Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 133(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38099496

RESUMO

Cell therapies such as tumor-infiltrating lymphocyte (TIL) therapy have shown promise in the treatment of patients with refractory solid tumors, with improvement in response rates and durability of responses nevertheless sought. To identify targets capable of enhancing the antitumor activity of T cell therapies, large-scale in vitro and in vivo clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screens were performed, with the SOCS1 gene identified as a top T cell-enhancing target. In murine CD8+ T cell-therapy models, SOCS1 served as a critical checkpoint in restraining the accumulation of central memory T cells in lymphoid organs as well as intermediate (Texint) and effector (Texeff) exhausted T cell subsets derived from progenitor exhausted T cells (Texprog) in tumors. A comprehensive CRISPR tiling screen of the SOCS1-coding region identified sgRNAs targeting the SH2 domain of SOCS1 as the most potent, with an sgRNA with minimal off-target cut sites used to manufacture KSQ-001, an engineered TIL therapy with SOCS1 inactivated by CRISPR/Cas9. KSQ-001 possessed increased responsiveness to cytokine signals and enhanced in vivo antitumor function in mouse models. These data demonstrate the use of CRISPR/Cas9 screens in the rational design of T cell therapies.


Assuntos
Sistemas CRISPR-Cas , Neoplasias , Humanos , Animais , Camundongos , RNA Guia de Sistemas CRISPR-Cas , Linfócitos do Interstício Tumoral , Imunoterapia Adotiva , Neoplasias/genética , Edição de Genes , Proteína 1 Supressora da Sinalização de Citocina/genética
2.
Cell Rep Med ; 4(1): 100899, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36652908

RESUMO

The non-canonical inflammasome sensor caspase-11 and gasdermin D (GSDMD) drive inflammation and pyroptosis, a type of immunogenic cell death that favors cell-mediated immunity (CMI) in cancer, infection, and autoimmunity. Here we show that caspase-11 and GSDMD are required for CD8+ and Th1 responses induced by nanoparticulate vaccine adjuvants. We demonstrate that nanoparticle-induced reactive oxygen species (ROS) are size dependent and essential for CMI, and we identify 50- to 60-nm nanoparticles as optimal inducers of ROS, GSDMD activation, and Th1 and CD8+ responses. We reveal a division of labor for IL-1 and IL-18, where IL-1 supports Th1 and IL-18 promotes CD8+ responses. Exploiting size as a key attribute, we demonstrate that biodegradable poly-lactic co-glycolic acid nanoparticles are potent CMI-inducing adjuvants. Our work implicates ROS and the non-canonical inflammasome in the mode of action of polymeric nanoparticulate adjuvants and establishes adjuvant size as a key design principle for vaccines against cancer and intracellular pathogens.


Assuntos
Inflamassomos , Nanopartículas , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Caspases/metabolismo , Interleucina-1/metabolismo
3.
Eur J Immunol ; 48(4): 705-715, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29349774

RESUMO

The effectiveness of many vaccines licensed for clinical use relates to the induction of neutralising antibodies, facilitated by the inclusion of vaccine adjuvants, particularly alum. However, the ability of alum to preferentially promote humoral rather than cellular, particularly Th1-type responses, is not well understood. We demonstrate that alum activates immunosuppressive mechanisms following vaccination, which limit its capacity to induce Th1 responses. One of the key cytokines limiting excessive immune responses is IL-10. Injection of alum primed draining lymph node cells for enhanced IL-10 secretion ex vivo. Moreover, at the site of injection, macrophages and dendritic cells were key sources of IL-10 expression. Alum strongly enhanced the transcription and secretion of IL-10 by macrophages and dendritic cells. The absence of IL-10 signalling did not compromise alum-induced cell infiltration into the site of injection, but resulted in enhanced antigen-specific Th1 responses after vaccination. In contrast to its decisive regulatory role in regulating Th1 responses, there was no significant change in antigen-specific IgG1 antibody production following vaccination with alum in IL-10-deficient mice. Overall, these findings indicate that injection of alum promotes IL-10, which can block Th1 responses and may explain the poor efficacy of alum as an adjuvant for inducing protective Th1 immunity.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alúmen/farmacologia , Células Dendríticas/imunologia , Interleucina-10/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Células Th1/imunologia , Animais , Células Cultivadas , Escherichia coli/imunologia , Feminino , Interleucina-10/biossíntese , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vacinas/imunologia
4.
Int J Nanomedicine ; 9: 5231-46, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25419128

RESUMO

Targeting antigen to dendritic cells (DCs) is a powerful and novel strategy for vaccination. Priming or loading DCs with antigen controls whether subsequent immunity will develop and hence whether effective vaccination can be achieved. The goal of our present work was to increase the potency of DC-based antitumor vaccines by overcoming inherent limitations associated with antigen stability and cross-presentation. Nanoparticles prepared from the biodegradable polymer poly(lactic-co-glycolic acid) have been extensively used in clinical settings for drug delivery and are currently the subject of intensive investigation as antigen delivery vehicles for vaccine applications. Here we describe a nanoparticulate delivery system with the ability to simultaneously carry a high density of protein-based antigen while displaying a DC targeting ligand on its surface. Utilizing a targeting motif specific for the DC-associated surface ligand DEC-205, we show that targeted nanoparticles encapsulating a MART-127-35 peptide are both internalized and cross-presented with significantly higher efficiency than isotype control-coated nanoparticles in human cells. In addition, the DEC-205-labeled nanoparticles rapidly escape from the DC endosomal compartment and do not colocalize with markers of early (EEA-1) or late endosome/lysosome (LAMP-1). This indicates that encapsulated antigens delivered by nanoparticles may have direct access to the class I cytoplasmic major histocompatibility complex loading machinery, overcoming the need for "classical" cross-presentation and facilitating heightened DC stimulation of anti-tumor CD8(+) T-cells. These results indicate that this delivery system provides a flexible and versatile methodology to deliver melanoma-associated antigen to DCs, with both high efficiency and heightened potency.


Assuntos
Antígenos CD/imunologia , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Ácido Láctico/química , Lectinas Tipo C/imunologia , Antígeno MART-1/administração & dosagem , Melanoma/imunologia , Nanopartículas/química , Ácido Poliglicólico/química , Receptores de Superfície Celular/imunologia , Apresentação de Antígeno/efeitos dos fármacos , Vacinas Anticâncer/imunologia , Humanos , Ácido Láctico/imunologia , Antígeno MART-1/imunologia , Melanoma/terapia , Antígenos de Histocompatibilidade Menor , Terapia de Alvo Molecular , Nanopartículas/administração & dosagem , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
5.
Nat Nanotechnol ; 9(8): 639-47, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25086604

RESUMO

Clinical translation of cell therapies requires strategies that can manufacture cells efficiently and economically. One promising way to reproducibly expand T cells for cancer therapy is by attaching the stimuli for T cells onto artificial substrates with high surface area. Here, we show that a carbon nanotube-polymer composite can act as an artificial antigen-presenting cell to efficiently expand the number of T cells isolated from mice. We attach antigens onto bundled carbon nanotubes and combined this complex with polymer nanoparticles containing magnetite and the T-cell growth factor interleukin-2 (IL-2). The number of T cells obtained was comparable to clinical standards using a thousand-fold less soluble IL-2. T cells obtained from this expansion were able to delay tumour growth in a murine model for melanoma. Our results show that this composite is a useful platform for generating large numbers of cytotoxic T cells for cancer immunotherapy.


Assuntos
Proteínas Imobilizadas/química , Melanoma/terapia , Nanotubos de Carbono/química , Polímeros/química , Linfócitos T/citologia , Linfócitos T/transplante , Animais , Antígenos/química , Antígenos/imunologia , Técnicas de Cultura de Células/métodos , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Humanos , Proteínas Imobilizadas/imunologia , Imunoterapia Adotiva , Interleucina-2/química , Interleucina-2/imunologia , Melanoma/imunologia , Camundongos , Linfócitos T/imunologia
6.
Eur J Immunol ; 42(10): 2709-19, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22777876

RESUMO

Alum is the principal vaccine adjuvant for clinical applications but it is a poor inducer of cellular immunity and is not an optimal adjuvant for vaccines where Th1 responses are required for protection. The mechanism underlying the inefficiency of alum in promoting Th1 responses is not fully understood. We show that aluminium hydroxide, aluminium phosphate, and calcium phosphate adjuvants inhibit the secretion of the Th1 polarizing cytokine, IL-12 by dendritic cells (DCs). Alum selectively inhibited DC expression of the IL-12p35 subunit and the inhibitory effect results from adjuvant-induced PI3 kinase signaling. To develop a more effective adjuvant for promoting cell-mediated immunity, we investigated alternative particulates and found that in contrast to alum, the cationic polysaccharide chitosan did not inhibit IL-12 secretion. A combination of chitosan and the TLR9 agonist CpG activated the NLRP3 inflammasome and enhanced secretion of IL-12 and the other key Th1 and Th17-cell polarizing cytokines. When used as an adjuvant, CpG-chitosan induced NLRP3-dependent antigen-specific Th1 and Th17 responses. A combination of alum and CpG also enhanced Th1 and Th17 responses but was less effective than CpG-chitosan. Therefore, chitosan is an attractive alternative to alum in adjuvants for vaccines where potent cell-mediated immunity is required.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alúmen/farmacologia , Células Dendríticas/imunologia , Subunidade p35 da Interleucina-12/metabolismo , Células Th1/imunologia , Células Th17/imunologia , Animais , Células Cultivadas , Quitosana/farmacologia , Células Dendríticas/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/imunologia , Imunidade Celular , Subunidade p35 da Interleucina-12/genética , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/imunologia
7.
Trends Biotechnol ; 29(6): 294-306, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21459467

RESUMO

Vaccine development has progressed significantly and has moved from whole microorganisms to subunit vaccines that contain only their antigenic proteins. Subunit vaccines are often less immunogenic than whole pathogens; therefore, adjuvants must amplify the immune response, ideally establishing both innate and adaptive immunity. Incorporation of antigens into biomaterials, such as liposomes and polymers, can achieve a desired vaccine response. The physical properties of these platforms can be easily manipulated, thus allowing for controlled delivery of immunostimulatory factors and presentation of pathogen-associated molecular patterns (PAMPs) that are targeted to specific immune cells. Targeting antigen to immune cells via PAMP-modified biomaterials is a new strategy to control the subsequent development of immunity and, in turn, effective vaccination. Here, we review the recent advances in both immunology and biomaterial engineering that have brought particulate-based vaccines to reality.


Assuntos
Adjuvantes Imunológicos/farmacologia , Materiais Biocompatíveis/farmacologia , Sistema Imunitário/efeitos dos fármacos , Imunidade Celular , Receptores de Reconhecimento de Padrão/efeitos dos fármacos , Vacinas/imunologia , Humanos
8.
J Biol Chem ; 286(11): 9587-97, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21228274

RESUMO

Autophagy is a key regulator of cellular homeostasis that can be activated by pathogen-associated molecules and recently has been shown to influence IL-1ß secretion by macrophages. However, the mechanisms behind this are unclear. Here, we describe a novel role for autophagy in regulating the production of IL-1ß in antigen-presenting cells. After treatment of macrophages with Toll-like receptor ligands, pro-IL-1ß was specifically sequestered into autophagosomes, whereas further activation of autophagy with rapamycin induced the degradation of pro-IL-1ß and blocked secretion of the mature cytokine. Inhibition of autophagy promoted the processing and secretion of IL-1ß by antigen-presenting cells in an NLRP3- and TRIF-dependent manner. This effect was reduced by inhibition of reactive oxygen species but was independent of NOX2. Induction of autophagy in mice in vivo with rapamycin reduced serum levels of IL-1ß in response to challenge with LPS. These data demonstrate that autophagy controls the production of IL-1ß through at least two separate mechanisms: by targeting pro-IL-1ß for lysosomal degradation and by regulating activation of the NLRP3 inflammasome.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Autofagia/fisiologia , Interleucina-1beta/metabolismo , Lisossomos/metabolismo , Macrófagos/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antibacterianos/farmacologia , Células Apresentadoras de Antígenos/citologia , Autofagia/efeitos dos fármacos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Feminino , Interleucina-1beta/genética , Ligantes , Lipopolissacarídeos/farmacologia , Lisossomos/genética , Macrófagos/citologia , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , NADPH Oxidase 2 , NADPH Oxidases , Proteína 3 que Contém Domínio de Pirina da Família NLR , Sirolimo/farmacologia , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
9.
Nat Immunol ; 11(10): 897-904, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20835230

RESUMO

Interleukin 1ß (IL-1ß) is an important inflammatory mediator of type 2 diabetes. Here we show that oligomers of islet amyloid polypeptide (IAPP), a protein that forms amyloid deposits in the pancreas during type 2 diabetes, triggered the NLRP3 inflammasome and generated mature IL-1ß. One therapy for type 2 diabetes, glyburide, suppressed IAPP-mediated IL-1ß production in vitro. Processing of IL-1ß initiated by IAPP first required priming, a process that involved glucose metabolism and was facilitated by minimally oxidized low-density lipoprotein. Finally, mice transgenic for human IAPP had more IL-1ß in pancreatic islets, which localized together with amyloid and macrophages. Our findings identify previously unknown mechanisms in the pathogenesis of type 2 diabetes and treatment of pathology caused by IAPP.


Assuntos
Amiloide/metabolismo , Proteínas de Transporte/metabolismo , Diabetes Mellitus Tipo 2/imunologia , Interleucina-1beta/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glibureto/farmacologia , Humanos , Hipoglicemiantes/farmacologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ratos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética
10.
Eur J Immunol ; 40(3): 634-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20201019

RESUMO

Adjuvants are essential for enhancing and directing immunity to vaccine antigens. Most adjuvants in clinical use are particulates, but how they drive innate and adaptive immune responses is unclear. A major recent advance was the demonstration that particulate adjuvants promote activation of the NLRP3 inflammasome. The mechanisms underlying this activation have been partly resolved and the role of NLRP3 in particulate adjuvant-induced adaptive immunity is currently the subject of intense interest.


Assuntos
Adjuvantes Imunológicos , Proteínas de Transporte/imunologia , Inflamação/imunologia , Complexos Multiproteicos/imunologia , Vacinas/imunologia , Animais , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR
11.
Proc Natl Acad Sci U S A ; 106(3): 870-5, 2009 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19139407

RESUMO

Many currently used and candidate vaccine adjuvants are particulate in nature, but their mechanism of action is not well understood. Here, we show that particulate adjuvants, including biodegradable poly(lactide-co-glycolide) (PLG) and polystyrene microparticles, dramatically enhance secretion of interleukin-1beta (IL-1beta) by dendritic cells (DCs). The ability of particulates to promote IL-1beta secretion and caspase 1 activation required particle uptake by DCs and NALP3. Uptake of microparticles induced lysosomal damage, whereas particle-mediated enhancement of IL-1beta secretion required phagosomal acidification and the lysosomal cysteine protease cathepsin B, suggesting a role for lysosomal damage in inflammasome activation. Although the presence of a Toll-like receptor (TLR) agonist was required to induce IL-1beta production in vitro, injection of the adjuvants in the absence of TLR agonists induced IL-1beta production at the injection site, indicating that endogenous factors can synergize with particulates to promote inflammasome activation. The enhancement of antigen-specific antibody production by PLG microparticles was independent of NALP3. However, the ability of PLG microparticles to promote antigen-specific IL-6 production by T cells and the recruitment and activation of a population of CD11b(+)Gr1(-) cells required NALP3. Our data demonstrate that uptake of microparticulate adjuvants by DCs activates the NALP3 inflammasome, and this contributes to their enhancing effects on innate and antigen-specific cellular immunity.


Assuntos
Adjuvantes Imunológicos/farmacologia , Proteínas de Transporte/fisiologia , Células Dendríticas/metabolismo , Vacinas/administração & dosagem , Adjuvantes Imunológicos/farmacocinética , Animais , Formação de Anticorpos , Caspase 1/fisiologia , Catepsina B/fisiologia , Movimento Celular , Células Cultivadas , Feminino , Interleucina-1beta/biossíntese , Ácido Láctico/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Poliestirenos/farmacologia , Receptores Toll-Like/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...