Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Transl Med ; 21(1): 483, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37468934

RESUMO

BACKGROUND: Viral therapies developed for cancer treatment have classically prioritized direct oncolytic effects over their immune activating properties. However, recent clinical insights have challenged this longstanding prioritization and have shifted the focus to more immune-based mechanisms. Through the potential utilization of novel, inherently immune-stimulating, oncotropic viruses there is a therapeutic opportunity to improve anti-tumor outcomes through virus-mediated immune activation. PV001-DV is an attenuated strain of Dengue virus (DEN-1 #45AZ5) with a favorable clinical safety profile that also maintains the potent immune stimulatory properties characterstic of Dengue virus infection. METHODS: In this study, we utilized in vitro tumor killing and immune multiplex assays to examine the anti-tumor effects of PV001-DV as a potential novel cancer immunotherapy. RESULTS: In vitro assays demonstrated that PV001-DV possesses the ability to directly kill human melanoma cells lines as well as patient melanoma tissue ex vivo. Importantly, further work demonstrated that, when patient peripheral blood mononuclear cells (PBMCs) were exposed to PV001-DV, a substantial induction in the production of apoptotic factors and immunostimulatory cytokines was detected. When tumor cells were cultured with the resulting soluble mediators from these PBMCs, rapid cell death of melanoma and breast cancer cell lines was observed. These soluble mediators also increased dengue virus binding ligands and immune checkpoint receptor, PD-L1 expression. CONCLUSIONS: The direct in vitro tumor-killing and immune-mediated tumor cytotoxicity facilitated by PV001-DV contributes support of its upcoming clinical evaluation in patients with advanced melanoma who have failed prior therapy.


Assuntos
Vírus da Dengue , Dengue , Melanoma , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Vírus da Dengue/fisiologia , Leucócitos Mononucleares , Melanoma/terapia , Células MCF-7 , Imunidade , Morte Celular , Terapia Viral Oncolítica/métodos
2.
Proc Natl Acad Sci U S A ; 117(2): 1119-1128, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31888983

RESUMO

Reprogramming the tumor microenvironment to increase immune-mediated responses is currently of intense interest. Patients with immune-infiltrated "hot" tumors demonstrate higher treatment response rates and improved survival. However, only the minority of tumors are hot, and a limited proportion of patients benefit from immunotherapies. Innovative approaches that make tumors hot can have immediate impact particularly if they repurpose drugs with additional cancer-unrelated benefits. The seasonal influenza vaccine is recommended for all persons over 6 mo without prohibitive contraindications, including most cancer patients. Here, we report that unadjuvanted seasonal influenza vaccination via intratumoral, but not intramuscular, injection converts "cold" tumors to hot, generates systemic CD8+ T cell-mediated antitumor immunity, and sensitizes resistant tumors to checkpoint blockade. Importantly, intratumoral vaccination also provides protection against subsequent active influenza virus lung infection. Surprisingly, a squalene-based adjuvanted vaccine maintains intratumoral regulatory B cells and fails to improve antitumor responses, even while protecting against active influenza virus lung infection. Adjuvant removal, B cell depletion, or IL-10 blockade recovers its antitumor effectiveness. Our findings propose that antipathogen vaccines may be utilized for both infection prevention and repurposing as a cancer immunotherapy.


Assuntos
Imunoterapia/métodos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/uso terapêutico , Injeções Intralesionais , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Linfócitos B , Fatores de Transcrição de Zíper de Leucina Básica/genética , Linfócitos T CD8-Positivos/imunologia , Humanos , Imunidade Celular , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana , Interleucina-10 , Pulmão/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Repressoras/genética , Estações do Ano , Pele , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Esqualeno/administração & dosagem , Microambiente Tumoral/efeitos dos fármacos , Vacinação
3.
Int J Mol Sci ; 25(1)2023 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-38203396

RESUMO

Breast cancer continues to have a high disease burden worldwide and presents an urgent need for novel therapeutic strategies to improve outcomes. The influenza vaccine offers a unique approach to enhance the anti-tumor immune response in patients with breast cancer. Our study explores the intratumoral use of the influenza vaccine in a triple-negative 4T1 mouse model of breast cancer. We show that the influenza vaccine attenuated tumor growth using a three-dose intratumoral regimen. More importantly, prior vaccination did not alter this improved anti-tumor response. Furthermore, we characterized the effect that the influenza vaccine has on the tumor microenvironment and the underlying mechanisms of action. We established that the vaccine facilitated favorable shifts in restructuring the tumor microenvironment. Additionally, we show that the vaccine's ability to bind sialic acid residues, which have been implicated in having oncogenic functions, emerged as a key mechanism of action. Influenza hemagglutinin demonstrated binding ability to breast cancer cells through sialic acid expression. When administered intratumorally, the influenza vaccine offers a promising therapeutic strategy for breast cancer patients by reshaping the tumor microenvironment and modestly suppressing tumor growth. Its interaction with sialic acids has implications for effective therapeutic application and future research.


Assuntos
Neoplasias da Mama , Vacinas contra Influenza , Animais , Camundongos , Humanos , Feminino , Neoplasias da Mama/terapia , Hemaglutininas , Microambiente Tumoral , Ácido N-Acetilneuramínico
4.
J Immunol ; 199(7): 2536-2546, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28814601

RESUMO

IL-15 is an essential cytokine known to promote T cell survival and activate the effector function of memory phenotype CD8 T cells. Blocking IL-15 signals also significantly impacts tissue-specific effector and memory CD8 T cell formation. In this study, we demonstrate that IL-15 influences the generation of memory CD8 T cells by first promoting their accumulation into mucosal tissues and second by sustaining expression of Bcl-6 and T-bet. We show that the mechanism for this recruitment is largely dependent on mammalian target of rapamycin and its subsequent inactivation of FoxO1. Last, we show that IL-15 complexes delivered locally to mucosal tissues without reinfection is an effective strategy to enhance establishment of tissue resident memory CD8 T cells within mucosal tissues. This study provides mechanistic insight into how IL-15 controls the generation of memory CD8 T cells and influences their trafficking and ability to take up residence within peripheral tissues.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Memória Imunológica , Interleucina-15/fisiologia , Mucosa/imunologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular , Proteína Forkhead Box O1/metabolismo , Interleucina-15/genética , Interleucina-15/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/citologia , Mucosa/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-6/genética , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Proteínas com Domínio T/genética , Subpopulações de Linfócitos T/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
5.
Nat Rev Immunol ; 6(8): 618-23, 2006 08.
Artigo em Inglês | MEDLINE | ID: mdl-16868553

RESUMO

The immune system has evolved by continuously increasing its complexity to provide the host with an advantage over infectious agents. The development of immunological memory engenders long-lasting protection and lengthens the lifespan of the host. The generation of subsets of memory T cells with distinct homing and functional properties increases our defensive capabilities. However, the developmental relationship of memory T-cell subsets is a matter of debate. In this Opinion article, in light of recent developments, we suggest that it is probable that two distinct lineages comprise the memory CD8+ T-cell population generated in response to infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD8-Positivos/citologia , Humanos , Selectina L/imunologia , Ativação Linfocitária/imunologia , Modelos Imunológicos , Subpopulações de Linfócitos T/citologia
6.
J Immunol ; 193(5): 2067-71, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25070853

RESUMO

Mucosal tissues are subject to frequent pathogen exposure and are major sites for transmission of infectious disease. CD8 T cells play a critical role in controlling mucosa-acquired infections even though their migration into mucosal tissues is tightly regulated. The mechanisms and signals that control the formation of tissue-resident memory CD8 T cells are poorly understood; however, one key regulator of memory CD8 T cell differentiation, mammalian target of rapamycin kinase, can be inhibited by rapamycin. We report that, despite enhancing the formation of memory CD8 T cells in secondary lymphoid tissues, rapamycin inhibits the formation of resident memory CD8 T cells in the intestinal and vaginal mucosa. The ability of rapamycin to block the formation of functional resident CD8 T cells in mucosal tissues protected mice from a model of CD8 T cell-mediated lethal intestinal autoimmunity. These findings demonstrate an opposing role for mammalian target of rapamycin in the formation of resident versus nonresident CD8 T cell immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade nas Mucosas/fisiologia , Memória Imunológica/fisiologia , Mucosa Intestinal/lesões , Modelos Imunológicos , Serina-Treonina Quinases TOR/imunologia , Animais , Autoimunidade/fisiologia , Linfócitos T CD8-Positivos/citologia , Feminino , Mucosa Intestinal/citologia , Camundongos , Vagina/citologia , Vagina/imunologia
7.
J Immunother Cancer ; 11(7)2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37433716

RESUMO

BACKGROUND: Immunotherapies are becoming front-line treatments for many advanced cancers, and combinations of two or more therapies are beginning to be investigated. Based on their individual antitumor capabilities, we sought to determine whether combination oncolytic virus (OV) and radiation therapy (RT) may improve cancer outcomes. METHODS: To investigate the activity of this combination therapy, we used in vitro mouse and human cancer cell lines as well as a mouse model of skin cancer. After initial results, we further included immune checkpoint blockade, whose addition constituted a triple combination immunotherapy. RESULTS: Our findings demonstrate that OV and RT reduce tumor growth via conversion of immunologically 'cold' tumors to 'hot', via a CD8+ T cell-dependent and IL-1α-dependent mechanism that is associated with increased PD-1/PD-L1 expression, and the triple combination of OV, RT, and PD-1 checkpoint inhibition impedes tumor growth and prolongs survival. Further, we describe the response of a PD-1-refractory patient with cutaneous squamous cell carcinoma who received the triple combination of OV, RT, and immune checkpoint inhibitor (ICI), and went on to experience unexpected, prolonged control and survival. He remains off-treatment and is without evidence of progression for >44 months since study entry. CONCLUSIONS: Effective systemic antitumor immune response is rarely elicited by a single therapy. In a skin cancer mouse model, we demonstrate improved outcomes with combination OV, RT, and ICI treatment, which is associated with mechanisms involving augmented CD8+ T cell infiltration and IL-1α expression. We report tumor reduction and prolonged survival of a patient with skin cancer treated with combination OV, RT, and ICI. Overall, our data provide strong rationale for combining OV, RT, and ICI for treatment of patients with ICI-refractory skin and potentially other cancers.


Assuntos
Carcinoma de Células Escamosas , Inibidores de Checkpoint Imunológico , Terapia Viral Oncolítica , Neoplasias Cutâneas , Animais , Humanos , Masculino , Camundongos , Carcinoma de Células Escamosas/terapia , Modelos Animais de Doenças , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia , Neoplasias Cutâneas/terapia , Linhagem Celular Tumoral , Terapia Combinada
8.
J Immunol Methods ; 505: 113253, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35358495

RESUMO

Understanding the dynamics of the tumor microenvironment (TME) has become vital in discovering new targets for effective immunotherapies and enhancing current treatments. However, localization and distribution of immune cells and treatment biomolecules are poorly characterized to date. In this study, a murine Luminal B mammary adenocarcinoma model received a combinatorial treatment of fluorescently labeled anti-PD-1-Cy3 and IL-15 complex-Cy5 injected interperitoneally and intratumorally, respectively. Fluorescent labeling allowed for the visualization of the distribution of IL-15 complexes and anti-PD-1, as well as their localization to immune cells in the TME and tumor-draining lymph node. Using fluorescent microscopy and light sheet microscopy of whole-clarified tumors and draining lymph nodes, the localization of IL-15 complexes was found to be distributed around the periphery of the tumor at 4 h post injection and medially located at the center of the tumor at 24 h post injection, corresponding with high densities of CD8 cells in the tumor present at 48 h and 72 h post injection. Anti-PD-1 was distributed around the perimeter of the tumor and colocalized to IL-15 in the draining lymph nodes 24 h post injection. Colocalization of IL-15 was also established with NK cells, CD8+ T cells, and macrophages. This study develops a novel method to spatiotemporally track fluorescently labeled immunotherapeutic biomolecules in vivo, with implications for optimizing and further understanding the pharmacokinetics of clinical immunotherapies.


Assuntos
Neoplasias , Microambiente Tumoral , Animais , Linfócitos T CD8-Positivos , Interleucina-15 , Linfonodos , Camundongos , Neoplasias/patologia
9.
Blood ; 114(10): 2121-30, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19617575

RESUMO

The ability of the adaptive immune system to respond rapidly and robustly upon repeated antigen exposure is known as immunologic memory, and it is thought that acquisition of memory T-cell function is an irreversible differentiation event. In this study, we report that many phenotypic and functional characteristics of antigen-specific CD8 memory T cells are lost when they are deprived of contact with dendritic cells. Under these circumstances, memory T cells reverted from G(1) to the G(0) cell-cycle state and responded to stimulation like naive T cells, as assessed by proliferation, dependence upon costimulation, and interferon-gamma production, without losing cell surface markers associated with memory. The memory state was maintained by signaling via members of the tumor necrosis factor receptor superfamily, CD27 and 4-1BB. Foxo1, a transcription factor involved in T-cell quiescence, was reduced in memory cells, and stimulation of naive CD8 cells via CD27 caused Foxo1 to be phosphorylated and emigrate from the nucleus in a phosphatidylinositol-3 kinase-dependent manner. Consistent with these results, maintenance of G(1) in vivo was compromised in antigen-specific memory T cells in vesicular stomatitis virus-infected CD27-deficient mice. Therefore, sustaining the functional phenotype of T memory cells requires active signaling and maintenance.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Comunicação Celular/imunologia , Células Dendríticas/imunologia , Memória Imunológica/imunologia , Transdução de Sinais/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Transporte Ativo do Núcleo Celular/genética , Transporte Ativo do Núcleo Celular/imunologia , Animais , Antígenos Virais/imunologia , Comunicação Celular/genética , Núcleo Celular/imunologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/imunologia , Fase G1/imunologia , Memória Imunológica/genética , Interferon gama/imunologia , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/imunologia , Fosforilação/genética , Fosforilação/imunologia , Fase de Repouso do Ciclo Celular/imunologia , Transdução de Sinais/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Estomatite Vesicular/imunologia , Vesiculovirus/imunologia
11.
Vaccines (Basel) ; 9(12)2021 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-34960243

RESUMO

Currently approximately 10 million people die each year due to cancer, and cancer is the cause of every sixth death worldwide. Tremendous efforts and progress have been made towards finding a cure for cancer. However, numerous challenges have been faced due to adverse effects of chemotherapy, radiotherapy, and alternative cancer therapies, including toxicity to non-cancerous cells, the inability of drugs to reach deep tumor tissue, and the persistent problem of increasing drug resistance in tumor cells. These challenges have increased the demand for the development of alternative approaches with greater selectivity and effectiveness against tumor cells. Cancer immunotherapy has made significant advancements towards eliminating cancer. Our understanding of cancer-directed immune responses and the mechanisms through which immune cells invade tumors have extensively helped us in the development of new therapies. Among immunotherapies, the application of bacteria and bacterial-based products has promising potential to be used as treatments that combat cancer. Bacterial targeting of tumors has been developed as a unique therapeutic option that meets the ongoing challenges of cancer treatment. In comparison with other cancer therapeutics, bacterial-based therapies have capabilities for suppressing cancer. Bacteria are known to accumulate and proliferate in the tumor microenvironment and initiate antitumor immune responses. We are currently well-informed regarding various methods by which bacteria can be manipulated by simple genetic engineering or synthetic bioengineering to induce the production of anti-cancer drugs. Further, bacterial-based cancer therapy (BBCT) can be either used as a monotherapy or in combination with other anticancer therapies for better clinical outcomes. Here, we review recent advances, current challenges, and prospects of bacteria and bacterial products in the development of BBCTs.

12.
Adv Exp Med Biol ; 684: 69-78, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20795541

RESUMO

Immunological memory is considered the hallmark of adaptive, or acquired, immunity. That ability of our immune system to recognize and respond to those pathogens we have encountered before not only typifies acquired immunity but has provided the basis for the most notable of medical interventions: vaccination. Yet, as much as we now know about this process, we are still on the cusp of fully understanding how memory T cells develop, how they are maintained and the importance of memory T-cell heterogeneity. In this review we will primarily focus on our understanding of CD8 T-cell memory generated during acute infections and how precursor frequency influences their development and functional attributes.


Assuntos
Memória Imunológica/imunologia , Células Precursoras de Linfócitos T/fisiologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/fisiologia , Animais , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular , Humanos , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Linfopoese , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transgenes
13.
Cell Rep ; 17(4): 957-965, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760326

RESUMO

In light of increased cancer prevalence and cancer-specific deaths in patients with infections, we investigated whether infections alter anti-tumor immune responses. We report that acute influenza infection of the lung promotes distal melanoma growth in the dermis and leads to accelerated cancer-specific host death. Furthermore, we show that during influenza infection, anti-melanoma CD8+ T cells are shunted from the tumor to the infection site, where they express high levels of the inhibitory receptor programmed cell death protein 1 (PD-1). Immunotherapy to block PD-1 reverses this loss of anti-tumor CD8+ T cells from the tumor and decreases infection-induced tumor growth. Our findings show that acute non-oncogenic infection can promote cancer growth, raising concerns regarding acute viral illness sequelae. They also suggest an unexpected role for PD-1 blockade in cancer immunotherapy and provide insight into the immune response when faced with concomitant challenges.


Assuntos
Melanoma/imunologia , Melanoma/patologia , Oncogenes , Infecções por Orthomyxoviridae/patologia , Doença Aguda , Animais , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Pulmão/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/metabolismo
14.
Adv Exp Med Biol ; 512: 141-6, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12405198

RESUMO

Our results indicate that a substantial proportion of the antimicrobial CD8 and CD4 T cell response is focused in non-lymphoid tissues. This finding makes teleological sense since maximum protection against infection is better served by the widespread presence of effector and memory cells. In the case of CD8 T cells, it appears that irrespective of the site at which initial activation of naive cells occurs, the end result is production of effector cells with broad migratory capabilities. Memory T cells perhaps have more restricted migratory abilities as compared to effector cells, although this needs to be tested definitively. Our results suggest that memory CD8 T cells in the intestinal LP may not be part of the recirculating pool of memory cells, though our data does not preclude the possibility that migrants from outside the mucosa contribute to the LP memory pool. Our data also demonstrates that CD8 memory T cells in non-lymphoid tissue exhibit heightened effector function as compared to their splenic counterparts. Whether these findings indicate the existence of distinct lineages of memory cells remains to be seen. The functional abilities of migrating memory cells could be modulated by the migration process and/or by the environmental milieu of a particular tissue. Although the development of CD8 memory T cells is a complex process requiring multiple signalling pathways, we identified IL-7 as an important player in memory generation. Much further work is needed to decipher the cellular and molecular mechanisms of memory induction as well as to learn the in vivo functional significance of memory cell subsets.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Animais , Linfócitos T CD8-Positivos/fisiologia , Quimiotaxia de Leucócito , Humanos , Infecções/imunologia , Ativação Linfocitária
15.
Oncoimmunology ; 2(2): e22731, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23525138

RESUMO

The major objectives of tumor vaccination are to induce the regression of established tumors and to favor the establishment of long-lasting tumor-specific immunity, capable of protecting the host from relapse. Immunotherapeutic strategies such as the administration of tumor-associated antigenic peptides offer one means to boost preexisting antitumor CD8+ T cell immunity. Our recent work reveals that established breast tumors are rejected and tumor recurrence prevented when low-dose irradiation is combined with the adoptive transfer of Mammaglobin A epitope-specific CD8+ T cells.

16.
PLoS One ; 7(7): e41240, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22911764

RESUMO

Adoptive T cell therapy has proven to be beneficial in a number of tumor systems by targeting the relevant tumor antigen. The tumor antigen targeted in our model is Mammaglobin-A, expressed by approximately 80% of human breast tumors. Here we evaluated the use of adoptively transferred Mammaglobin-A specific CD8 T cells in combination with low dose irradiation to induce breast tumor rejection and prevent relapse. We show Mammaglobin-A specific CD8 T cells generated by DNA vaccination with all epitopes (Mammaglobin-A2.1, A2.2, A2.4 and A2.6) and full-length DNA in vivo resulted in heterogeneous T cell populations consisting of both effector and central memory CD8 T cell subsets. Adoptive transfer of spleen cells from all Mammaglobin-A2 immunized mice into tumor-bearing SCID/beige mice induced tumor regression but this anti-tumor response was not sustained long-term. Additionally, we demonstrate that only the adoptive transfer of Mammaglobin-A2 specific CD8 T cells in combination with a single low dose of irradiation prevents tumors from recurring. More importantly we show that this single dose of irradiation results in the down regulation of the macrophage scavenger receptor 1 on dendritic cells within the tumor and reduces lipid uptake by tumor resident dendritic cells potentially enabling the dendritic cells to present tumor antigen more efficiently and aid in tumor clearance. These data reveal the potential for adoptive transfer combined with a single low dose of total body irradiation as a suitable therapy for the treatment of established breast tumors and the prevention of tumor recurrence.


Assuntos
Transferência Adotiva , Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Mamoglobina A/imunologia , Irradiação Corporal Total , Animais , Neoplasias da Mama/mortalidade , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Epitopos de Linfócito T/genética , Feminino , Humanos , Memória Imunológica , Interferon gama/biossíntese , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Selectina L/metabolismo , Metabolismo dos Lipídeos , Mamoglobina A/química , Camundongos , Camundongos SCID , Camundongos Transgênicos , Receptores Depuradores Classe A/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Vacinas de DNA/imunologia
17.
Nat Med ; 18(3): 422-8, 2012 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-22366950

RESUMO

CD4-unhelped CD8(+) T cells are functionally defective T cells primed in the absence of CD4(+) T cell help. Given the co-stimulatory role of natural-killer group 2, member D protein (NKG2D) on CD8(+) T cells, we investigated its ability to rescue these immunologically impotent cells. We demonstrate that augmented co-stimulation through NKG2D during priming paradoxically rescues memory, but not effector, CD8(+) T cell responses. NKG2D-mediated rescue is characterized by reversal of elevated transcription factor T-box expressed in T cells (T-bet) expression and recovery of interleukin-2 and interferon-γ production and cytolytic responses. Rescue is abrogated in CD8(+) T cells lacking NKG2D. Augmented co-stimulation through NKG2D confers a high rate of survival to mice lacking CD4(+) T cells in a CD4-dependent influenza model and rescues HIV-specific CD8(+) T cell responses from CD4-deficient HIV-positive donors. These findings demonstrate that augmented co-stimulation through NKG2D is effective in rescuing CD4-unhelped CD8(+) T cells from their pathophysiological fate and may provide therapeutic benefits.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/imunologia , Influenza Humana/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Citotoxicidade Imunológica , Modelos Animais de Doenças , Expressão Gênica , HIV-1/imunologia , Humanos , Imunidade Celular , Influenza Humana/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-2/imunologia , Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas com Domínio T/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Vacinas de DNA/imunologia
18.
PLoS One ; 6(2): e16529, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21364749

RESUMO

Memory CD4 T cells play a vital role in protection against re-infection by pathogens as diverse as helminthes or influenza viruses. Inducible costimulator (ICOS) is highly expressed on memory CD4 T cells and has been shown to augment proliferation and survival of activated CD4 T cells. However, the role of ICOS costimulation on the development and maintenance of memory CD4 T cells remains controversial. Herein, we describe a significant defect in the number of effector memory (EM) phenotype cells in ICOS(-/-) and ICOSL(-/-) mice that becomes progressively more dramatic as the mice age. This decrease was not due to a defect in the homeostatic proliferation of EM phenotype CD4 T cells in ICOS(-/-) or ICOSL(-/-) mice. To determine whether ICOS regulated the development or survival of EM CD4 T cells, we utilized an adoptive transfer model. We found no defect in development of EM CD4 T cells, but long-term survival of ICOS(-/-) EM CD4 T cells was significantly compromised compared to wild-type cells. The defect in survival was specific to EM cells as the central memory (CM) ICOS(-/-) CD4 T cells persisted as well as wild type cells. To determine the physiological consequences of a specific defect in EM CD4 T cells, wild-type and ICOS(-/-) mice were infected with influenza virus. ICOS(-/-) mice developed significantly fewer influenza-specific EM CD4 T cells and were more susceptible to re-infection than wild-type mice. Collectively, our findings demonstrate a role for ICOS costimulation in the maintenance of EM but not CM CD4 T cells.


Assuntos
Antígenos de Diferenciação de Linfócitos T/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/fisiologia , Memória Imunológica/genética , Proteínas/fisiologia , Animais , Antígenos de Diferenciação de Linfócitos T/genética , Linfócitos T CD4-Positivos/metabolismo , Sobrevivência Celular/genética , Citoproteção/genética , Citoproteção/imunologia , Predisposição Genética para Doença , Memória Imunológica/fisiologia , Ligante Coestimulador de Linfócitos T Induzíveis , Proteína Coestimuladora de Linfócitos T Induzíveis , Vírus da Influenza A/fisiologia , Interleucina-4/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Fenótipo , Proteínas/genética
20.
J Immunol ; 179(1): 36-40, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17579018

RESUMO

Memory CD8 T cells, essential for defense against intracellular pathogens, are heterogeneous with respect to phenotype and function. Constitutively lytic effector memory cells primarily reside in nonlymphoid tissues, whereas secondary lymphoid tissues contain functionally quiescent central memory cells. However, the mechanism by which functionally distinct memory populations are maintained is unknown. In this study, we show that resting CD8 memory cells modified their functional abilities upon entry into nonlymphoid tissues, as exemplified by the induction of granzyme B and lytic activity. Contemporaneously, the costimulator CD27 was down-regulated. These findings hold important implications for memory cell lineage development and tissue-specific immunity.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Movimento Celular/imunologia , Memória Imunológica , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/transplante , Citotoxicidade Imunológica , Regulação para Baixo/imunologia , Granzimas/biossíntese , Imunofenotipagem , Fígado/citologia , Fígado/imunologia , Pulmão/citologia , Pulmão/imunologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/biossíntese , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA