Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 11(12)2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-38164757

RESUMO

INTRODUCTION: The clinical benefit of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab has been well established but limited by immune-related adverse events, especially when ipilimumab is used in combination with anti-PD-(L)1 mAb therapy. To overcome these limitations, we have developed XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 mAb. METHODS: XTX101 consists of an anti-human CTLA-4 mAb covalently linked to masking peptides that block the complementarity-determining regions, thereby minimizing the mAb binding to CTLA-4. The masking peptides are designed to be released by proteases that are typically dysregulated within the tumor microenvironment (TME), resulting in activation of XTX101 intratumorally. Mutations within the Fc region of XTX101 were included to enhance affinity for FcγRIII, which is expected to enhance potency through antibody-dependent cellular cytotoxicity. RESULTS: Biophysical, biochemical, and cell-based assays demonstrate that the function of XTX101 depends on proteolytic activation. In human CTLA-4 transgenic mice, XTX101 monotherapy demonstrated significant tumor growth inhibition (TGI) including complete responses, increased intratumoral CD8+T cells, and regulatory T cell depletion within the TME while maintaining minimal pharmacodynamic effects in the periphery. XTX101 in combination with anti-PD-1 mAb treatment resulted in significant TGI and was well tolerated in mice. XTX101 was activated in primary human tumors across a range of tumor types including melanoma, renal cell carcinoma, colon cancer and lung cancer in an ex vivo assay system. CONCLUSIONS: These data demonstrate that XTX101 retains the full potency of an Fc-enhanced CTLA-4 antagonist within the TME while minimizing the activity in non-tumor tissue, supporting the further evaluation of XTX101 in clinical studies.


Assuntos
Antineoplásicos , Melanoma , Humanos , Camundongos , Animais , Antígeno CTLA-4 , Ipilimumab/uso terapêutico , Antineoplásicos/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Melanoma/tratamento farmacológico , Modelos Animais de Doenças , Camundongos Transgênicos , Peptídeos/uso terapêutico , Microambiente Tumoral
2.
ACS Chem Biol ; 14(4): 806-818, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30875193

RESUMO

Drug discovery research on new pain targets with human genetic validation, including the voltage-gated sodium channel NaV1.7, is being pursued to address the unmet medical need with respect to chronic pain and the rising opioid epidemic. As part of early research efforts on this front, we have previously developed NaV1.7 inhibitory peptide-antibody conjugates with tarantula venom-derived GpTx-1 toxin peptides with an extended half-life (80 h) in rodents but only moderate in vitro activity (hNaV1.7 IC50 = 250 nM) and without in vivo activity. We identified the more potent peptide JzTx-V from our natural peptide collection and improved its selectivity against other sodium channel isoforms through positional analogueing. Here we report utilization of the JzTx-V scaffold in a peptide-antibody conjugate and architectural variations in the linker, peptide loading, and antibody attachment site. We found conjugates with 100-fold improved in vitro potency relative to those of complementary GpTx-1 analogues, but pharmacokinetic and bioimaging analyses of these JzTx-V conjugates revealed a shorter than expected plasma half-life in vivo with accumulation in the liver. In an attempt to increase circulatory serum levels, we sought the reduction of the net +6 charge of the JzTx-V scaffold while retaining a desirable NaV in vitro activity profile. The conjugate of a JzTx-V peptide analogue with a +2 formal charge maintained NaV1.7 potency with 18-fold improved plasma exposure in rodents. Balancing the loss of peptide and conjugate potency associated with the reduction of net charge necessary for improved target exposure resulted in a compound with moderate activity in a NaV1.7-dependent pharmacodynamic model but requires further optimization to identify a conjugate that can fully engage NaV1.7 in vivo.


Assuntos
Imunoconjugados , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Peptídeos/química , Venenos de Aranha/química , Bloqueadores do Canal de Sódio Disparado por Voltagem , Animais , Anticorpos/química , Descoberta de Drogas , Humanos , Imunoconjugados/química , Imunoconjugados/farmacocinética , Masculino , Camundongos , Terapia de Alvo Molecular , Canal de Sódio Disparado por Voltagem NAV1.7/imunologia , Peptídeos/farmacocinética , Venenos de Aranha/farmacocinética , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacocinética
3.
Proc Natl Acad Sci U S A ; 111(14): 5153-8, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24706856

RESUMO

Mice genetically engineered to be humanized for their Ig genes allow for human antibody responses within a mouse background (HumAb mice), providing a valuable platform for the generation of fully human therapeutic antibodies. Unfortunately, existing HumAb mice do not have fully functional immune systems, perhaps because of the manner in which their genetic humanization was carried out. Heretofore, HumAb mice have been generated by disrupting the endogenous mouse Ig genes and simultaneously introducing human Ig transgenes at a different and random location; KO-plus-transgenic humanization. As we describe in the companion paper, we attempted to make mice that more efficiently use human variable region segments in their humoral responses by precisely replacing 6 Mb of mouse Ig heavy and kappa light variable region germ-line gene segments with their human counterparts while leaving the mouse constant regions intact, using a unique in situ humanization approach. We reasoned the introduced human variable region gene segments would function indistinguishably in their new genetic location, whereas the retained mouse constant regions would allow for optimal interactions and selection of the resulting antibodies within the mouse environment. We show that these mice, termed VelocImmune mice because they were generated using VelociGene technology, efficiently produce human:mouse hybrid antibodies (that are rapidly convertible to fully human antibodies) and have fully functional humoral immune systems indistinguishable from those of WT mice. The efficiency of the VelocImmune approach is confirmed by the rapid progression of 10 different fully human antibodies into human clinical trials.


Assuntos
Formação de Anticorpos , Genes de Imunoglobulinas , Alelos , Animais , Linfócitos B/imunologia , Citometria de Fluxo , Humanos , Camundongos , Mutação
4.
Proc Natl Acad Sci U S A ; 111(14): 5147-52, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24706858

RESUMO

Genetic humanization, which involves replacing mouse genes with their human counterparts, can create powerful animal models for the study of human genes and diseases. One important example of genetic humanization involves mice humanized for their Ig genes, allowing for human antibody responses within a mouse background (HumAb mice) and also providing a valuable platform for the generation of fully human antibodies as therapeutics. However, existing HumAb mice do not have fully functional immune systems, perhaps because of the manner in which they were genetically humanized. Heretofore, most genetic humanizations have involved disruption of the endogenous mouse gene with simultaneous introduction of a human transgene at a new and random location (so-called KO-plus-transgenic humanization). More recent efforts have attempted to replace mouse genes with their human counterparts at the same genetic location (in situ humanization), but such efforts involved laborious procedures and were limited in size and precision. We describe a general and efficient method for very large, in situ, and precise genetic humanization using large compound bacterial artificial chromosome-based targeting vectors introduced into mouse ES cells. We applied this method to genetically humanize 3-Mb segments of both the mouse heavy and κ light chain Ig loci, by far the largest genetic humanizations ever described. This paper provides a detailed description of our genetic humanization approach, and the companion paper reports that the humoral immune systems of mice bearing these genetically humanized loci function as efficiently as those of WT mice.


Assuntos
Genes de Imunoglobulinas , Animais , Cromossomos Artificiais Bacterianos , Células-Tronco Embrionárias/imunologia , Recombinação Homóloga , Humanos , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Transgenes
6.
Sci Transl Med ; 4(162): 162ra153, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197570

RESUMO

Fibroblast growth factor 21 (FGF21) is a distinctive member of the FGF family with potent beneficial effects on lipid, body weight, and glucose metabolism and has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to native FGF21, we have developed a monoclonal antibody, mimAb1, that binds to ßKlotho with high affinity and specifically activates signaling from the ßKlotho/FGFR1c (FGF receptor 1c) receptor complex. In obese cynomolgus monkeys, injection of mimAb1 led to FGF21-like metabolic effects, including decreases in body weight, plasma insulin, triglycerides, and glucose during tolerance testing. Mice with adipose-selective FGFR1 knockout were refractory to FGF21-induced improvements in glucose metabolism and body weight. These results in obese monkeys (with mimAb1) and in FGFR1 knockout mice (with FGF21) demonstrated the essential role of FGFR1c in FGF21 function and suggest fat as a critical target tissue for the cytokine and antibody. Because mimAb1 depends on ßKlotho to activate FGFR1c, it is not expected to induce side effects caused by activating FGFR1c alone. The unexpected finding of an antibody that can activate FGF21-like signaling through cell surface receptors provided preclinical validation for an innovative therapeutic approach to diabetes and obesity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Diabetes Mellitus/tratamento farmacológico , Fatores de Crescimento de Fibroblastos/imunologia , Glucuronidase/metabolismo , Obesidade/tratamento farmacológico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Anticorpos Monoclonais/farmacologia , Peso Corporal/genética , Diabetes Mellitus/sangue , Epitopos/química , Glucose/metabolismo , Glucuronidase/imunologia , Humanos , Proteínas Klotho , Macaca fascicularis , Camundongos , Obesidade/sangue , Obesidade/complicações , Fosfatos/sangue , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/agonistas , Transdução de Sinais/efeitos dos fármacos , Triglicerídeos/sangue
7.
Nat Immunol ; 13(2): 129-35, 2011 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-22197977

RESUMO

The importance of immunoproteasomes to antigen presentation has been unclear because animals totally lacking immunoproteasomes had not been available. Having now developed mice lacking the three immunoproteasome catalytic subunits, we found that the dendritic cells of these mice had defects in presenting several major histocompatibility complex (MHC) class I epitopes. During viral infection in vivo, the presentation of a majority of MHC class I epitopes was markedly reduced in immunoproteasome-deficient animals compared with wild-type animals, whereas presentation of MHC class II peptides was unaffected. According to mass spectrometry, the repertoire of MHC class I-presented peptides was ∼50% different from that in wild-type mice, and these differences were sufficient to stimulate robust transplant rejection of wild-type cells in mutant mice. These results indicated that immunoproteasomes were more important in antigen presentation than previously thought.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Animais , Apresentação de Antígeno/genética , Células Dendríticas/imunologia , Epitopos/imunologia , Feminino , Rejeição de Enxerto/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/genética
8.
J Pharmacol Sci ; 115(2): 105-11, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21157117

RESUMO

The cytokine, interleukin (IL)-19, is a member of the IL-10 family that includes IL-20, IL-22, IL-24, and IL-26. Recent studies have shown that IL-19 is produced by keratinocytes, epithelial cells, macrophages, and B-cells. Little is known about the exact biological role of IL-19 in immunological regulation, although there is an increasing body of data demonstrating that IL-19 is associated with the development of Th2 responses and the pathogenesis of psoriasis. In this review, I shall attempt to discuss current knowledge about the role of IL-19 on macrophages and the potential role in inflammatory bowel disease.


Assuntos
Colo/imunologia , Imunidade Inata , Fatores Imunológicos/imunologia , Doenças Inflamatórias Intestinais/imunologia , Interleucinas/imunologia , Macrófagos/imunologia , Animais , Linfócitos B/imunologia , Humanos , Psoríase/imunologia , Psoríase/fisiopatologia
9.
J Pharmacol Sci ; 115(2): 105-111, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-32272526

RESUMO

The cytokine, interleukin (IL)-19, is a member of the IL-10 family that includes IL-20, IL-22, IL-24, and IL-26. Recent studies have shown that IL-19 is produced by keratinocytes, epithelial cells, macrophages, and B-cells. Little is known about the exact biological role of IL-19 in immunological regulation, although there is an increasing body of data demonstrating that IL-19 is associated with the development of Th2 responses and the pathogenesis of psoriasis. In this review, I shall attempt to discuss current knowledge about the role of IL-19 on macrophages and the potential role in inflammatory bowel disease.

10.
Inflamm Bowel Dis ; 16(6): 1017-28, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19834971

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) results from the chronic dysregulation of the mucosal immune system and the aberrant activation of both the innate and the adaptive immune responses. We used two complementary models of colonic inflammation to examine the roles of interleukin (IL)-19 in colonic inflammation and thus its possible role in IBD. METHODS: Using gene-targeting, we generated IL-19-deficient mice. To study the activation of the innate immune response during colonic inflammation we characterized an innate immune-mediated model of colitis induced by dextran sulfate sodium (DSS). DSS can induce not only acute colitis but also chronic colitis. In addition to the acute DSS-induced colitis model, we used a chronic DSS-induced colitis model that is associated with the activation of both Th1 and Th2 cytokines as well as innate immune response in the colon. RESULTS: We show that IL-19-deficient mice are more susceptible to experimental acute colitis induced by DSS, and this increased susceptibility is correlated with the accumulation of macrophages and the increased production of IFN-gamma, IL-1beta, IL-6, IL-12, TNF-alpha, and KC. Additionally, cytokine production in IL-19-deficient macrophages was enhanced on stimulation of lipopolysaccharide (LPS) through reduced phosphorylation of STAT1 and STAT3. Moreover, our results clearly demonstrate that IL-19 is required for B-cell infiltration during chronic DSS-induced colitis, which may be mediated by IL-13 and IL-6. CONCLUSIONS: The finding that IL-19 drives pathogenic innate immune responses in the colon suggests that the selective targeting of IL-19 may be an effective therapeutic approach in the treatment of human IBD.


Assuntos
Colite/imunologia , Imunidade Inata , Interleucina-10/fisiologia , Imunidade Adaptativa , Animais , Colite/genética , Citocinas/análise , Sulfato de Dextrana/imunologia , Modelos Animais de Doenças , Predisposição Genética para Doença , Interleucina-10/genética , Interleucina-10/farmacologia , Interleucinas , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th1/imunologia , Células Th2/imunologia
11.
J Exp Med ; 206(4): 937-52, 2009 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-19349464

RESUMO

Differentiation and recruitment of alternatively activated macrophages (AAMacs) are hallmarks of several inflammatory conditions associated with infection, allergy, diabetes, and cancer. AAMacs are defined by the expression of Arginase 1, chitinase-like molecules, and resistin-like molecule (RELM) alpha/FIZZ1; however, the influence of these molecules on the development, progression, or resolution of inflammatory diseases is unknown. We describe the generation of RELM-alpha-deficient (Retnla(-/-)) mice and use a model of T helper type 2 (Th2) cytokine-dependent lung inflammation to identify an immunoregulatory role for RELM-alpha. After challenge with Schistosoma mansoni (Sm) eggs, Retnla(-/-) mice developed exacerbated lung inflammation compared with their wild-type counterparts, characterized by excessive pulmonary vascularization, increased size of egg-induced granulomas, and elevated fibrosis. Associated with increased disease severity, Sm egg-challenged Retnla(-/-) mice exhibited elevated expression of pathogen-specific CD4(+) T cell-derived Th2 cytokines. Consistent with immunoregulatory properties, recombinant RELM-alpha could bind to macrophages and effector CD4(+) Th2 cells and inhibited Th2 cytokine production in a Bruton's tyrosine kinase-dependent manner. Additionally, Retnla(-/-) AAMacs promoted exaggerated antigen-specific Th2 cell differentiation. Collectively, these data identify a previously unrecognized role for AAMac-derived RELM-alpha in limiting the pathogenesis of Th2 cytokine-mediated pulmonary inflammation, in part through the regulation of CD4(+) T cell responses.


Assuntos
Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Pneumopatias/imunologia , Ativação de Macrófagos , Animais , Linfócitos T CD4-Positivos/imunologia , Regulação da Expressão Gênica , Genes Reporter , Granuloma/genética , Granuloma/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Lectinas Tipo C/imunologia , Ativação Linfocitária , Receptor de Manose , Lectinas de Ligação a Manose/imunologia , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/imunologia
12.
J Immunol ; 181(7): 4709-15, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802073

RESUMO

The secreted goblet cell-derived protein resistin-like molecule beta (RELMbeta) has been implicated in divergent functions, including a direct effector function against parasitic helminths and a pathogenic function in promoting inflammation in models of colitis and ileitis. However, whether RELMbeta influences CD4(+) T cell responses in the intestine is unknown. Using a natural model of intestinal inflammation induced by chronic infection with gastrointestinal helminth Trichuris muris, we identify dual functions for RELMbeta in augmenting CD4(+) Th1 cell responses and promoting infection-induced intestinal inflammation. Following exposure to low-dose Trichuris, wild-type C57BL/6 mice exhibit persistent infection associated with robust IFN-gamma production and intestinal inflammation. In contrast, infected RELMbeta(-/-) mice exhibited a significantly reduced expression of parasite-specific CD4(+) T cell-derived IFN-gamma and TNF-alpha and failed to develop Trichuris-induced intestinal inflammation. In in vitro T cell differentiation assays, recombinant RELMbeta activated macrophages to express MHC class II and secrete IL-12/23p40 and enhanced their ability to mediate Ag-specific IFN-gamma expression in CD4(+) T cells. Taken together, these data suggest that goblet cell-macrophage cross-talk, mediated in part by RELMbeta, can promote adaptive CD4(+) T cell responses and chronic inflammation following intestinal helminth infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Caliciformes/imunologia , Hormônios Ectópicos/fisiologia , Mediadores da Inflamação/fisiologia , Interferon gama/biossíntese , Enteropatias Parasitárias/imunologia , Enteropatias Parasitárias/patologia , Tricuríase/imunologia , Doença Aguda , Adjuvantes Imunológicos/fisiologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular/imunologia , Doença Crônica , Células Caliciformes/metabolismo , Hormônios Ectópicos/genética , Peptídeos e Proteínas de Sinalização Intercelular , Enteropatias Parasitárias/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Resistina/genética , Resistina/fisiologia , Tricuríase/metabolismo , Tricuríase/patologia , Trichuris/imunologia
13.
J Immunol ; 180(3): 1704-12, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18209067

RESUMO

Previous experiments using enzyme inhibitors, cell lysates, and purified enzyme have suggested that puromycin-sensitive aminopeptidase (PSA) plays a role in creating and destroying MHC class I-presented peptides although its precise contribution to these processes is unknown. To examine the importance of this enzyme in MHC class I Ag presentation, we have generated PSA-deficient mice and cell lines from these animals. PSA-deficient mice are smaller and do not reproduce as well as wild type mice. In addition, dendritic cells from PSA-deficient mice display more MHC class I molecules on the cell surface, suggesting that PSA normally limits Ag presentation by destroying certain peptides in these key APCs. Surprisingly, MHC class I levels are not altered on other PSA-deficient cells and the processing and presentation of peptide precursors in PSA-deficient fibroblasts is normal. Moreover, PSA-deficient mice have normal numbers of T cells in the periphery, and respond as well as wild type mice to eight epitopes from three viruses. These data indicate that PSA may play a role in limiting MHC class I Ag presentation in dendritic cells in vivo but that it is not essential for generating most MHC class I-presented peptides or for stimulating CTL responses to several Ags.


Assuntos
Aminopeptidases/fisiologia , Apresentação de Antígeno , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Sequência de Aminoácidos , Aminopeptidases/genética , Animais , Linfócitos T CD8-Positivos/enzimologia , Células Dendríticas/enzimologia , Epitopos/imunologia , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Peptídeos/imunologia , Viroses/imunologia , Vírus/imunologia
14.
Immunity ; 27(4): 647-59, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17919941

RESUMO

The cytokine interleukin-22 (IL-22) is primarily expressed by T helper 17 (Th17) CD4(+) T cells and is highly upregulated during chronic inflammatory diseases. IL-22 receptor expression is absent on immune cells, but is instead restricted to the tissues, providing signaling directionality from the immune system to the tissues. However, the role of IL-22 in inflammatory responses has been confounded by data suggesting both pro- and anti-inflammatory functions. Herein, we provide evidence that during inflammation, IL-22 played a protective role in preventing tissue injury. Hepatocytes from mice deficient in IL-22 were highly sensitive to the detrimental immune response associated with hepatitis. Additionally, IL-22-expressing Th17 cells provided protection during hepatitis in IL-22-deficient mice. On the other hand, interleukin-17 (IL-17), which is coexpressed with IL-22 and can induce similar cellular responses, had no observable role in liver inflammation. Our data suggest that IL-22 serves as a protective molecule to counteract the destructive nature of the immune response to limit tissue damage.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Hepatite/imunologia , Hepatócitos/imunologia , Interleucina-17/imunologia , Interleucinas/imunologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Hepatite/metabolismo , Hepatócitos/metabolismo , Interleucina-17/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Interleucinas/metabolismo , Camundongos , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/metabolismo , Interleucina 22
15.
Am J Physiol Lung Cell Mol Physiol ; 293(2): L305-13, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17545488

RESUMO

Resistin-like molecule (RELM)-beta is a cysteine-rich cytokine implicated in insulin resistance and asthmatic responses, but its function remains an enigma. We now report that RELM-beta has a role in promoting airway inflammation and lung remodeling in the mouse lung. RELM-beta is strongly induced by diverse allergens and T helper type 2 (Th2) cytokines by an IL-13- and STAT6-dependent mechanism. To understand the in vivo role of RELM-beta, we delivered recombinant murine RELM-beta intratracheally to naïve mice. RELM-beta induced dose-dependent leukocyte accumulation (most prominently involving macrophages) and goblet cell hyperplasia. The most prominent effect induced by RELM-beta was increased perivascular and peribronchial collagen deposition. Mice genetically deficient in RELM-beta had reduced accumulation of collagen and goblet cell hyperplasia in an experimental model of allergic airway inflammation. In vitro experiments demonstrated that RELM-beta had fibroblast motogenic activity. These results identify RELM-beta as a Th2-associated cytokine with potent inflammatory and remodeling activity.


Assuntos
Alérgenos/imunologia , Hormônios Ectópicos/imunologia , Pulmão/imunologia , Pneumonia/imunologia , Alérgenos/metabolismo , Animais , Asma/imunologia , Asma/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Movimento Celular , Colágeno/metabolismo , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Células Caliciformes/imunologia , Hormônios Ectópicos/genética , Hormônios Ectópicos/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Células NIH 3T3 , Pneumonia/metabolismo , Pneumonia/patologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Fator de Transcrição STAT6/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Células Th2/metabolismo
16.
Cell ; 126(5): 941-54, 2006 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16959573

RESUMO

Sir2 is an NAD-dependent deacetylase that connects metabolism with longevity in yeast, flies, and worms. Mammals have seven Sir2 homologs (SIRT1-7). We show that SIRT4 is a mitochondrial enzyme that uses NAD to ADP-ribosylate and downregulate glutamate dehydrogenase (GDH) activity. GDH is known to promote the metabolism of glutamate and glutamine, generating ATP, which promotes insulin secretion. Loss of SIRT4 in insulinoma cells activates GDH, thereby upregulating amino acid-stimulated insulin secretion. A similar effect is observed in pancreatic beta cells from mice deficient in SIRT4 or on the dietary regimen of calorie restriction (CR). Furthermore, GDH from SIRT4-deficient or CR mice is insensitive to phosphodiesterase, an enzyme that cleaves ADP-ribose, suggesting the absence of ADP-ribosylation. These results indicate that SIRT4 functions in beta cell mitochondria to repress the activity of GDH by ADP-ribosylation, thereby downregulating insulin secretion in response to amino acids, effects that are alleviated during CR.


Assuntos
Glutamato Desidrogenase/metabolismo , Células Secretoras de Insulina/enzimologia , Ilhotas Pancreáticas/enzimologia , Sirtuínas/metabolismo , Adenosina Difosfato Ribose/metabolismo , Aminoácidos/farmacologia , Animais , Restrição Calórica , Linhagem Celular Tumoral , Regulação para Baixo , Glucose/farmacologia , Glutamato Desidrogenase/genética , Glutamina/farmacologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mitocôndrias/enzimologia , NAD/metabolismo , Processamento de Proteína Pós-Traducional , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Sirtuína 1 , Sirtuínas/genética , Transfecção
17.
J Immunol ; 177(5): 2760-4, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16920909

RESUMO

Although IL-12 and IL-23 share the common p40 subunit, IL-23, rather than IL-12, seems to drive the pathogenesis of experimental autoimmune encephalomyelitis and arthritis, because IL-23/p19 knockout mice are protected from disease. In contrast, we describe in this study that newly created LacZ knockin mice deficient for IL-23 p19 were highly susceptible for the development of experimental T cell-mediated TNBS colitis and showed even more severe colitis than wild-type mice by endoscopic and histologic criteria. Subsequent studies revealed that dendritic cells from p19-deficient mice produce elevated levels of IL-12, and that IL-23 down-regulates IL-12 expression upon TLR ligation. Finally, in vivo blockade of IL-12 p40 in IL-23-deficient mice rescued mice from lethal colitis. Taken together, our data identify cross-regulation of IL-12 expression by IL-23 as novel key regulatory pathway during initiation of T cell dependent colitis.


Assuntos
Colite/metabolismo , Interleucina-12/biossíntese , Interleucinas/metabolismo , Linfócitos T/metabolismo , Animais , Células Cultivadas , Colite/genética , Colite/patologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Regulação para Baixo , Interleucina-23 , Subunidade p19 da Interleucina-23 , Interleucinas/genética , Camundongos , Camundongos Transgênicos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Taxa de Sobrevida
18.
J Allergy Clin Immunol ; 118(1): 257-68, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16815164

RESUMO

BACKGROUND: Resistin-like molecule (RELM) beta is a cysteine-rich cytokine expressed in the gastrointestinal tract and implicated in insulin resistance and gastrointestinal nematode immunity; however, its function primarily remains an enigma. OBJECTIVE: We sought to elucidate the function of RELM-beta in the gastrointestinal tract. METHODS: We generated RELM-beta gene-targeted mice and examined colonic epithelial barrier function, gene expression profiles, and susceptibility to acute colonic inflammation. RESULTS: We show that RELM-beta is constitutively expressed in the colon by goblet cells and enterocytes and has a role in homeostasis, as assessed by alterations in colon mRNA transcripts and epithelial barrier function in the absence of RELM-beta. Using acute colonic inflammatory models, we demonstrate that RELM-beta has a central role in the regulation of susceptibility to colonic inflammation. Mechanistic studies identify that RELM-beta regulates expression of type III regenerating gene (REG) (REG3beta and gamma), molecules known to influence nuclear factor kappaB signaling. CONCLUSIONS: These data define a critical role for RELM-beta in the maintenance of colonic barrier function and gastrointestinal innate immunity. CLINICAL IMPLICATIONS: These findings identify RELM-beta as an important molecule in homeostatic gastrointestinal function and colonic inflammation, and as such, these results have implications for a variety of human inflammatory gastrointestinal conditions, including allergic gastroenteropathies.


Assuntos
Colite/etiologia , Colo/fisiologia , Hormônios Ectópicos/fisiologia , Animais , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-13/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas a Pancreatite , Permeabilidade , Proteínas/genética
19.
Cell ; 124(2): 315-29, 2006 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-16439206

RESUMO

The Sir2 histone deacetylase functions as a chromatin silencer to regulate recombination, genomic stability, and aging in budding yeast. Seven mammalian Sir2 homologs have been identified (SIRT1-SIRT7), and it has been speculated that some may have similar functions to Sir2. Here, we demonstrate that SIRT6 is a nuclear, chromatin-associated protein that promotes resistance to DNA damage and suppresses genomic instability in mouse cells, in association with a role in base excision repair (BER). SIRT6-deficient mice are small and at 2-3 weeks of age develop abnormalities that include profound lymphopenia, loss of subcutaneous fat, lordokyphosis, and severe metabolic defects, eventually dying at about 4 weeks. We conclude that one function of SIRT6 is to promote normal DNA repair, and that SIRT6 loss leads to abnormalities in mice that overlap with aging-associated degenerative processes.


Assuntos
Envelhecimento/metabolismo , Doenças Genéticas Inatas/genética , Instabilidade Genômica , Sirtuínas/genética , Sirtuínas/fisiologia , Animais , Proliferação de Células , Cromatina/metabolismo , Dano ao DNA , Reparo do DNA , Doenças Genéticas Inatas/patologia , Humanos , Antígeno Ki-1/metabolismo , Linfócitos/imunologia , Camundongos , Camundongos Knockout , Fenótipo , Tolerância a Radiação , Transdução de Sinais , Sirtuínas/deficiência
20.
J Immunol ; 175(10): 6605-14, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16272315

RESUMO

To detect viral infections and tumors, CD8+ T lymphocytes monitor cells for the presence of antigenic peptides bound to MHC class I molecules. The majority of MHC class I-presented peptides are generated from the cleavage of cellular and viral proteins by the ubiquitin-proteasome pathway. Many of the oligopeptides produced by this process are too long to stably bind to MHC class I molecules and require further trimming for presentation. Leucine aminopeptidase (LAP) is an IFN-inducible cytosolic aminopeptidase that can trim precursor peptides to mature epitopes and has been thought to play an important role in Ag presentation. To examine the role of LAP in generating MHC class I peptides in vivo, we generated LAP-deficient mice and LAP-deficient cell lines. These mutant mice and cells are viable and grow normally. The trimming of peptides in LAP-deficient cells is not reduced under basal conditions or after stimulation with IFN. Similarly, there is no reduction in presentation of peptides from precursor or full-length Ag constructs or in the overall supply of peptides from cellular proteins to MHC class I molecules even after stimulation with IFN. After viral infection, LAP-deficient mice generate normal CTL responses to seven epitopes from three different viruses. These data demonstrate that LAP is not an essential enzyme for generating most MHC class I-presented peptides and reveal redundancy in the function of cellular aminopeptidases.


Assuntos
Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe I/metabolismo , Leucil Aminopeptidase/metabolismo , Sequência de Aminoácidos , Animais , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/metabolismo , Sequência de Bases , Linhagem Celular , Citosol/imunologia , Citosol/metabolismo , Epitopos/química , Epitopos/metabolismo , Células HeLa , Antígenos de Histocompatibilidade Classe I/química , Humanos , Leucil Aminopeptidase/deficiência , Leucil Aminopeptidase/genética , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/imunologia , Peptídeos/metabolismo , RNA Interferente Pequeno/genética , Linfócitos T Citotóxicos/enzimologia , Linfócitos T Citotóxicos/imunologia , Transfecção , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...