitigated in CFs from IL-6 KO mice. Moreover, IL-6 knockout alleviated the up-regulation of TGFß1 in diabetic hearts of mice and cultured CFs treated with high glucose or IL-6. Furthermore, the expression of miR-29 reduced upon IL-6 treatment, while increased in IL-6 KO hearts. Overexpression of miR-29 blocked the pro-fibrotic effects of IL-6 on cultured CFs. In summary, deletion of IL-6 is able to mitigate myocardial fibrosis and improve cardiac function of diabetic mice. The mechanism involves the regulation of IL-6 on TGFß1 and miR-29 pathway. This study indicates the therapeutic potential of IL-6 suppression on diabetic cardiomyopathy disease associated with fibrosis.


Assuntos
Cardiomiopatias Diabéticas/genética , Interleucina-6/genética , MicroRNAs/genética , Miocárdio/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta1/genética , Animais , Animais Recém-Nascidos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Cardiomiopatias Diabéticas/induzido quimicamente , Cardiomiopatias Diabéticas/diagnóstico por imagem , Ecocardiografia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose/genética , Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Coração/fisiopatologia , Interleucina-6/sangue , Interleucina-6/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estreptozocina , Fator de Crescimento Transformador beta1/metabolismo
18.
Discov Med ; 20(110): 185-90, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26562471

RESUMO

Relapsed and refractory acute lymphoblastic leukemia (ALL) remains difficult to treat, with minimal improvement in outcomes despite advances in upfront therapy and improved survival for de novo ALL. Targeted immunotherapy for cancer represents a promising new treatment and utilizing the immune system to target and eradicate malignant cells in the body has emerged as a potent therapy. Administration of cytotoxic T cells genetically engineered to express a chimeric antigen receptor (CAR) recognizing CD19 have been shown to induce complete responses in patients with B-cell lineage ALL. So far, six clinical trials including 79 ALL patients treated with CD19-CAR T cells have been published, and the results from these trials are exciting with impressive clinical responses. Thus, CAR T cell therapy represents a potential useful tool to ALL. However, the majority of CAR cell studies have observed severe therapy associated toxicities, which needs attention. In this review, we mainly focus on CD19-CAR T cells, clinical trials for ALL as well as toxicities and challenges for CD19-CAR T therapy.


Assuntos
Antígenos CD19/metabolismo , Linfócitos B/imunologia , Linhagem da Célula/imunologia , Imunoterapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Humanos , Imunoterapia/efeitos adversos
19.
Artigo em Zh | MEDLINE | ID: mdl-12567991

RESUMO

OBJECTIVE: To construct single chain Fv (scFv) gene of anti-idiotypic monoclonal antibody NP30 of Schistosoma japonicum. METHODS: The heavy and light chain variable region genes of anti-idiotypic monoclonal antibody NP30 of Schistosoma japonicum were inserted into two corresponding sites of expression vector pTHA90, and a scFv gene was constructed with a short peptide (Gly4Ser)3 linker gene. The recombinants were determined by digesting with XhoI/SpeI, XbaI/EcoRI and XhoI/EcoRI, and then were introduced into E. coli Top10. The antigen binding activity of expressed product was detected with ELISA. RESULTS: The recombinants were determined by digesting with endonucleases and expected bands were identified. The value of expressed scFv was 3 times higher than negative control by ELISA(OD492 = 1.06). CONCLUSION: The scFv gene of anti-idiotypic monoclonal antibody NP30 of Schistosoma japonicum was successfully cloned, and the expressed scFv fragment could interact specifically with antigen NP48.


Assuntos
Anticorpos Anti-Idiotípicos/genética , Anticorpos Anti-Helmínticos/genética , Anticorpos Monoclonais/genética , Fragmentos de Imunoglobulinas/genética , Schistosoma japonicum/imunologia , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Anti-Helmínticos/biossíntese , Anticorpos Monoclonais/biossíntese , Clonagem Molecular , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Leves de Imunoglobulina/genética , Região Variável de Imunoglobulina/genética
20.
Methods Mol Biol ; 1139: 119-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24619675

RESUMO

Gene silencing with RNAi is an invaluable technique in cell biology to knock down the target gene expression. Dendritic cells (DC) are the most effective antigen-presenting cells (APC), and the efficacy of antigen presentation is tightly controlled by the stimulatory as well as inhibitory mechanisms. In recent studies, RNAi technology has been employed to silence the expression of the intrinsic inhibitors of antigen presentation in DC, improving the efficacy of DC vaccines against tumor antigens in pre-clinical studies. Here, we describe the technique of using siRNA oligonucleotides, adenovirus expressing shRNA (Ad-shRNA), or lentivirus expressing shRNA (Lv-shRNA) to knock down inhibitors of antigen presentation in both mouse and human DC.


Assuntos
Células Dendríticas/metabolismo , Técnicas de Silenciamento de Genes/métodos , Interferência de RNA , Adenoviridae/genética , Animais , Apresentação de Antígeno , Células da Medula Óssea/citologia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Interleucina-4/genética , Lentivirus/genética , Camundongos , Monócitos/citologia , RNA Interferente Pequeno/genética , Transdução Genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA