Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
EMBO Rep ; 21(6): e50162, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32314873

RESUMO

The latency associated with bone metastasis emergence in castrate-resistant prostate cancer is attributed to dormancy, a state in which cancer cells persist prior to overt lesion formation. Using single-cell transcriptomics and ex vivo profiling, we have uncovered the critical role of tumor-intrinsic immune signaling in the retention of cancer cell dormancy. We demonstrate that loss of tumor-intrinsic type I IFN occurs in proliferating prostate cancer cells in bone. This loss suppresses tumor immunogenicity and therapeutic response and promotes bone cell activation to drive cancer progression. Restoration of tumor-intrinsic IFN signaling by HDAC inhibition increased tumor cell visibility, promoted long-term antitumor immunity, and blocked cancer growth in bone. Key findings were validated in patients, including loss of tumor-intrinsic IFN signaling and immunogenicity in bone metastases compared to primary tumors. Data herein provide a rationale as to why current immunotherapeutics fail in bone-metastatic prostate cancer, and provide a new therapeutic strategy to overcome the inefficacy of immune-based therapies in solid cancers.


Assuntos
Neoplasias Ósseas , Neoplasias da Próstata , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Humanos , Interferons , Masculino , Neoplasias da Próstata/genética , Transdução de Sinais
2.
Cancer Immunol Immunother ; 70(8): 2125-2138, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33449132

RESUMO

Competent type I IFN signaling is the lynchpin of most immune surveillance mechanisms and has recently proven critical to the efficacy of several anticancer agents. Expression of the type I IFN receptor, IFNAR, underpins type I IFN responsiveness in all cells and facilitates the activation and cytotoxic potential of lymphocytes, while loss of IFNAR on lymphocytes has previously been associated with tumor progression and poor patient survival. This study underscores the importance of intact type I IFN signaling to NK cells in the regulation of tumorigenesis and metastasis, whereby ablation of NK cell IFNAR1 impairs antitumor activity and tumor clearance. Using a preclinical model of triple negative breast cancer, we identified that intact IFNAR on NK cells is required for an effective response to type I IFN-inducing immunotherapeutics that may be mediated by pathways associated with NK cell degranulation. Taken together, these data provide a rationale for considering the IFNAR status on NK cells when devising therapeutic strategies aimed at inducing systemic type I IFN signaling in breast cancer.


Assuntos
Neoplasias da Mama/imunologia , Interferon Tipo I/imunologia , Células Matadoras Naturais/imunologia , Animais , Carcinogênese/imunologia , Linhagem Celular Tumoral , Feminino , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/imunologia
3.
J Immunol ; 202(12): 3370-3380, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31092636

RESUMO

The importance of antiviral CD8+ T cell recognition of alternative reading frame (ARF)-derived peptides is uncertain. In this study, we describe an epitope (NS1-ARF21-8) present in a predicted 14-residue peptide encoded by the +1 register of NS1 mRNA in the influenza A virus (IAV). NS1-ARF21-8 elicits a robust, highly functional CD8+ T cell response in IAV-infected BALB/c mice. NS1-ARF21-8 is presented from unspliced NS mRNA, likely from downstream initiation on a Met residue that comprises the P1 position of NS1-ARF21-8 Derived from a 14-residue peptide with no apparent biological function and negligible impacts on IAV infection, infectivity, and pathogenicity, NS1-ARF21-8 provides a clear demonstration of how immunosurveillance exploits natural errors in protein translation to provide antiviral immunity. We further show that IAV infection enhances a model cellular ARF translation, which potentially has important implications for virus-induced autoimmunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/metabolismo , Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , Infecções por Orthomyxoviridae/imunologia , Proteínas não Estruturais Virais/metabolismo , Processamento Alternativo , Animais , Modelos Animais de Doenças , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Vigilância Imunológica , Camundongos , Camundongos Endogâmicos BALB C , Fases de Leitura Aberta/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia
4.
Immunol Cell Biol ; 98(7): 563-576, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32330333

RESUMO

The participation of dendritic cells (DCs) in CD8+ T-cell-mediated allograft rejection is a long-standing question of great clinical relevance for tissue transplantation. Here, we show that Batf3-/- mice demonstrate delayed allo-skin graft rejection and are deficient in priming allo-specific CD8+ T cells. Batf3-/- mouse priming is restored by transferring either purified CD8α+ or CD103+ DCs, demonstrating the critical role of these cells in alloreactivity. Using Db -restricted antiviral F5 transgenic T-cell receptor T cells, which we demonstrate are alloreactive with H-2Kd , we show that cross-dressing of CD8α+ and CD103+ primes CD8+ T-cell or allo-specific responses in vitro and in vivo. These findings suggest novel strategies for moderating tissue rejection based on interfering with DC cross-dressing or subsequent interaction with T cells.


Assuntos
Linfócitos T CD8-Positivos , Células Dendríticas , Rejeição de Enxerto/imunologia , Animais , Antígenos CD , Antígenos CD8 , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Cadeias alfa de Integrinas , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Pele
5.
Immunol Cell Biol ; 97(9): 774-786, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31116465

RESUMO

Memory regulatory T cells (mTregs) have been demonstrated to persist long-term in hosts after the resolution of primary influenza A virus (IAV) infection. However, whether such IAV infection-experienced (IAV-experienced) mTregs differentiate into a phenotypically and functionally distinct Treg subset and what function they play at the infection site remains poorly defined. In this study, we characterized the phenotype, examined the responsiveness and assessed the suppressive function of IAV-experienced memory Tregs (mTregs). In comparison with inexperienced naïve Tregs (nTregs), mTregs exhibited elevated expression of CD39, CD69, CD103, cytotoxic T lymphocyte-associated antigen-4, leukocyte function-associated antigen-1 and programmed cell death-1 and could be activated in an antigen-specific manner in vitro and in vivo. When mTregs and nTregs were adoptively cotransferred into recipient mice, mTregs had a competitive advantage in migrating to the IAV-infected lungs. mTregs were more capable of controlling in vitro proliferation of CD4+ and CD8+ T cells and suppressed CD40 and CD86 upregulation on bone marrow-derived dendritic cells. Adoptively transferred mTregs, but not adoptively transferred nTregs, significantly attenuated body weight loss, lung pathology and immune cell infiltration into the infected lungs after IAV infection. These results suggest that mTregs generated after IAV infection differentiate into a phenotypically distinct and functionally enhanced Treg subset that can be activated in an antigen-specific manner to exert immunosuppression. We propose vaccination to induce such mTregs as a potential novel strategy to protect against severe IAV infection.


Assuntos
Memória Imunológica , Vírus da Influenza A/imunologia , Pulmão/imunologia , Pulmão/virologia , Infecções por Orthomyxoviridae/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Movimento Celular/imunologia , Proliferação de Células , Células Dendríticas/imunologia , Regulação para Baixo , Feminino , Pulmão/patologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Monócitos/patologia , Infiltração de Neutrófilos , Infecções por Orthomyxoviridae/virologia , Fenótipo , Redução de Peso
6.
BMC Cancer ; 19(1): 924, 2019 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-31521127

RESUMO

BACKGROUND: Current therapies fail to cure over a third of osteosarcoma patients and around three quarters of those with metastatic disease. "Smac mimetics" (also known as "IAP antagonists") are a new class of anti-cancer agents. Previous work revealed that cells from murine osteosarcomas were efficiently sensitized by physiologically achievable concentrations of some Smac mimetics (including GDC-0152 and LCL161) to killing by the inflammatory cytokine TNFα in vitro, but survived exposure to Smac mimetics as sole agents. METHODS: Nude mice were subcutaneously or intramuscularly implanted with luciferase-expressing murine 1029H or human KRIB osteosarcoma cells. The impacts of treatment with GDC-0152, LCL161 and/or doxorubicin were assessed by caliper measurements, bioluminescence, 18FDG-PET and MRI imaging, and by weighing resected tumors at the experimental endpoint. Metastatic burden was examined by quantitative PCR, through amplification of a region of the luciferase gene from lung DNA. ATP levels in treated and untreated osteosarcoma cells were compared to assess in vitro sensitivity. Immunophenotyping of cells within treated and untreated tumors was performed by flow cytometry, and TNFα levels in blood and tumors were measured using cytokine bead arrays. RESULTS: Treatment with GDC-0152 or LCL161 suppressed the growth of subcutaneously or intramuscularly implanted osteosarcomas. In both models, co-treatment with doxorubicin and Smac mimetics impeded average osteosarcoma growth to a greater extent than either drug alone, although these differences were not statistically significant. Co-treatments were also more toxic. Co-treatment with LCL161 and doxorubicin was particularly effective in the KRIB intramuscular model, impeding primary tumor growth and delaying or preventing metastasis. Although the Smac mimetics were effective in vivo, in vitro they only efficiently killed osteosarcoma cells when TNFα was supplied. Implanted tumors contained high levels of TNFα, produced by infiltrating immune cells. Spontaneous osteosarcomas that arose in genetically-engineered immunocompetent mice also contained abundant TNFα. CONCLUSIONS: These data imply that Smac mimetics can cooperate with TNFα secreted by tumor-associated immune cells to kill osteosarcoma cells in vivo. Smac mimetics may therefore benefit osteosarcoma patients whose tumors contain Smac mimetic-responsive cancer cells and TNFα-producing infiltrating cells.


Assuntos
Antineoplásicos/farmacologia , Cicloexanos/farmacologia , Pirróis/farmacologia , Tiazóis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Imageamento por Ressonância Magnética/métodos , Camundongos , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Immunol ; 198(10): 3835-3845, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28363906

RESUMO

CD8+ T cell immunosurveillance is based on recognizing oligopeptides presented by MHC class I molecules. Despite decades of study, the importance of protein ubiquitylation to peptide generation remains uncertain. In this study, we examined the ability of MLN7243, a recently described ubiquitin-activating enzyme E1 inhibitor, to block overall cytosolic peptide generation and generation of specific peptides from vaccinia- and influenza A virus-encoded proteins. We show that MLN7243 rapidly inhibits ubiquitylation in a variety of cell lines and can profoundly reduce the generation of cytosolic peptides. Kinetic analysis of specific peptide generation reveals that ubiquitylation of defective ribosomal products is rate limiting in generating class I peptide complexes. More generally, our findings demonstrate that the requirement for ubiquitylation in MHC class I-restricted Ag processing varies with class I allomorph, cell type, source protein, and peptide context. Thus, ubiquitin-dependent and -independent pathways robustly contribute to MHC class I-based immunosurveillance.


Assuntos
Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe I/imunologia , Nucleosídeos/farmacologia , Peptídeos/imunologia , Sulfonamidas/farmacologia , Linfócitos T/imunologia , Animais , Linhagem Celular , Citosol/química , Citosol/imunologia , Inibidores Enzimáticos/farmacologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Vírus da Influenza A/química , Vírus da Influenza A/imunologia , Cinética , Ligantes , Camundongos , Monitorização Imunológica , Peptídeos/metabolismo , Pirazóis , Pirimidinas , Sulfetos , Ubiquitinação , Vaccinia virus/química , Vaccinia virus/imunologia
8.
Immunol Cell Biol ; 96(9): 981-993, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29738610

RESUMO

The role of the immunoproteasome is perceived as confined to adaptive immune responses given its ability to produce peptides ideal for MHC Class-I binding. Here, we demonstrate that the immunoproteasome subunit, LMP2, has functions beyond its immunomodulatory role. Using LMP2-deficient mice, we demonstrate that LMP2 is crucial for lymphocyte development and survival in the periphery. Moreover, LMP2-deficient lymphocytes show impaired degradation of key BH3-only proteins, resulting in elevated levels of pro-apoptotic BIM and increased cell death. Interestingly, LMP2 is the sole immunoproteasome subunit required for BIM degradation. Together, our results suggest LMP2 has important housekeeping functions and represents a viable therapeutic target for cancer.


Assuntos
Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/imunologia , Cisteína Endopeptidases/imunologia , Linfócitos/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Animais , Western Blotting , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Cisteína Endopeptidases/deficiência , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/deficiência
9.
Immunity ; 28(6): 787-98, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18549799

RESUMO

The nature of crosspriming immunogens for CD8(+) T cell responses is highly controversial. By using a panel of T cell receptor-like antibodies specific for viral peptides bound to mouse D(b) major histocompatibility complex class I molecules, we show that an exceptional peptide (PA(224-233)) expressed as a viral minigene product formed a sizeable cytosolic pool continuously presented for hours after protein synthesis was inhibited. PA(224-233) pool formation required active cytosolic heat-shock protein 90 but not ER g96 and uniquely enabled crosspriming by this peptide. These findings demonstrate that exceptional class I binding oligopeptides that escape proteolytic degradation are potent crosspriming agents. Thus, the feeble immunogenicity of natural proteasome products in crosspriming can be attributed to their evanescence in donor cells and not an absolute inability of cytosolic oligopeptides to be transferred to and presented by professional antigen-presenting cells.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Vírus da Influenza A/imunologia , Peptídeos/imunologia , Animais , Anticorpos/imunologia , Antígenos Virais/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Feminino , Proteínas de Choque Térmico HSP90/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Camundongos , Peptídeos/metabolismo , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo
10.
Proteomics ; 15(11): 1808-12, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25643623

RESUMO

Differentiation of CD8(+) T lymphocytes into effector and memory cells is key for an adequate immune response and relies on complex interplay of pathways that convey signals from the cell surface to the nucleus. In this study, we investigated the proteome of four cytotoxic T-cell subtypes; naïve, recently activated effector, effector, and memory cells. Cells were fractionated into membrane, cytosol, soluble nuclear, chromatin-bound, and cytoskeletal compartments. Following LC-MS/MS analysis, identified peptides were analyzed via MaxQuant. Compartment fractionation and gel-LC-MS separation resulted in 2399 proteins identified in total. Comparison between the different subsets resulted in 146 significantly regulated proteins for naïve and effector cells, followed by 116 for activated, and 55 for memory cells. Besides Granzyme B signaling (for activated and/ or effector cells vs. naïve cells), the most prominent changes occurred in the TCA cycle and aspartate degradation. These changes suggest that correct balancing of metabolism is key for differentiation processes. All MS data have been deposited in the ProteomeXchange with identifier PXD001065 (http://proteomecentral.proteomexchange.org/dataset/PXD001065).


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Proteoma/análise , Animais , Cromatografia Líquida de Alta Pressão , Bases de Dados de Proteínas , Espectrometria de Massas , Camundongos Endogâmicos C57BL , Proteoma/isolamento & purificação , Espectrometria de Massas em Tandem
11.
Eur J Immunol ; 44(12): 3500-3, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25399798

RESUMO

Vertebrates have evolved to express major histocompatibility complexes (MHCs) that present peptides of the intra-cellular proteome for immunosurveillance against viruses and tumor. The MHC class I (MHC-I) processing and presentation pathway allows for scrutiny of all cellular proteins and peptides by CD8(+) cytotoxic T cells. The proteasome is part of the specialised machinery that degrades proteins down to peptides with the correct sequence for MHC-I binding. However, much conjecture lies as to how the various proteasome isoforms and their active subunits create antigenic peptides, and if the specialised immunoproteasome solely performs this job. In this issue of the European Journal of Immunology, Mishto et al. [Eur. J. Immunol. 2014. 44: 3508-3521] address this question through systematic biochemical peptide degradation studies and provide new insights into the functions of proteasome ß-subunits.


Assuntos
Apresentação de Antígeno/fisiologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Peptídeos/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Proteólise , Animais , Humanos
12.
Immunol Cell Biol ; 93(5): 500-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25559620

RESUMO

T-cell repertoire is selected according to self peptide-MHC (major histocompatibility complex) complexes in the thymus. Although most peripheral T cells recognize specific pathogen-derived peptides complexed to self-MHC exclusively, some possess cross-reactivity to other self or foreign peptides presented by self-MHC molecules; a phenomenon often termed T-cell receptor (TCR) promiscuity or degeneracy. TCR promiscuity has been attributed to various autoimmune conditions. On the other hand, it is considered a mechanism for a relatively limited TCR repertoire to deal with a potentially much larger antigenic peptide repertoire. Such property has also been utilized to bypass self-tolerance for cancer vaccine development. Although many studies explored such degeneracy for peptide of the same length, few studies reported such properties for peptides of different length. In this study, we finely characterized the CD8(+) T-cell response specific for a 11mer peptide derived from influenza A viral polymerase basic protein 2. The short-term T-cell line, despite possessing highly biased TCR, was able to react with multiple peptides of different length sharing the same core sequence. Out data clearly showed the importance of detailed and quantitative assessments for such T-cell specificity. Our data also emphasize the importance of biochemical demonstration of the naturally presented minimal peptide.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígeno de Histocompatibilidade H-2D/metabolismo , Orthomyxoviridae/imunologia , Fragmentos de Peptídeos/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Vacinas Anticâncer , Linhagem Celular , Reações Cruzadas , Cisteína Endopeptidases/genética , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/genética , Ligação Proteica , RNA Polimerase Dependente de RNA/genética , Especificidade do Receptor de Antígeno de Linfócitos T , Proteínas Virais/genética
13.
J Virol ; 88(20): 11760-73, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25078703

RESUMO

Antigen-specific CD4(+) T cells are essential for effective virus-specific host responses, with recent human challenge studies (in volunteers) establishing their importance for influenza A virus (IAV)-specific immunity. However, while many IAV CD4(+) T cell epitopes have been identified, few are known to stimulate immunodominant CD4(+) T cell responses. Moreover, much remains unclear concerning the major antigen(s) responded to by the human CD4(+) T cells and the extents and magnitudes of these responses. We initiated a systematic screen of immunodominant CD4(+) T cell responses to IAV in healthy individuals. Using in vitro expanded-multispecificity IAV-specific T cell lines and individual IAV protein antigens produced by recombinant vaccinia viruses, we found that the internal matrix protein 1 (M1) and nucleoprotein (NP) were the immunodominant targets of CD4(+) T cell responses. Ten epitopes derived from M1 and NP were definitively characterized. Furthermore, epitope sequence conservation analysis established that immunodominance correlated with an increased frequency of mutations, reflecting the fact that these prominent epitopes are under greater selective pressure. Such evidence that particular CD4(+) T cells are important for protection/recovery is of value for the development of novel IAV vaccines and for our understanding of different profiles of susceptibility to these major pathogens. Importance: Influenza virus causes half a million deaths annually. CD4(+) T cell responses have been shown to be important for protection against influenza and for recovery. CD4(+) T cell responses are also critical for efficient CD8(+) T cell response and antibody response. As immunodominant T cells generally play a more important role, characterizing these immunodominant responses is critical for influenza vaccine development. We show here that the internal matrix protein 1 (M1) and nucleoprotein (NP), rather than the surface proteins reported previously, are the immunodominant targets of CD4(+) T cell responses. Interestingly, these immunodominant epitope regions accumulated many mutations over time, which likely indicates increased immune pressure. These findings have significant implications for the design of T cell-based influenza vaccines.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vírus da Influenza A/imunologia , Proteínas Virais/imunologia , Células Cultivadas , Humanos
14.
J Immunol ; 191(1): 52-9, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23709680

RESUMO

The three proteasome subunits with proteolytic activity are encoded by standard or immunoproteasome genes. Many proteasomes expressed by normal cells and cells exposed to cytokines are "mixed", that is, contain both standard and immunoproteasome subunits. Using a panel of 38 defined influenza A virus-derived epitopes recognized by C57BL/6 mouse CD8(+) T cells, we used mice with targeted disruption of ß1i, ß2i, or ß5i/ß2i genes to examine the contribution of mixed proteasomes to the immunodominance hierarchy of antiviral CD8(+) T cells. We show that each immunoproteasome subunit has large effects on the primary and recall immunodominance hierarchies due to modulating both the available T cell repertoire and generation of individual epitopes as determined both biochemically and kinetically in Ag presentation assays. These findings indicate that mixed proteasomes function to enhance the diversity of peptides and support a broad CD8(+) T cell response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Vírus da Influenza A/imunologia , Biossíntese Peptídica , Complexo de Endopeptidases do Proteassoma/imunologia , Regulação para Cima/imunologia , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/biossíntese , Animais , Linfócitos T CD8-Positivos/enzimologia , Técnicas de Cocultura , Vírus da Influenza A/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Biossíntese Peptídica/genética , Biossíntese Peptídica/imunologia , Complexo de Endopeptidases do Proteassoma/administração & dosagem , Complexo de Endopeptidases do Proteassoma/genética , Células Tumorais Cultivadas
15.
Eur J Immunol ; 43(2): 533-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23124877

RESUMO

CD4(+) CD25(+) FoxP3(+) naturally occurring regulatory T (Treg) cells play a crucial role in the maintenance of immune tolerance and in preventing autoimmune pathology. Interventions that expand Treg cells are highly desirable, as they may offer novel treatment options in a variety of autoimmune and transplantation settings. Paralleling previous preclinical studies, we demonstrate here that administration of the hematopoietic growth factor Flt3L to human subjects increases the frequency and absolute number of Treg cells, and reduces the ratio of CD8(+) T cells to Treg cells in the peripheral blood. The increase in Treg cells was due to enhanced Treg-cell proliferation rather than release of Treg cells from the thymus. Further studies revealed that Flt3L-induced proliferation of Treg cells was an indirect effect that occurred via the interaction of Treg cells with the Flt3L-expanded pool of CD1c(+) myeloid dendritic cells. On the basis of these findings, Flt3L may represent a promising agent for promoting immune tolerance in a variety of clinical settings.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/imunologia , Proteínas de Membrana/imunologia , Linfócitos T Reguladores/imunologia , Tirosina Quinase 3 Semelhante a fms/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Processos de Crescimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Proteínas de Membrana/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Linfócitos T Reguladores/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo
16.
Proc Natl Acad Sci U S A ; 108(22): 9178-83, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21562214

RESUMO

Immunodominant T-cell responses are important for virus clearance. However, the identification of immunodominant T-cell peptide + HLA glycoprotein epitopes has been hindered by the extent of HLA polymorphism and the limitations of predictive algorithms. A simple, systematic approach has been used here to screen for immunodominant CD8(+) T-cell specificities. The analysis targeted healthy HLA-A2(+) donors to allow comparison with responses to the well-studied influenza matrix protein 1 epitope. Although influenza matrix protein 1 was consistently detected in all individual samples in our study, the response to this epitope was only immunodominant in three of eight, whereas for the other five, prominent CD8(+) T-cell responses tended to focus on various peptides from the influenza nucleoprotein that were not presented by HLA-A2. Importantly, with the four immunodominant T-cell epitopes identified here, only one would have been detected by the current prediction programs. The other three peptides would have been either considered too long or classified as not containing typical HLA binding motifs. Our data stress the importance of systematic analysis for discovering HLA-dependent, immunodominant CD8(+) T-cell epitopes derived from viruses and tumors. Focusing on HLA-A2 and predictive algorithms may be too limiting as we seek to develop targeted immunotherapy and vaccine strategies that depend on T cell-mediated immunity.


Assuntos
Linfócitos T CD8-Positivos/citologia , Antígeno HLA-A2/genética , Vírus da Influenza A/metabolismo , Biologia Computacional/métodos , Epitopos/química , Epitopos de Linfócito T/química , Antígeno HLA-A2/imunologia , Humanos , Epitopos Imunodominantes/imunologia , Imunoterapia/métodos , Interferon gama/metabolismo , Leucócitos Mononucleares/citologia , Nucleoproteínas/metabolismo , Orthomyxoviridae/metabolismo , Peptídeos/química , Vírus/metabolismo
17.
J Immunol ; 184(8): 4115-22, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20228196

RESUMO

Proteasomes are multisubunit proteases that initiate degradation of many Ags presented by MHC class I molecules. Vertebrates express alternate forms of each of the three catalytic proteasome subunits: standard subunits, and immunosubunits, which are constitutively expressed by APCs and are induced in other cell types by exposure to cytokines. The assembly of mixed proteasomes containing standard subunits and immunosubunits is regulated in a tissue specific manner. In this study, we report that the presence of mixed proteasomes in immune cells in LMP2(-/-) mice compromises multiple components that contribute to the generation of antiviral Ab responses, including splenic B cell numbers, survival and function of adoptively transferred B cells, Th cell function, and dendritic cell secretion of IL-6, TNF-alpha, IL-1beta, and type I IFNs. These defects did not result from compromised overall protein degradation, rather they were associated with altered NF-kappaB activity. These findings demonstrate an important role for immunoproteasomes in immune cell function beyond their contribution to Ag processing.


Assuntos
Anticorpos Antivirais/biossíntese , Cisteína Endopeptidases/fisiologia , Imunidade Inata , Vírus da Influenza A/imunologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Subunidades Proteicas/fisiologia , Animais , Anticorpos Antivirais/metabolismo , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Linfócitos B/enzimologia , Linfócitos B/imunologia , Linfócitos B/virologia , Células Cultivadas , Cisteína Endopeptidases/deficiência , Cisteína Endopeptidases/genética , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunidade Inata/genética , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Complexo de Endopeptidases do Proteassoma/deficiência , Complexo de Endopeptidases do Proteassoma/genética , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia
18.
Nat Commun ; 13(1): 6539, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36344500

RESUMO

Cancers evade the immune system through the process of cancer immunoediting. While immune checkpoint inhibitors are effective for reactivating tumour immunity in some cancer types, many other solid cancers, including breast cancer, remain largely non-responsive. Understanding how non-responsive cancers evade immunity and whether this occurs at the clonal level will improve immunotherapeutic design. Here we use DNA barcoding to track murine mammary cancer cell clones during immunoediting and determine clonal transcriptional profiles that allow immune evasion following anti-PD1 plus anti-CTLA4 immunotherapy. Clonal diversity is significantly restricted by immunotherapy treatment in both primary tumours and metastases, demonstrating selection for pre-existing breast cancer cell populations and ongoing immunoediting during metastasis and treatment. Immunotherapy resistant clones express a common gene signature associated with poor survival of basal-like breast cancer patient cohorts. At least one of these genes has an existing small molecule that can potentially be used to improve immunotherapy response.


Assuntos
Neoplasias da Mama , Código de Barras de DNA Taxonômico , Humanos , Camundongos , Animais , Feminino , Imunoterapia , Fatores Imunológicos , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Estudos Longitudinais
19.
Cancer Res Commun ; 2(7): 706-724, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-36923279

RESUMO

Inhibiting the androgen receptor (AR), a ligand-activated transcription factor, with androgen deprivation therapy is a standard-of-care treatment for metastatic prostate cancer. Paradoxically, activation of AR can also inhibit the growth of prostate cancer in some patients and experimental systems, but the mechanisms underlying this phenomenon are poorly understood. This study exploited a potent synthetic androgen, methyltestosterone (MeT), to investigate AR agonist-induced growth inhibition. MeT strongly inhibited growth of prostate cancer cells expressing AR, but not AR-negative models. Genes and pathways regulated by MeT were highly analogous to those regulated by DHT, although MeT induced a quantitatively greater androgenic response in prostate cancer cells. MeT potently downregulated DNA methyltransferases, leading to global DNA hypomethylation. These epigenomic changes were associated with dysregulation of transposable element expression, including upregulation of endogenous retrovirus (ERV) transcripts after sustained MeT treatment. Increased ERV expression led to accumulation of double-stranded RNA and a "viral mimicry" response characterized by activation of IFN signaling, upregulation of MHC class I molecules, and enhanced recognition of murine prostate cancer cells by CD8+ T cells. Positive associations between AR activity and ERVs/antiviral pathways were evident in patient transcriptomic data, supporting the clinical relevance of our findings. Collectively, our study reveals that the potent androgen MeT can increase the immunogenicity of prostate cancer cells via a viral mimicry response, a finding that has potential implications for the development of strategies to sensitize this cancer type to immunotherapies. Significance: Our study demonstrates that potent androgen stimulation of prostate cancer cells can elicit a viral mimicry response, resulting in enhanced IFN signaling. This finding may have implications for the development of strategies to sensitize prostate cancer to immunotherapies.


Assuntos
Neoplasias da Próstata , Receptores Androgênicos , Masculino , Humanos , Animais , Camundongos , Receptores Androgênicos/genética , Androgênios/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Antagonistas de Androgênios/farmacologia , Linfócitos T CD8-Positivos/metabolismo , DNA
20.
Nat Commun ; 12(1): 3950, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34168137

RESUMO

The concept that extracellular vesicles (EVs) from the diet can be absorbed by the intestinal tract of the consuming organism, be bioavailable in various organs, and in-turn exert phenotypic changes is highly debatable. Here, we isolate EVs from both raw and commercial bovine milk and characterize them by electron microscopy, nanoparticle tracking analysis, western blotting, quantitative proteomics and small RNA sequencing analysis. Orally administered bovine milk-derived EVs survive the harsh degrading conditions of the gut, in mice, and is subsequently detected in multiple organs. Milk-derived EVs orally administered to mice implanted with colorectal and breast cancer cells reduce the primary tumor burden. Intriguingly, despite the reduction in primary tumor growth, milk-derived EVs accelerate metastasis in breast and pancreatic cancer mouse models. Proteomic and biochemical analysis reveal the induction of senescence and epithelial-to-mesenchymal transition in cancer cells upon treatment with milk-derived EVs. Timing of EV administration is critical as oral administration after resection of the primary tumor reverses the pro-metastatic effects of milk-derived EVs in breast cancer models. Taken together, our study provides context-based and opposing roles of milk-derived EVs as metastasis inducers and suppressors.


Assuntos
Vesículas Extracelulares , Leite/citologia , Neoplasias Experimentais/patologia , Administração Oral , Animais , Disponibilidade Biológica , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Bovinos , Linhagem Celular Tumoral , Proliferação de Células , Transição Epitelial-Mesenquimal , Vesículas Extracelulares/química , Vesículas Extracelulares/genética , Feminino , Humanos , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/terapia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa