Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Ther ; 32(4): 1000-1015, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38414243

RESUMO

Adoptive cell therapy (ACT) using T cells expressing chimeric antigen receptors (CARs) is an area of intense investigation in the treatment of malignancies and chronic viral infections. One of the limitations of ACT-based CAR therapy is the lack of in vivo persistence and maintenance of optimal cell function. Therefore, alternative strategies that increase the function and maintenance of CAR-expressing T cells are needed. In our studies using the humanized bone marrow/liver/thymus (BLT) mouse model and nonhuman primate (NHP) model of HIV infection, we evaluated two CAR-based gene therapy approaches. In the ACT approach, we used cytokine enhancement and preconditioning to generate greater persistence of anti-HIV CAR+ T cells. We observed limited persistence and expansion of anti-HIV CAR T cells, which led to minimal control of the virus. In our stem cell-based approach, we modified hematopoietic stem/progenitor cells (HSPCs) with anti-HIV CAR to generate anti-HIV CAR T cells in vivo. We observed CAR-expressing T cell expansion, which led to better plasma viral load suppression. HSPC-derived CAR cells in infected NHPs showed superior trafficking and persistence in multiple tissues. Our results suggest that a stem cell-based CAR T cell approach may be superior in generating long-term persistence and functional antiviral responses against HIV infection.


Assuntos
Infecções por HIV , HIV-1 , Receptores de Antígenos Quiméricos , Camundongos , Animais , Linfócitos T , Receptores de Antígenos Quiméricos/genética , Células-Tronco Hematopoéticas , Imunoterapia Adotiva
2.
Mol Ther Methods Clin Dev ; 30: 276-287, 2023 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-37575091

RESUMO

Hematopoietic stem cell gene therapy has been successfully used for a number of genetic diseases and is also being explored for HIV. However, toxicity of the conditioning regimens has been a major concern. Here we compared current conditioning approaches in a clinically relevant nonhuman primate model. We first customized various aspects of the therapeutic approach, including mobilization and cell collection protocols, conditioning regimens that support engraftment with minimal collateral damage, and cell manufacturing and infusing schema that reflect and build on current clinical approaches. Through a series of iterative in vivo experiments in two macaque species, we show that busulfan conditioning significantly spares lymphocytes and maintains a superior immune response to mucosal challenge with simian/human immunodeficiency virus, compared to total body irradiation and melphalan regimens. Comparative mobilization experiments demonstrate higher cell yield relative to our historical standard, primed bone marrow and engraftment of CRISPR-edited hematopoietic stem and progenitor cells (HSPCs) after busulfan conditioning. Our findings establish a detailed workflow for preclinical HSPC gene therapy studies in the nonhuman primate model, which in turn will support testing of novel conditioning regimens and more advanced HSPC gene editing techniques tailored to any disease of interest.

3.
JCI Insight ; 7(22)2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36509289

RESUMO

A hallmark of HIV-1 infection is chronic inflammation, even in patients treated with antiretroviral therapy (ART). Chronic inflammation drives HIV-1 pathogenesis, leading to loss of CD4+ T cells and exhaustion of antiviral immunity. Therefore, strategies to safely reduce systematic inflammation are needed to halt disease progression and restore defective immune responses. Autophagy is a cellular mechanism for disposal of damaged organelles and elimination of intracellular pathogens. Autophagy is pivotal for energy homeostasis and plays critical roles in regulating immunity. However, how it regulates inflammation and antiviral T cell responses during HIV infection is unclear. Here, we demonstrate that autophagy is directly linked to IFN-I signaling, which is a key driver of immune activation and T cell exhaustion during chronic HIV infection. Impairment of autophagy leads to spontaneous IFN-I signaling, and autophagy induction reduces IFN-I signaling in monocytic cells. Importantly, in HIV-1-infected humanized mice, autophagy inducer rapamycin treatment significantly reduced persistent IFN-I-mediated inflammation and improved antiviral T cell responses. Cotreatment of rapamycin with ART led to significantly reduced viral rebound after ART withdrawal. Taken together, our data suggest that therapeutically targeting autophagy is a promising approach to treat persistent inflammation and improve immune control of HIV replication.


Assuntos
Infecções por HIV , HIV-1 , Interferon Tipo I , Camundongos , Animais , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Autofagia
4.
J Vis Exp ; (188)2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-36282697

RESUMO

The human immunodeficiency virus (HIV-1) pandemic continues to spread unabated worldwide, and currently, there is no vaccine available against HIV. Although combinational antiretroviral therapy (cART) has been successful in suppressing viral replication, it cannot completely eradicate the reservoir from HIV-infected individuals. A safe and effective cure strategy for HIV infection will require multipronged methods, and therefore the advancements of animal models for HIV-1 infection are pivotal for the development of HIV cure research. Humanized mice recapitulate key features of HIV-1 infection. The humanized mouse model can be infected by HIV-1 and viral replication can be controlled with cART regimens. Moreover, cART interruption results in a prompt viral rebound in humanized mice. However, administration of cART to the animal can be ineffective, difficult, or toxic, and many clinically relevant cART regimens are unable to be optimally utilized. Along with being potentially unsafe for researchers, administration of cART by a commonly used intensive daily injection procedure induces stress by physical restraint of the animal. The novel oral cART method to treat HIV-1 infected humanized mice described in this article resulted in suppression of viremia below the detection level, increased rate of CD4+ restoration, and improved overall health in HIV-1 infected humanized mice.


Assuntos
Infecções por HIV , HIV-1 , Camundongos , Humanos , Animais , Infecções por HIV/tratamento farmacológico , Antirretrovirais/uso terapêutico , Antirretrovirais/farmacologia , Viremia/tratamento farmacológico , Replicação Viral , Carga Viral , Linfócitos T CD4-Positivos
5.
PLoS Pathog ; 17(8): e1009895, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34460861

RESUMO

[This corrects the article DOI: 10.1371/journal.ppat.1009404.].

6.
PLoS Pathog ; 17(4): e1009404, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33793675

RESUMO

Due to the durability and persistence of reservoirs of HIV-1-infected cells, combination antiretroviral therapy (ART) is insufficient in eradicating infection. Achieving HIV-1 cure or sustained remission without ART treatment will require the enhanced and persistent effective antiviral immune responses. Chimeric Antigen Receptor (CAR) T-cells have emerged as a powerful immunotherapy and show promise in treating HIV-1 infection. Persistence, trafficking, and maintenance of function remain to be a challenge in many of these approaches, which are based on peripheral T cell modification. To overcome many of these issues, we have previously demonstrated successful long-term engraftment and production of anti-HIV CAR T cells in modified hematopoietic stem cells (HSCs) in vivo. Here we report the development and in vivo testing of second generation CD4-based CARs (CD4CAR) against HIV-1 infection using a HSCs-based approach. We found that a modified, truncated CD4-based CAR (D1D2CAR) allows better CAR-T cell differentiation from gene modified HSCs, and maintains similar CTL activity as compared to the full length CD4-based CAR. In addition, D1D2CAR does not mediate HIV infection or stimulation mediated by IL-16, suggesting lower risk of off-target effects. Interestingly, stimulatory domains of 4-1BB but not CD28 allowed successful hematopoietic differentiation and improved anti-viral function of CAR T cells from CAR modified HSCs. Addition of 4-1BB to CD4 based CARs led to faster suppression of viremia during early untreated HIV-1 infection. D1D2CAR 4-1BB mice had faster viral suppression in combination with ART and better persistence of CAR T cells during ART. In summary, our data indicate that the D1D2CAR-41BB is a superior CAR, showing better HSC differentiation, viral suppression and persistence, and less deleterious functions compared to the original CD4CAR, and should continue to be pursued as a candidate for clinical study.


Assuntos
Infecções por HIV/virologia , Células-Tronco Hematopoéticas/citologia , Ativação Linfocitária , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Animais , Infecções por HIV/imunologia , HIV-1/imunologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/uso terapêutico
7.
Artigo em Inglês | MEDLINE | ID: mdl-32903563

RESUMO

The HIV reservoir remains to be a difficult barrier to overcome in order to achieve a therapeutic cure for HIV. Several strategies have been developed to purge the reservoir, including the "kick and kill" approach, which is based on the notion that reactivating the latent reservoir will allow subsequent elimination by the host anti-HIV immune cells. However, clinical trials testing certain classes of latency reactivating agents (LRAs) have so far revealed the minimal impact on reducing the viral reservoir. A robust immune response to reactivated HIV expressing cells is critical for this strategy to work. A current focus to enhance anti-HIV immunity is through the use of chimeric antigen receptors (CARs). Currently, HIV-specific CARs are being applied to peripheral T cells, NK cells, and stem cells to boost recognition and killing of HIV infected cells. In this review, we summarize current developments in engineering HIV directed CAR-expressing cells to facilitate HIV elimination. We also summarize current LRAs that enhance the "kick" strategy and how new generation and combinations of LRAs with HIV specific CAR T cell therapies could provide an optimal strategy to target the viral reservoir and achieve HIV clearance from the body.


Assuntos
Infecções por HIV , HIV-1 , Linfócitos T CD4-Positivos , Infecções por HIV/terapia , Humanos , Células Matadoras Naturais , Latência Viral
8.
Front Immunol ; 9: 746, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29755454

RESUMO

HIV and cancer remain prevailing sources of morbidity and mortality worldwide. There are current efforts to discover novel therapeutic strategies for the treatment or cure of these diseases. Humanized mouse models provide the investigative tool to study the interaction between HIV or cancer and the human immune system in vivo. These humanized models consist of immunodeficient mice transplanted with human cells, tissues, or hematopoietic stem cells that result in reconstitution with a nearly full human immune system. In this review, we discuss preclinical studies evaluating therapeutic approaches in stem cell-based gene therapy and T cell-based immunotherapies for HIV and cancer using a humanized mouse model and some recent advances in using checkpoint inhibitors to improve antiviral or antitumor responses.


Assuntos
Modelos Animais de Doenças , Terapia Genética , Infecções por HIV/terapia , Imunoterapia , Neoplasias/terapia , Animais , Transplante de Células-Tronco Hematopoéticas , Humanos , Interferon Tipo I/imunologia , Camundongos , Receptor de Morte Celular Programada 1/imunologia
10.
PLoS Pathog ; 13(12): e1006753, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29284044

RESUMO

Chimeric Antigen Receptor (CAR) T-cells have emerged as a powerful immunotherapy for various forms of cancer and show promise in treating HIV-1 infection. However, significant limitations are persistence and whether peripheral T cell-based products can respond to malignant or infected cells that may reappear months or years after treatment remains unclear. Hematopoietic Stem/Progenitor Cells (HSPCs) are capable of long-term engraftment and have the potential to overcome these limitations. Here, we report the use of a protective CD4 chimeric antigen receptor (C46CD4CAR) to redirect HSPC-derived T-cells against simian/human immunodeficiency virus (SHIV) infection in pigtail macaques. CAR-containing cells persisted for more than 2 years without any measurable toxicity and were capable of multilineage engraftment. Combination antiretroviral therapy (cART) treatment followed by cART withdrawal resulted in lower viral rebound in CAR animals relative to controls, and demonstrated an immune memory-like response. We found CAR-expressing cells in multiple lymphoid tissues, decreased tissue-associated SHIV RNA levels, and substantially higher CD4/CD8 ratios in the gut as compared to controls. These results show that HSPC-derived CAR T-cells are capable of long-term engraftment and immune surveillance. This study demonstrates for the first time the safety and feasibility of HSPC-based CAR therapy in a large animal preclinical model.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , Infecções por HIV/terapia , Células-Tronco Hematopoéticas/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Animais , Linfócitos T CD4-Positivos/transplante , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Modelos Animais de Doenças , Terapia Genética/métodos , Infecções por HIV/virologia , Transplante de Células-Tronco Hematopoéticas/métodos , Imunoterapia/métodos , Macaca nemestrina , Masculino , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
11.
Curr Biol ; 27(17): 2630-2639.e6, 2017 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-28823678

RESUMO

Many chemosensory stimuli evoke innate behavioral responses that can be either appetitive or aversive, depending on an animal's age, prior experience, nutritional status, and environment [1-9]. However, the circuit mechanisms that enable these valence changes are poorly understood. Here, we show that Caenorhabditis elegans can alternate between attractive or aversive responses to carbon dioxide (CO2), depending on its recently experienced CO2 environment. Both responses are mediated by a single pathway of interneurons. The CO2-evoked activity of these interneurons is subject to extreme experience-dependent modulation, enabling them to drive opposite behavioral responses to CO2. Other interneurons in the circuit regulate behavioral sensitivity to CO2 independent of valence. A combinatorial code of neuropeptides acts on the circuit to regulate both valence and sensitivity. Chemosensory valence-encoding interneurons exist across phyla, and valence is typically determined by whether appetitive or aversive interneuron populations are activated. Our results reveal an alternative mechanism of valence determination in which the same interneurons contribute to both attractive and aversive responses through modulation of sensory neuron to interneuron synapses. This circuit design represents a previously unrecognized mechanism for generating rapid changes in innate chemosensory valence.


Assuntos
Caenorhabditis elegans/fisiologia , Dióxido de Carbono/fisiologia , Quimiotaxia , Interneurônios/fisiologia , Animais
12.
Transl Res ; 187: 83-92, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28755872

RESUMO

HIV infection continues to be a life-long chronic disease in spite of the success of antiretroviral therapy (ART) in controlling viral replication and preventing disease progression. However, because of the high cost of treatment, severe side effects, and inefficiency in curing the disease with ART, there is a call for alternative therapies that will provide a functional cure for HIV. Cytotoxic T lymphocytes (CTLs) are vital in the control and clearance of viral infections and therefore immune-based therapies have attempted to engineer HIV-specific CTLs that would be able to clear the infection from the body. The development of chimeric antigen receptors (CARs) provides an opportunity to engineer superior HIV-specific CTLs that will be independent of the major histocompatibility complex for target recognition. A CD4-based CAR has been previously tested in clinical trials to test the antiviral efficacy of peripheral T cells armed with this CD4-based CAR. The results from these clinical trials showed the safety and feasibility of CAR T cell therapy for HIV infection; however, minimal antiviral efficacy was seen. In this review, we will discuss the various strategies being developed to enhance the therapeutic potency of anti-HIV CARs with the goal of generating superior antiviral responses that will lead to life-long HIV immunity and clearance of the virus from the body.


Assuntos
Infecções por HIV/terapia , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Antígenos CD4 , Regulação da Expressão Gênica , Humanos , Imunoterapia Adotiva , Receptores de Antígenos/genética
13.
Immunotherapy ; 9(5): 401-410, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28357916

RESUMO

Despite the success of combination antiretroviral therapy (cART) for suppressing HIV and improving patients' quality of life, HIV persists in cART-treated patients and remains an incurable disease. Financial burdens and health consequences of lifelong cART treatment call for novel HIV therapies that result in a permanent cure. Cellular immunity is central in controlling HIV replication. However, HIV adopts numerous strategies to evade immune surveillance. Engineered immunity via genetic manipulation could offer a functional cure by generating cells that have enhanced antiviral activity and are resistant to HIV infection. Recently, encouraging reports from several human clinical trials using an anti-CD19 chimeric antigen receptor (CAR) modified T-cell therapy for treating B-cell malignancies have provided valuable insights and generated remarkable enthusiasm in engineered T-cell therapy. In this review, we discuss the development of HIV-specific chimeric antigen receptors and the use of stem cell based therapies to generate lifelong anti-HIV immunity.


Assuntos
Infecções por HIV/terapia , HIV/fisiologia , Imunidade Celular , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Células-Tronco/fisiologia , Animais , Antígenos CD19/imunologia , Linfócitos B/imunologia , Infecções por HIV/genética , Humanos , Evasão da Resposta Imune , Receptores de Antígenos/genética , Linfócitos T/imunologia
14.
Infect Immun ; 83(3): 1130-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25561714

RESUMO

Entomopathogenic nematodes (EPNs) in the genera Heterorhabditis and Steinernema are lethal parasites of insects that are of interest as models for understanding parasite-host interactions and as biocontrol agents for insect pests. EPNs harbor a bacterial endosymbiont in their gut that assists in insect killing. EPNs are capable of infecting and killing a wide range of insects, yet how the nematodes and their bacterial endosymbionts interact with the insect immune system is poorly understood. Here, we develop a versatile model system for understanding the insect immune response to parasitic nematode infection that consists of seven species of EPNs as model parasites and five species of Drosophila fruit flies as model hosts. We show that the EPN Steinernema carpocapsae, which is widely used for insect control, is capable of infecting and killing D. melanogaster larvae. S. carpocapsae is associated with the bacterium Xenorhabdus nematophila, and we show that X. nematophila induces expression of a subset of antimicrobial peptide genes and suppresses the melanization response to the nematode. We further show that EPNs vary in their virulence toward D. melanogaster and that Drosophila species vary in their susceptibilities to EPN infection. Differences in virulence among different EPN-host combinations result from differences in both rates of infection and rates of postinfection survival. Our results establish a powerful model system for understanding mechanisms of host-parasite interactions and the insect immune response to parasitic nematode infection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/biossíntese , Drosophila melanogaster/imunologia , Drosophila/imunologia , Nematoides/patogenicidade , Animais , Drosophila/parasitologia , Drosophila melanogaster/parasitologia , Especificidade de Hospedeiro , Interações Hospedeiro-Parasita , Larva/crescimento & desenvolvimento , Larva/metabolismo , Larva/microbiologia , Larva/patogenicidade , Longevidade/imunologia , Nematoides/crescimento & desenvolvimento , Nematoides/metabolismo , Nematoides/microbiologia , Especificidade da Espécie , Simbiose , Fatores de Tempo , Virulência , Xenorhabdus
15.
J Neurosci ; 33(23): 9675-83, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23739964

RESUMO

Sensory behaviors are often flexible, allowing animals to generate context-appropriate responses to changing environmental conditions. To investigate the neural basis of behavioral flexibility, we examined the regulation of carbon dioxide (CO2) response in the nematode Caenorhabditis elegans. CO2 is a critical sensory cue for many animals, mediating responses to food, conspecifics, predators, and hosts (Scott, 2011; Buehlmann et al., 2012; Chaisson and Hallem, 2012). In C. elegans, CO2 response is regulated by the polymorphic neuropeptide receptor NPR-1: animals with the N2 allele of npr-1 avoid CO2, whereas animals with the Hawaiian (HW) allele or an npr-1 loss-of-function (lf) mutation appear virtually insensitive to CO2 (Hallem and Sternberg, 2008; McGrath et al., 2009). Here we show that ablating the oxygen (O2)-sensing URX neurons in npr-1(lf) mutants restores CO2 avoidance, suggesting that NPR-1 enables CO2 avoidance by inhibiting URX neurons. URX was previously shown to be activated by increases in ambient O2 (Persson et al., 2009; Zimmer et al., 2009; Busch et al., 2012). We find that, in npr-1(lf) mutants, O2-induced activation of URX inhibits CO2 avoidance. Moreover, both HW and npr-1(lf) animals avoid CO2 under low O2 conditions, when URX is inactive. Our results demonstrate that CO2 response is determined by the activity of O2-sensing neurons and suggest that O2-dependent regulation of CO2 avoidance is likely to be an ecologically relevant mechanism by which nematodes navigate gas gradients.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Dióxido de Carbono/metabolismo , Quimiotaxia/fisiologia , Neurônios/metabolismo , Oxigênio/metabolismo , Animais , Animais Geneticamente Modificados , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Caenorhabditis elegans/efeitos dos fármacos , Dióxido de Carbono/farmacologia , Quimiotaxia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Oxigênio/farmacologia
16.
Clin Cancer Res ; 15(19): 6167-76, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19789304

RESUMO

PURPOSE: The goal of this study was to investigate the therapeutic potential of a novel immunotherapy strategy resulting in immunity to localized or metastatic human papillomavirus 16-transformed murine tumors. EXPERIMENTAL DESIGN: Animals bearing E7-expressing tumors were coimmunized by lymph node injection with E7 49-57 antigen and TLR3-ligand (synthetic dsRNA). Immune responses were measured by flow cytometry and antitumor efficacy was evaluated by tumor size and survival. In situ cytotoxicity assays and identification of tumor-infiltrating lymphocytes and T regulatory cells were used to assess the mechanisms of treatment resistance in bulky disease. Chemotherapy with cyclophosphamide was explored to augment immunotherapy in late-stage disease. RESULTS: In therapeutic and prophylactic settings, immunization resulted in a considerable expansion of E7 49-57 antigen-specific T lymphocytes in the range of 1/10 CD8(+) T cells. The resulting immunity was effective in suppressing disease progression and mortality in a pulmonary metastatic disease model. Therapeutic immunization resulted in control of isolated tumors up to a certain volume, and correlated with antitumor immune responses measured in blood. In situ analysis showed that within bulky tumors, T-cell function was affected by negative regulatory mechanisms linked to an increase in T regulatory cells and could be overcome by cyclophosphamide treatment in conjunction with immunization. CONCLUSIONS: This study highlights a novel cancer immunotherapy platform with potential for translatability to the clinic and suggests its potential usefulness for controlling metastatic disease, solid tumors of limited size, or larger tumors when combined with cytotoxic agents that reduce the number of tumor-infiltrating T regulatory cells.


Assuntos
Papillomavirus Humano 16/fisiologia , Imunidade Celular/fisiologia , Imunoterapia/métodos , Linfonodos/imunologia , Neoplasias/patologia , Neoplasias/terapia , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Transformação Celular Viral/imunologia , Terapia Combinada , Citotoxinas/administração & dosagem , Feminino , Papillomavirus Humano 16/imunologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neoplasias/imunologia , Neoplasias/prevenção & controle , Proteínas E7 de Papillomavirus/metabolismo , Carga Tumoral/imunologia
17.
Immunol Lett ; 127(1): 60-7, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19751765

RESUMO

Elevated Programmed Death-1 (PD-1) expression can inhibit T cell activity and is a potential barrier to achieving persisting and optimal immunity via therapeutic vaccination. Using a direct lymph node-targeted vaccination procedure that enabled uncoupling of synthetic peptide (signal 1, TCR-mediated) and adjuvant (signal 2, non-TCR-mediated), we evaluated the impact of varied doses of Toll-like receptor (TLR)-9 ligand CpG oligodeoxynucleotide (ODN) adjuvant on epitope-specific CD8(+) T cell-associated PD-1 expression. Peptide vaccination without adjuvant yielded CD8(+) T cells with significantly elevated PD-1 expression. This conferred impaired function ex vivo, but was reversible by antibody-mediated PD-1 blockade. By comparison, peptide vaccination with escalating doses of CpG ODN adjuvant yielded higher magnitudes of CD8(+) T cells with progressively lower PD-1 expression and greater ex vivo function. CpG ODN adjuvant in context of titrated peptide doses for vaccination yielded the lowest overall PD-1 expression levels, demonstrating that fine-tuning both TCR-independent (adjuvant dose) and -dependent (antigen dose) stimuli can synergize to co-regulate PD-1 expression on epitope-specific CD8(+) T cells. These data hint at strategies to elicit PD-1(low) CD8(+) T cells using TLR-9 ligand adjuvants, and also shed light on the PD-1-regulated homeostasis of CD8(+) T cells.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Regulação da Expressão Gênica , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Imunológicos/biossíntese , Receptor Toll-Like 9/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , DNA/administração & dosagem , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Antígenos HLA-A/genética , Antígeno HLA-A2 , Imunização , Ligantes , Antígeno MART-1 , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/imunologia , Oligodesoxirribonucleotídeos , Fragmentos de Peptídeos/imunologia , Receptor de Morte Celular Programada 1 , Receptores de Antígenos de Linfócitos T/imunologia , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Receptor Toll-Like 9/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA