Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Immunol ; 192(12): 6111-9, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24829407

RESUMO

Cancer progression is associated with inflammation, increased metabolic demand, infection, cachexia, and eventually death. Myeloid-derived suppressor cells (MDSCs) commonly expand during cancer and are associated with adaptive immune suppression and inflammatory metabolite production. We propose that cancer-induced cachexia is driven at least in part by the expansion of MDSCs. MDSC expansion in 4T1 mammary carcinoma-bearing hosts is associated with induction of a hepatic acute-phase protein response and altered host energy and fat metabolism, and eventually reduced survival to polymicrobial sepsis and endotoxemia. Similar results are also seen in mice bearing a Lewis lung carcinoma and a C26 colon adenocarcinoma. However, a similar cachexia response is not seen with equivalent growth of the 66C4 subclone of 4T1, in which MDSC expansion does not occur. Importantly, reducing MDSC numbers in 4T1-bearing animals can ameliorate some of these late responses and reduce susceptibility to inflammation-induced organ injury and death. In addition, administering MDSCs from both tumor- and nontumor-bearing mice can produce an acute-phase response. Thus, we propose a previously undescribed mechanism for the development of cancer cachexia, whereby progressive MDSC expansion contributes to changes in host protein and energy metabolism and reduced resistance to infection.


Assuntos
Caquexia/imunologia , Tolerância Imunológica , Células Mieloides/imunologia , Neoplasias Experimentais/imunologia , Animais , Caquexia/etiologia , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/patologia , Neoplasias Experimentais/patologia
2.
J Exp Med ; 204(6): 1463-74, 2007 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-17548519

RESUMO

Polymicrobial sepsis alters the adaptive immune response and induces T cell suppression and Th2 immune polarization. We identify a GR-1(+)CD11b(+) population whose numbers dramatically increase and remain elevated in the spleen, lymph nodes, and bone marrow during polymicrobial sepsis. Phenotypically, these cells are heterogeneous, immature, predominantly myeloid progenitors that express interleukin 10 and several other cytokines and chemokines. Splenic GR-1(+) cells effectively suppress antigen-specific CD8(+) T cell interferon (IFN) gamma production but only modestly suppress antigen-specific and nonspecific CD4(+) T cell proliferation. GR-1(+) cell depletion in vivo prevents both the sepsis-induced augmentation of Th2 cell-dependent and depression of Th1 cell-dependent antibody production. Signaling through MyD88, but not Toll-like receptor 4, TIR domain-containing adaptor-inducing IFN-beta, or the IFN-alpha/beta receptor, is required for complete GR-1(+)CD11b(+) expansion. GR-1(+)CD11b(+) cells contribute to sepsis-induced T cell suppression and preferential Th2 polarization.


Assuntos
Antígeno CD11b/metabolismo , Tolerância Imunológica/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Células Progenitoras Mieloides/imunologia , Receptores de Quimiocinas/metabolismo , Sepse/imunologia , Células Th2/imunologia , Animais , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Citocinas/metabolismo , Citometria de Fluxo , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Progenitoras Mieloides/metabolismo , Transdução de Sinais/imunologia
3.
Rapid Commun Mass Spectrom ; 27(23): 2639-47, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24591025

RESUMO

RATIONALE: Human genetics studies in African Americans have shown a strong correlation between polymorphisms in the ApoL1 gene and chronic kidney disease (CKD). To gain further insight into the etiology of ApoL1-associated kidney diseases, the determination of circulating levels of both wild type as well as ApoL1 variants could be of significant use. To date, antibodies that discriminate between all three ApoL1 variant forms (wild type, G1 and G2) are not available. We aimed to develop a rapid method for detecting and quantifying ApoL1 variants and total levels in plasma. METHODS: Ultra-performance liquid chromatography (UPLC) and tandem mass spectrometry (MS/MS) in multiple-reaction monitoring acquisition mode was used to quantify ApoL1. RESULTS: We demonstrated that it is feasible to detect and quantify ApoL1 variants (wild type, G1 and G2), and total ApoL1 concentrations in plasma. ApoL1 genotypes determined by LC/MS agreed perfectly with the traditional method DNA sequencing for 74 human subjects. The method exhibited at least three orders of linearity with a lower limit of quantification of 10 nM. Moreover, the method can readily be multiplexed for the quantification of a panel of protein markers in a single sample. CONCLUSIONS: The method reported herein obviates the need to perform DNA genotyping of ApoL1 variants, which is of significant value in cases where stored samples are unsuitable for DNA analysis. More importantly, the method could potentially be of use in the early identification of individuals at risk of developing CKD, and for the stratification of patients for treatment with future ApoL1-modifying therapies.


Assuntos
Apolipoproteínas/sangue , Apolipoproteínas/genética , Cromatografia Líquida de Alta Pressão/métodos , Variação Genética , Nefropatias/sangue , Lipoproteínas HDL/sangue , Lipoproteínas HDL/genética , Espectrometria de Massas em Tandem/métodos , Sequência de Aminoácidos , Animais , Apolipoproteína L1 , Genótipo , Humanos , Nefropatias/diagnóstico , Nefropatias/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular
4.
J Immunol ; 187(2): 951-9, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21670317

RESUMO

The role of IL-23 in the development of arthritis and bone metabolism was studied using systemic IL-23 exposure in adult mice via hydrodynamic delivery of IL-23 minicircle DNA in vivo and in mice genetically deficient in IL-23. Systemic IL-23 exposure induced chronic arthritis, severe bone loss, and myelopoiesis in the bone marrow and spleen, which resulted in increased osteoclast differentiation and systemic bone loss. The effect of IL-23 was partly dependent on CD4(+) T cells, IL-17A, and TNF, but could not be reproduced by overexpression of IL-17A in vivo. A key role in the IL-23-induced arthritis was made by the expansion and activity of myeloid cells. Bone marrow macrophages derived from IL-23p19(-/-) mice showed a slower maturation into osteoclasts with reduced tartrate-resistant acid phosphatase-positive cells and dentine resorption capacity in in vitro osteoclastogenesis assays. This correlated with fewer multinucleated osteoclast-like cells and more trabecular bone volume and number in 26-wk-old male IL-23p19(-/-) mice compared with control animals. Collectively, our data suggest that systemic IL-23 exposure induces the expansion of a myeloid lineage osteoclast precursor, and targeting IL-23 pathway may combat inflammation-driven bone destruction as observed in rheumatoid arthritis and other autoimmune arthritides.


Assuntos
Artrite Experimental/imunologia , Artrite Experimental/patologia , Reabsorção Óssea/imunologia , Diferenciação Celular/imunologia , Subunidade p19 da Interleucina-23/fisiologia , Osteoclastos/imunologia , Osteoclastos/patologia , Animais , Artrite Experimental/genética , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Células CHO , Diferenciação Celular/genética , Doença Crônica , Cricetinae , Cricetulus , DNA de Cinetoplasto/biossíntese , DNA de Cinetoplasto/genética , Células HEK293 , Humanos , Subunidade p19 da Interleucina-23/deficiência , Subunidade p19 da Interleucina-23/isolamento & purificação , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Índice de Gravidade de Doença , Baço/imunologia , Baço/metabolismo , Baço/patologia
5.
J Immunol ; 187(2): 911-8, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21690321

RESUMO

Neutrophils are essential for successful host eradication of bacterial pathogens and for survival to polymicrobial sepsis. During inflammation, the bone marrow provides a large reserve of neutrophils that are released into the peripheral circulation where they traverse to sites of infection. Although neutrophils are essential for survival, few studies have investigated the mechanisms responsible for neutrophil mobilization from the bone marrow during polymicrobial sepsis. Using a cecal ligation and puncture model of polymicrobial sepsis, we demonstrated that neutrophil mobilization from the bone marrow is not dependent on TLR4, MyD88, TRIF, IFNARα/ß, or CXCR2 pathway signaling during sepsis. In contrast, we observed that bone marrow CXCL12 mRNA abundance and specific CXCL12 levels are sharply reduced, whereas splenic CXCR4 mRNA and cell surface expression are increased during sepsis. Blocking CXCL12 activity significantly reduced blood neutrophilia by inhibiting bone marrow release of granulocytes during sepsis. However, CXCL12 inhibition had no impact on the expansion of bone marrow neutrophil precursors and hematopoietic progenitors. Bone marrow neutrophil retention by CXCL12 blockade prevented blood neutrophilia, inhibited peritoneal neutrophil accumulation, allowed significant peritoneal bacterial invasion, and increased polymicrobial sepsis mortality. We concluded that changes in the pattern of CXCL12 signaling during sepsis are essential for neutrophil bone marrow mobilization and host survival but have little impact on bone marrow granulopoiesis.


Assuntos
Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Quimiocina CXCL12/fisiologia , Infiltração de Neutrófilos/imunologia , Sepse/imunologia , Sepse/microbiologia , Transdução de Sinais/imunologia , Doença Aguda , Animais , Células da Medula Óssea/microbiologia , Quimiocina CXCL12/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mielopoese/genética , Mielopoese/imunologia , Infiltração de Neutrófilos/genética , Sepse/mortalidade , Transdução de Sinais/genética , Análise de Sobrevida
6.
Mol Med ; 17(3-4): 281-92, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21085745

RESUMO

Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells whose numbers dramatically increase in chronic and acute inflammatory diseases, including cancer, autoimmune disease, trauma, burns and sepsis. Studied originally in cancer, these cells are potently immunosuppressive, particularly in their ability to suppress antigen-specific CD8(+) and CD4(+) T-cell activation through multiple mechanisms, including depletion of extracellular arginine, nitrosylation of regulatory proteins, and secretion of interleukin 10, prostaglandins and other immunosuppressive mediators. However, additional properties of these cells, including increased reactive oxygen species and inflammatory cytokine production, as well as their universal expansion in nearly all inflammatory conditions, suggest that MDSCs may be more of a normal component of the inflammatory response ("emergency myelopoiesis") than simply a pathological response to a growing tumor. Recent evocative data even suggest that the expansion of MDSCs in acute inflammatory processes, such as burns and sepsis, plays a beneficial role in the host by increasing immune surveillance and innate immune responses. Although clinical efforts are currently underway to suppress MDSC numbers and function in cancer to improve antineoplastic responses, such approaches may not be desirable or beneficial in other clinical conditions in which immune surveillance and antimicrobial activities are required.


Assuntos
Ativação Linfocitária/imunologia , Células Mieloides/imunologia , Sepse/imunologia , Ferimentos e Lesões/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Humanos , Modelos Imunológicos
7.
Mol Ther ; 17(3): 508-15, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19127252

RESUMO

Intravascular delivery (1.5 x 10(9) particles and higher) of recombinant adenovirus (rAd) induces myeloid cell mediated, self-limiting hemodynamic responses in normal mice. However, we observed anaphylactoid-type reactions and exacerbated hemodynamic events following rAd injection in mice bearing malignant 4T1 mammary carcinoma. Because 4T1 tumors induce significant CD11b(+)Gr-1(+) myeloid cell expansion and activation, we set to determine whether this causes rAd-induced exaggerated responses. When treated with a single intravenous dose (1 x 10(10) particles) of rAd, mice implanted with 4T1 carcinoma succumbed due to the anaphylactoid-type reactions. In contrast, normal mice and mice implanted with a related mammary carcinoma (66cl4) that does not induce CD11b(+)Gr-1(+) cell expansion, showed minimal responses. Depletion of phagocytic CD11b(+)Gr-1(+) cells prior to rAd delivery protected 4T1 tumor-bearing animals, whereas passive transfer of CD11b(+)Gr-1(+) cells from 4T1 tumor-bearing animals was sufficient to convey susceptibility to anaphylactoid-type reactions in normal animals. We further show that there is upregulation of nitric oxide and leukotriene signaling pathways in the 4T1 tumor-induced CD11b(+)Gr-1(+) myeloid cells and that pretreating mice with inhibitors of nitric oxide synthetase and leukotrienes can attenuate the anaphylactoid-type reactions. These data show that malignant tumor growth can alter CD11b(+)Gr-1(+) myeloid cells, rendering hosts susceptible to anaphylactoid-type reactions upon intravascular treatment with rAd.


Assuntos
Adenoviridae/imunologia , Anafilaxia/metabolismo , Antígeno CD11b/imunologia , Diferenciação Celular , Terapia Genética/efeitos adversos , Neoplasias/patologia , Receptores de Quimiocinas/metabolismo , Adenoviridae/genética , Anafilaxia/fisiopatologia , Animais , Sistema Cardiovascular/metabolismo , Linhagem Celular Tumoral , Suscetibilidade a Doenças , Feminino , Frequência Cardíaca , Humanos , Antagonistas de Leucotrienos/farmacologia , Lipossomos , Camundongos , Células Mieloides/metabolismo , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/terapia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Fagócitos , Receptores de Quimiocinas/imunologia , Receptores de Leucotrienos/metabolismo
8.
Front Immunol ; 11: 573405, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117369

RESUMO

The molecule "T cell immunoreceptor with immunoglobulin and ITIM domain," or TIGIT, has recently received much attention as a promising target in the treatment of various malignancies. In spite of the quick progression of anti-TIGIT antibodies into clinical testing both as monotherapy and in combination with programmed cell death-1 (PD-1)-directed immune checkpoint blockade, the molecular mechanism behind the observed therapeutic benefits remains poorly understood. Here we demonstrate, using mouse tumor models, that TIGIT blocking antibodies with functional Fc binding potential induce effective anti-tumor response. Our observations reveal that the anti-TIGIT therapeutic effect is not achieved by depletion of intratumoral regulatory T cells (Treg) or any cell population expressing TIGIT, but instead is mediated by possible "reverse activating signals" through FcγRs on myeloid cells, inducing expression of various mediators such as cytokines and chemokines. Furthermore, we discovered an induction of a robust and persistent granzyme B and perforin response, distinct from a predominantly interferon-γ (IFN-γ)-driven anti-PD-1 blockade. Our observations, for the first time, provide the basis for a mechanistic hypothesis involving the requirement of a functional Fc domain of anti-TIGIT monoclonal antibodies, of which various isotypes are currently under intense clinical investigation.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Inibidores de Checkpoint Imunológico/farmacologia , Células Mieloides/efeitos dos fármacos , Receptores de IgG/metabolismo , Receptores Imunológicos/metabolismo , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Granzimas/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral
9.
J Immunother Cancer ; 8(2)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33127658

RESUMO

BACKGROUND: Programmed cell death protein 1 (PD-1) and CTLA4 combination blockade enhances clinical efficacy in melanoma compared with targeting either checkpoint alone; however, clinical response improvement is coupled with increased risk of developing immune-related adverse events (irAE). Delineating the mechanisms of checkpoint blockade-mediated irAE has been hampered by the lack of animal models that replicate these clinical events. METHODS: We have developed a mouse model of checkpoint blockade-mediated enterocolitis via prolonged administration of an Fc-competent anti-CTLA4 antibody. RESULTS: Sustained treatment with Fc-effector, but not Fc-mutant or Fc-null, anti-CTLA4 antagonist for 7 weeks resulted in enterocolitis. Moreover, combining Fc-null or Fc-mutant CTLA4 antagonists with PD-1 blockade results in potent antitumor combination efficacy indicating that Fc-effector function is not required for combination benefit. CONCLUSION: These data suggest that using CTLA4 antagonists with no Fc-effector function can mitigate gut inflammation associated with anti-CTLA4 antibody therapy yet retain potent antitumor activity in combination with PD-1 blockade.


Assuntos
Antígeno CTLA-4/antagonistas & inibidores , Inflamação/fisiopatologia , Receptor de Morte Celular Programada 1/metabolismo , Animais , Humanos , Camundongos
11.
JPEN J Parenter Enteral Nutr ; 32(6): 651-5, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18974247

RESUMO

Cachexia accompanies many chronic inflammatory diseases, including cancer. Lean tissue wasting is only one component of the cancer cachexia response, which also includes anemia, anorexia, a hepatic acute phase protein response, and increased susceptibility to secondary infections. The etiologies of cancer cachexia are multifactorial and include an overproduction of inflammatory mediators, including cytokines produced by inappropriate activation of innate immunity. However, anticytokine therapies have generally not been seriously considered for cancer cachexia, in large part because of the overlapping activities of several inflammatory cytokines and the inability to prospectively identify the contributions of individual mediators. In contrast, recent evidence has focused on an immature myeloid cell population that expands dramatically in the tumors and secondary lymphoid organs of animals with some actively growing tumors. These immature GR-1(+)CD11b(+) cells are metabolically active and secrete large quantities of inflammatory cytokines and chemokines with the potential to produce cachexia. Their expansion is temporally associated with the development of cachexia. Future studies are required to determine whether therapeutic efforts intended to block the expansion of these cells can prevent the lean tissue wasting that accompanies active tumor growth.


Assuntos
Caquexia/etiologia , Citocinas/fisiologia , Metabolismo Energético/fisiologia , Inflamação/metabolismo , Neoplasias/metabolismo , Proteínas de Fase Aguda/metabolismo , Reação de Fase Aguda/metabolismo , Caquexia/metabolismo , Humanos , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Neoplasias/fisiopatologia
12.
Shock ; 23(6): 507-15, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15897802

RESUMO

Control of dendritic cell (DC) function is critical for strategies to modulate innate and acquired immune responses. We examined whether transduction of murine DCs with adenoviral vectors (Adv) expressing interleukin (IL)-10 could alter their phenotype and T cell stimulatory function. Murine bone marrow-derived DCs were transduced with AdV encoding human IL-10 or green fluorescent protein (GFP). Whereas transduction of immature DCs with AdV/GFP resulted in dose-dependent maturation, DCs transduced with Adv/IL-10 maintained an immature state with low major histocompatibility complex (MHC) class II, CD86, and IL-12 expression. The Adv/IL-10 transduced DCs were phenotypically unique, characterized by suppression of IL-12 expression, failure to stimulate Th1 or Th2 cytokine responses, and retained capacity to endocytose antigen. Importantly, Adv/IL-10-transduced DCs were biologically active in vivo, in that administration of these DCs into mice before a generalized peritonitis significantly improved survival. We conclude that Adv/IL-10 transduction of DCs provides an efficient means to modulate DC function. The capacity to modify DCs by adenoviral expression of IL-10 may provide a novel ex vivo or in vivo approach to mitigate acute and chronic inflammatory diseases like sepsis.


Assuntos
Adenoviridae/genética , Células Dendríticas/citologia , Interleucina-10/genética , Interleucina-10/uso terapêutico , Sepse/terapia , Adenoviridae/metabolismo , Animais , Antígenos CD/biossíntese , Antígeno B7-2 , Complexo CD3/biossíntese , Antígenos CD40/biossíntese , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Endocitose , Feminino , Citometria de Fluxo , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Interleucina-12/metabolismo , Lipopolissacarídeos/metabolismo , Linfonodos/patologia , Linfócitos/citologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Sepse/metabolismo , Linfócitos T/citologia , Células Th1 , Células Th2/metabolismo , Fatores de Tempo
13.
Clin Cancer Res ; 10(21): 7199-206, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15534093

RESUMO

PURPOSE: The purpose of this study was to assess the impact of anti-adenovirus neutralizing antibodies (AdNAbs) on the distribution, tolerability, and efficacy of intravenously administered oncolytic adenovirus. A translational model was developed to evaluate the impact of humoral immunity on intravenous administration of oncolytic adenovirus in humans. EXPERIMENTAL DESIGN: Initially, severe combined immunodeficient (SCID)/beige mice were passively immunized with various amounts of human sera to establish a condition of preexisting humoral immunity similar to humans. A replication-deficient adenovirus encoding beta-galactosidase (rAd-betagal) was injected intravenously into these mice. An AdNAb titer that mitigated galactosidase transgene expression was determined. A xenograft tumor-bearing nude mouse model was developed to assess how a similar in vivo titer would impact the activity of 01/PEME, an oncolytic adenovirus, after intravenous administration. RESULTS: In SCID/beige mice, there was a dose dependence between AdNAbs and galactosidase transgene expression; 90% of transgene expression was inhibited when the titer was 80. A similar titer reconstituted in the nude mice with human serum, as was done in the SCID/beige mice, did not abrogate the antitumor efficacy of the replicating adenovirus after intravenous administration. Viral DNA increased in tumors over time. CONCLUSIONS: In intravenous administration, preexisting AdNAb titer of 80 significantly attenuated the activity of a 2.5 x 10(12) particles per kilogram dose of nonreplicating adenovirus; the same titer had no affect on the activity of an equivalent dose of replicating adenovirus. Our results suggest that a majority of patients with preexisting adenovirus immunity would be candidates for intravenous administration of oncolytic adenovirus.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Animais , Peso Corporal , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Camundongos SCID , Transplante de Neoplasias , Reação em Cadeia da Polimerase , Fatores de Tempo , beta-Galactosidase/genética
14.
J Biotechnol ; 208: 13-21, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26015261

RESUMO

Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.


Assuntos
Anticorpos Monoclonais , Anticorpos Antineoplásicos , Engenharia Celular , Mieloma Múltiplo/tratamento farmacológico , Neoplasias Experimentais/tratamento farmacológico , Pichia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/química , Anticorpos Antineoplásicos/genética , Linhagem Celular Tumoral , Glicosilação , Células HEK293 , Humanos , Camundongos SCID , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Endotoxin Res ; 10(6): 393-401, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15588421

RESUMO

Adenovirus-based gene therapy offers a unique opportunity to target gene expression to the liver by systemic delivery. However, systemic administration of a first generation adenoviral construct elicits an inflammatory response leading to TNF-alpha-dependent liver injury. The aim of this study was to evaluate whether the systemic administration of recombinant adenovirus exacerbates a subsequent TNF-alpha-dependent liver injury induced by D-galactosamine and lipopolysaccharide. Surprisingly, low-dose adenovirus administration (10(5) particles) protects, while high-dose adenovirus (10(10) particles) is associated with an exaggerated hepatic inflammatory response from a subsequent D-galactosamine and lipopolysaccharide challenge. This exacerbation is TNF-alpha dependent, since treatment with a TNF inhibitor fully protects against the liver injury. Moreover, intravenous administration of an adenoviral construct expressing the anti-inflammatory protein interleukin-10 reduces TNF-alpha appearance and attenuates the increased hepatocyte injury. Taken together, this report demonstrates potential additive effects of TNF-alpha responses induced by adenovirus and other inflammatory signals, and suggests that the response can be mitigated by relative adenovirus particle dose or by inhibitors, such as TNF-binding protein or interleukin 10.


Assuntos
Adenoviridae/genética , Terapia Genética , Lipopolissacarídeos/toxicidade , Hepatopatias/terapia , Adenoviridae/imunologia , Animais , Apoptose/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas , Relação Dose-Resposta Imunológica , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Injeções Intraperitoneais , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolissacarídeos/administração & dosagem , Fígado/efeitos dos fármacos , Fígado/patologia , Hepatopatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
16.
Hum Immunol ; 65(11): 1344-55, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15556685

RESUMO

Recombinant adenoviruses (rAd) are efficient tools for genetic modification of human dendritic cells (DC) in vitro. Infection of DCs by rAd encoding beta-galactosidase (betagal) results in partial maturation of DCs, as witnessed by the upregulation of major histocompatibility complex and costimulatory molecules. Accordingly, these DCs are more potent stimulators of Th1-type proliferative responses. We now demonstrate that infection of immature DCs with rAd encoding human interleukin (IL)-10 results in the secretion by the DCs of large amounts of IL-10, while not affecting expression of activation markers indicative of partial DC maturation. In contrast to rAd-betagal-infected DCs, rAdIL-10-infected DCs are very poor stimulators of monoclonal and polyclonal Th1-type responses. Instead, stimulation of nonpolarized CD4+ T-cell cultures with rAdIL-10-infected DCs selectively activates and expands an IL-10-producing CD4+ T-cell subset capable of suppressing Th1 responses in vitro. Our data argue that rAd-infected human DCs genetically engineered to produce IL-10 may be exploited for the modulation of harmful Th1-type responses in transplantation and autoimmune diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Interleucina-10/genética , Ativação Linfocitária/imunologia , Células Th1/imunologia , Adenoviridae/genética , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Proliferação de Células , Citocinas/metabolismo , Células Dendríticas/metabolismo , Vetores Genéticos/genética , Humanos , Interleucina-10/metabolismo , Células Th1/metabolismo , Transfecção
17.
Cancer Res ; 72(14): 3570-81, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22581824

RESUMO

The presence of activated intratumoral T cells correlates clinically with better prognosis in patients with cancer. Although tumor vaccines can increase the number of tumor-specific CD8(+) T cells in systemic circulation, they frequently fail to increase the number of active and tumor reactive T cells within the tumor. Here we show that treatment with the pleiotropic cytokine interleukin-10 (IL-10) induces specific activation of tumor-resident CD8(+) T cells as well as their intratumoral expansion in several mouse tumor models. We found that inhibition of T-cell trafficking from lymphoid organs did not impair IL-10-induced tumor rejection or the activation of tumor-resident CD8(+) T cells. Tumor-resident CD8(+) T cells expressed elevated levels of the IL-10 receptor and were directly activated by IL-10, resulting in prominent phosphorylation of STAT3 and STAT1. Although CD4(+) T cells, regulatory T cells, NK cells, and dendritic cells have been reported as prominent targets of IL-10 in the tumor microenvironment, we found that expression of the IL-10R was required only on CD8(+) T cells to facilitate IL-10-induced tumor rejection as well as in situ expansion and proliferation of tumor-resident CD8 T cells. Together, our findings indicate that IL-10 activates CD8(+) T-cell-mediated tumor control and suggest that IL-10 may represent a potential tumor immunotherapy in human patients with cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interleucina-10/farmacologia , Ativação Linfocitária/imunologia , Neoplasias/imunologia , Animais , Feminino , Humanos , Sistema Linfático/imunologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Interleucina-10/metabolismo , Linfócitos T Reguladores/imunologia
18.
Nat Med ; 18(7): 1069-76, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22772566

RESUMO

The spondyloarthropathies are a group of rheumatic diseases that are associated with inflammation at anatomically distal sites, particularly the tendon-bone attachments (entheses) and the aortic root. Serum concentrations of interleukin-23 (IL-23) are elevated and polymorphisms in the IL-23 receptor are associated with ankyosing spondylitis, however, it remains unclear whether IL-23 acts locally at the enthesis or distally on circulating cell populations. We show here that IL-23 is essential in enthesitis and acts on previously unidentified IL-23 receptor (IL-23R)(+), RAR-related orphan receptor γt (ROR-γt)(+)CD3(+)CD4(-)CD8(-), stem cell antigen 1 (Sca1)(+) entheseal resident T cells. These cells allow entheses to respond to IL-23 in vitro-in the absence of further cellular recruitment--and to elaborate inflammatory mediators including IL-6, IL-17, IL-22 and chemokine (C-X-C motif) ligand 1 (CXCL1). Notably, the in vivo expression of IL-23 is sufficient to phenocopy the human disease, with the specific and characteristic development of enthesitis and entheseal new bone formation in the initial complete absence of synovitis. As in the human condition, inflammation also develops in vivo at the aortic root and valve, which are structurally similar to entheses. The presence of these entheseal resident cells and their production of IL-22, which activates signal transducer and activator of transcription 3 (STAT3)-dependent osteoblast-mediated bone remodeling, explains why dysregulation of IL-23 results in inflammation at this precise anatomical site.


Assuntos
Interleucina-23/imunologia , Espondiloartropatias/imunologia , Linfócitos T/imunologia , Tendões/imunologia , Animais , Antígenos CD/metabolismo , Aorta/patologia , Artrite Experimental/complicações , Artrite Experimental/imunologia , Artrite Experimental/patologia , Remodelação Óssea , Complexo CD3/metabolismo , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Modelos Animais de Doenças , Extremidades/patologia , Citometria de Fluxo , Humanos , Imunização Passiva , Inflamação/complicações , Inflamação/imunologia , Inflamação/patologia , Interleucina-17 , Interleucinas , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Osteogênese/imunologia , Periósteo/crescimento & desenvolvimento , Receptores de Interleucina/metabolismo , Espondiloartropatias/complicações , Espondiloartropatias/patologia , Tendões/patologia , Células Th17 , Interleucina 22
19.
Int Immunopharmacol ; 11(7): 780-2, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21315784

RESUMO

Recent investigations of regulatory myeloid cell mobilization and amplification in response to cancer and chronic inflammation associated with infectious disease and autoimmunity have provided numerous insights into the suppressive mechanisms, pathobiology and potential pharmaceutical modulation of MDSC. Controversies have emerged as hematology investigators, have suggested that regulatory myeloid cells, including MDSC merely reflect normal physiological responses to inflammatory and growth factor stimuli. A recurring theme at the recent "International Immunopharmacology Conference on Regulatory Myeloid cells" held in Arlington, Virginia, October 21-24, 2010, focused on efforts to more clearly elucidate molecular features of MDSC from the immature myeloid cells observed in the circulation in response to acute infection and inflammatory responses. Evidence for the potential contribution of regulatory myeloid cells to pathologic processes and sequelae associated with numerous inflammatory and malignant diseases also suggested that these cells may represent a significant deviation from the normal physiologic responses to acute inflammatory stimuli. The relevance of these research efforts was evident from the discussions by numerous clinical investigations regarding their efforts to modulate MDSC in cancer patients to augment immunotherapeutic treatments.


Assuntos
Doenças Autoimunes/imunologia , Imunoterapia , Infecções/imunologia , Células Mieloides/imunologia , Neoplasias/imunologia , Animais , Doenças Autoimunes/terapia , Biomarcadores/metabolismo , Diferenciação Celular , Humanos , Imunomodulação , Imunoterapia/tendências , Infecções/terapia , Inflamação , Camundongos , Neoplasias/terapia
20.
PLoS One ; 6(7): e22256, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21799806

RESUMO

C17 was first described about ten years ago as a gene expressed in CD34+ cells. A more recent study has suggested a role for C17 in chondrogenesis and development of cartilage. However, based on sequence analysis, we believe that C17 has homology to IL-2 and hence we present the hypothesis that C17 is a cytokine possessing immune-regulatory properties. We provide evidence that C17 is a secreted protein preferentially expressed in chondrocytes, hence in cartilage-rich tissues. Systemic expression of C17 in vivo reduces disease in a collagen antibody-induced arthritis model in mice (CAIA). Joint protection is evident by delayed disease onset, minimal edema, bone protection and absence of diverse histological features of disease. Expression of genes typically associated with acute joint inflammation and erosion of cartilage or bone is blunted in the presence of C17. Consistent with the observed reduction in bone erosion, we demonstrate reduced levels of RANKL in the paws and sera of mice over-expressing C17. Administration of C17 at the peak of disease, however, had no effect on disease progression, indicating that C17's immune-regulatory activity must be most prominent prior to or at the onset of severe joint inflammation. Based on this data we propose C17 as a cytokine that s contributes to immune homeostasis systemically or in a tissue-specific manner in the joint.


Assuntos
Artrite/metabolismo , Proteínas Sanguíneas/metabolismo , Citocinas/metabolismo , Articulações/metabolismo , Articulações/patologia , Sequência de Aminoácidos , Animais , Artrite/imunologia , Artrite/patologia , Artrite/terapia , Biomarcadores/metabolismo , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Doenças Ósseas/complicações , Doenças Ósseas/metabolismo , Cartilagem/metabolismo , Condrócitos/metabolismo , Citocinas/química , Citocinas/genética , Regulação da Expressão Gênica , Células HEK293 , Homeostase/imunologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/terapia , Articulações/imunologia , Masculino , Camundongos , Dados de Sequência Molecular , Ligante RANK/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA