Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Sci Adv ; 9(25): eade7890, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37352349

RESUMEN

Peptides from degradation of intracellular proteins are continuously displayed by major histocompatibility complex (MHC) class I. To better understand origins of these peptides, we performed a comprehensive census of the class I peptide repertoire in the presence and absence of ubiquitin-proteasome system (UPS) activity upon developing optimized methodology to enrich for and quantify these peptides. Whereas most class I peptides are dependent on the UPS for their generation, a surprising 30%, enriched in peptides of mitochondrial origin, appears independent of the UPS. A further ~10% of peptides were found to be dependent on the proteasome but independent of ubiquitination for their generation. Notably, clinically achievable partial inhibition of the proteasome resulted in display of atypical peptides. Our results suggest that generation of MHC class I•peptide complexes is more complex than previously recognized, with UPS-dependent and UPS-independent components; paradoxically, alternative protein degradation pathways also generate class I peptides when canonical pathways are impaired.


Asunto(s)
Presentación de Antígeno , Complejo de la Endopetidasa Proteasomal , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Antígenos de Histocompatibilidad Clase I/metabolismo , Péptidos/metabolismo , Ubiquitina/metabolismo
2.
Cytotherapy ; 23(2): 131-136, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33309258

RESUMEN

BACKGROUND AIMS: To investigate the feasibility of using CD4 + T cells genetically modified to express an allo-HLA directed CAR and FOXP3 to suppress T cell proliferation and cytokine secretion in GvHD. METHODS: Human CD4+ T cells from A*02:01 negative donors were transduced to express A*02 CAR and FOXP3 and co-cultured in mixed lymphocyte reaction assays to demonstrate T cell suppression. A*02- CAR/FOXP CD4+ T cells were then injected into mice engrafted with allogeneic T cells in a GvHD mouse model. RESULTS: CD4+ T cells genetically modified to express allo-HLA-directed CAR and FOXP3 proliferate rapidly, downregulate CD127 and interferon-γ, express high CD25 and Helios and convert to a stable antigen-dependent suppressive phenotype. In mixed lymphocyte reaction assays, these cells potently suppressed T-cell proliferation and secreted IL-10. In a graft-versus-host disease model, A*02-CAR/FOXP3 CD4+ T cells outperformed polyclonal Tregs by reducing liver and lung inflammation, inhibiting pro-inflammatory cytokine production and limiting grafted CD3+ T-cell expansion. CONCLUSIONS: CD4 + T cells expressing allo-antigen directed HLA-specific CAR and FOXP3 act as potent, specific and stable suppressors of inflammation that out-perform their Treg counterparts both in vitro and in vivo.


Asunto(s)
Linfocitos T CD4-Positivos , Enfermedad Injerto contra Huésped , Receptores Quiméricos de Antígenos , Linfocitos T Reguladores , Animales , Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/genética , Antígenos HLA-A , Humanos , Ratones , Fenotipo , Linfocitos T Reguladores/inmunología
3.
NPJ Vaccines ; 5(1): 106, 2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33298945

RESUMEN

Cancer vaccines using synthetic long peptides (SLP) targeting tumor antigens have been tested in the clinic but the outcomes have been unimpressive, perhaps because these peptides elicit predominantly CD4+ T cell responses. We hypothesized that enhanced delivery of peptide antigens to, and uptake in, secondary lymphoid tissues should elicit more robust CD8+ and CD4+ T cell responses and improved anti-tumor responses. Here, we have designed SLP-containing cationic lipoplexes (SLP-Lpx) that improve delivery of peptides to myeloid cells in the spleen and lymphatics. Using the G12D KRAS mutations as neoantigens, we found that vaccination of mice with naked synthetic peptides harboring the G12D mutation with CpG adjuvant stimulated mainly CD4+ T cell responses with limited tumor growth inhibition. On the other hand, immunization with SLP-Lpx stimulated both CD4+ and CD8+ T cells and suppressed tumor growth in a CD8+ T cell-dependent manner. Combination of the SLP-Lpx vaccines with a checkpoint inhibitor led to profound growth suppression of established tumors. These studies suggest that preferential targeting of peptides derived from neoantigens to the spleen via lipoplexes elicits potent CD4+ and CD8+ T cell responses that inhibit tumor growth.

4.
Front Pharmacol ; 10: 51, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30837866

RESUMEN

There is an unmet need in severe asthma where approximately 40% of patients exhibit poor ß-agonist responsiveness, suffer daily symptoms and show frequent exacerbations. Antagonists of the Ca2+-activated Cl- channel, TMEM16A, offers a new mechanism to bronchodilate airways and block the multiple contractiles operating in severe disease. To identify TMEM16A antagonists we screened a library of ∼580,000 compounds. The anthelmintics niclosamide, nitazoxanide, and related compounds were identified as potent TMEM16A antagonists that blocked airway smooth muscle depolarization and contraction. To evaluate whether TMEM16A antagonists resist use- and inflammatory-desensitization pathways limiting ß-agonist action, we tested their efficacy under harsh conditions using maximally contracted airways or airways pretreated with a cytokine cocktail. Stunningly, TMEM16A antagonists fully bronchodilated airways, while the ß-agonist isoproterenol showed only partial effects. Thus, antagonists of TMEM16A and repositioning of niclosamide and nitazoxanide represent an important additional treatment for patients with severe asthma and COPD that is poorly controlled with existing therapies. It is of note that drug repurposing has also attracted wide interest in niclosamide and nitazoxanide as a new treatment for cancer and infectious disease. For the first time we identify TMEM16A as a molecular target for these drugs and thus provide fresh insights into their mechanism for the treatment of these disorders in addition to respiratory disease.

5.
PLoS One ; 12(7): e0180190, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28727837

RESUMEN

Inflammatory bowel disease (IBD) is associated with a loss of intestinal barrier function and dysregulated immune responses. It has been shown that short chain fatty acids (SCFAs) are protective in IBD and that GPR43 mediates the protective effects of SCFAs. In this study, we investigated the effects of SCFAs in comparison to highly specific GPR43 agonists on human intestinal epithelial and immune cells. Our results confirm that SCFAs are enhancers of barrier function in intestinal epithelial cells. Additionally, SCFAs also displayed potent immunoregulatory properties based upon the ability to inhibit LPS-induced cytokine production in PBMC, and human T cell proliferation and cytokine production. Unexpectedly, and in contrast to the current belief, specific GPR43 agonists failed to exhibit similar barrier enhancing and anti-inflammatory properties. These findings demonstrate that SCFA possess broad protective functions in IBD and agonizing GPR43 alone is unlikely to be beneficial in patients.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Receptores de Superficie Celular/agonistas , Animales , Células CACO-2 , Línea Celular , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Ácidos Grasos Volátiles , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Ratones
6.
J Immunol ; 199(2): 707-717, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28615416

RESUMEN

Atopic dermatitis (AD) is a common inflammatory skin disease affecting up to 20% of children and 3% of adults worldwide and is associated with dysregulation of the skin barrier. Although type 2 responses are implicated in AD, emerging evidence indicates a potential role for the IL-17A signaling axis in AD pathogenesis. In this study we show that in the filaggrin mutant mouse model of spontaneous AD, IL-17RA deficiency (Il17ra-/- ) resulted in severe exacerbation of skin inflammation. Interestingly, Il17ra-/- mice without the filaggrin mutation also developed spontaneous progressive skin inflammation with eosinophilia, as well as increased levels of thymic stromal lymphopoietin (TSLP) and IL-5 in the skin. Il17ra-/- mice have a defective skin barrier with altered filaggrin expression. The barrier dysregulation and spontaneous skin inflammation in Il17ra-/- mice was dependent on TSLP, but not the other alarmins IL-25 and IL-33. The associated skin inflammation was mediated by IL-5-expressing pathogenic effector Th2 cells and was independent of TCRγδ T cells and IL-22. An absence of IL-17RA in nonhematopoietic cells, but not in the hematopoietic cells, was required for the development of spontaneous skin inflammation. Skin microbiome dysbiosis developed in the absence of IL-17RA, with antibiotic intervention resulting in significant amelioration of skin inflammation and reductions in skin-infiltrating pathogenic effector Th2 cells and TSLP. This study describes a previously unappreciated protective role for IL-17RA signaling in regulation of the skin barrier and maintenance of skin immune homeostasis.


Asunto(s)
Dermatitis Atópica/inmunología , Receptores de Interleucina-17/inmunología , Receptores de Interleucina-17/metabolismo , Piel/crecimiento & desarrollo , Piel/patología , Animales , Citocinas/inmunología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Disbiosis , Eosinofilia/inmunología , Proteínas Filagrina , Regulación de la Expresión Génica , Homeostasis , Interleucina-33/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Interleucinas/genética , Interleucinas/inmunología , Proteínas de Filamentos Intermediarios/deficiencia , Proteínas de Filamentos Intermediarios/genética , Ratones , Microbiota , Mutación , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Interleucina-17/deficiencia , Receptores de Interleucina-17/genética , Transducción de Señal , Piel/inmunología , Piel/microbiología , Células Th2/inmunología , Linfopoyetina del Estroma Tímico , Interleucina-22
7.
J Vis Exp ; (122)2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28448027

RESUMEN

Obstructive respiratory diseases like asthma and chronic obstructive pulmonary disease (COPD) are currently treated by inhaled anti-inflammatory and bronchodilator drugs. Despite the availability of multiple treatments, both diseases are growing public health concerns. The majority of asthma patients are well controlled on current inhaled therapies but a substantial number of patients with severe asthma are not. Asthma affects an estimated 300 million people worldwide and approximately 20 percent have a severe form of the disease. In contrast to asthma, there are few effective therapies for COPD. An estimated 10% of the population has COPD and the trend in death rates is increasing for COPD while decreasing for other major diseases. Although developing drugs for inhaled delivery is challenging, the nose-only inhalation unit enables direct delivery of novel drugs to the lung of rodents for pre-clinical efficacy and safety/toxicology studies. Inhaled drug delivery has multiple advantages for respiratory diseases, where high concentration in the lung improves efficacy and minimizes systemic side effects. Inhaled corticosteroids and bronchodilators benefit from these advantages and inhaled delivery may also hold potential for future biologic therapies. The inhalation unit described herein can generate, sample for characterization, and uniformly deposit a drug aerosol in the lungs of rodents. This enables the pre-clinical determination of the efficacy and safety of drug doses deposited in the lungs of rodents, key data required before initiating clinical development.


Asunto(s)
Broncodilatadores/administración & dosificación , Sistemas de Liberación de Medicamentos/instrumentación , Diseño de Equipo , Nariz , Administración por Inhalación , Administración Intranasal , Aerosoles , Animales , Humanos , Ratones , Polvos
8.
PLoS Genet ; 13(3): e1006659, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28273074

RESUMEN

IL-33 is a tissue-derived cytokine that induces and amplifies eosinophilic inflammation and has emerged as a promising new drug target for asthma and allergic disease. Common variants at IL33 and IL1RL1, encoding the IL-33 receptor ST2, associate with eosinophil counts and asthma. Through whole-genome sequencing and imputation into the Icelandic population, we found a rare variant in IL33 (NM_001199640:exon7:c.487-1G>C (rs146597587-C), allele frequency = 0.65%) that disrupts a canonical splice acceptor site before the last coding exon. It is also found at low frequency in European populations. rs146597587-C associates with lower eosinophil counts (ß = -0.21 SD, P = 2.5×10-16, N = 103,104), and reduced risk of asthma in Europeans (OR = 0.47; 95%CI: 0.32, 0.70, P = 1.8×10-4, N cases = 6,465, N controls = 302,977). Heterozygotes have about 40% lower total IL33 mRNA expression than non-carriers and allele-specific analysis based on RNA sequencing and phased genotypes shows that only 20% of the total expression is from the mutated chromosome. In half of those transcripts the mutation causes retention of the last intron, predicted to result in a premature stop codon that leads to truncation of 66 amino acids. The truncated IL-33 has normal intracellular localization but neither binds IL-33R/ST2 nor activates ST2-expressing cells. Together these data demonstrate that rs146597587-C is a loss of function mutation and support the hypothesis that IL-33 haploinsufficiency protects against asthma.


Asunto(s)
Asma/genética , Eosinófilos/metabolismo , Interleucina-33/genética , Mutación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Empalme Alternativo , Animales , Sitios de Unión , Bioensayo , Niño , Preescolar , Dinamarca , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Heterocigoto , Humanos , Islandia , Lactante , Recién Nacido , Intrones , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Países Bajos , Adulto Joven
9.
Eur Respir J ; 48(3): 715-25, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27338192

RESUMEN

Severe asthma represents a major unmet clinical need. Eosinophilic inflammation persists in the airways of many patients with uncontrolled asthma, despite high-dose inhaled corticosteroid therapy. Suppressors of cytokine signalling (SOCS) are a family of molecules involved in the regulation of cytokine signalling via inhibition of the Janus kinase-signal transducers and activators of transcription pathway. We examined SOCS expression in the airways of asthma patients and investigated whether this is associated with persistent eosinophilia.Healthy controls, mild/moderate asthmatics and severe asthmatics were studied. Whole genome expression profiling, quantitative PCR and immunohistochemical analysis were used to examine expression of SOCS1, SOCS2 and SOCS3 in bronchial biopsies. Bronchial epithelial cells were utilised to examine the role of SOCS1 in regulating interleukin (IL)-13 signalling in vitroSOCS1 gene expression was significantly lower in the airways of severe asthmatics compared with mild/moderate asthmatics, and was inversely associated with airway eosinophilia and other measures of T-helper type 2 (Th2) inflammation. Immunohistochemistry demonstrated SOCS1 was predominantly localised to the bronchial epithelium. SOCS1 overexpression inhibited IL-13-mediated chemokine ligand (CCL) 26 (eotaxin-3) mRNA expression in bronchial epithelial cells.Severe asthma patients with persistent airway eosinophilia and Th2 inflammation have reduced airway epithelial SOCS1 expression. SOCS1 inhibits epithelial IL-13 signalling, supporting its key role in regulating Th2-driven eosinophilia in severe asthma.


Asunto(s)
Asma/metabolismo , Células Epiteliales/metabolismo , Interleucina-13/metabolismo , Eosinofilia Pulmonar/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Adulto , Asma/tratamiento farmacológico , Biopsia , Bronquios/metabolismo , Broncoscopía , Estudios de Casos y Controles , Línea Celular , Quimiocina CCL26/metabolismo , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica , Humanos , Inflamación , Masculino , Persona de Mediana Edad , Eosinofilia Pulmonar/tratamiento farmacológico , Mucosa Respiratoria/metabolismo , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Células Th2/citología , Adulto Joven
10.
Sci Transl Med ; 7(303): 303ra140, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26333936

RESUMEN

Sepsis is the most frequent cause of death in hospitalized patients, and severe sepsis is a leading contributory factor to acute respiratory distress syndrome (ARDS). At present, there is no effective treatment for these conditions, and care is primarily supportive. Murine sialic acid-binding immunoglobulin-like lectin-E (Siglec-E) and its human orthologs Siglec-7 and Siglec-9 are immunomodulatory receptors found predominantly on hematopoietic cells. These receptors are important negative regulators of acute inflammatory responses and are potential targets for the treatment of sepsis and ARDS. We describe a Siglec-targeting platform consisting of poly(lactic-co-glycolic acid) nanoparticles decorated with a natural Siglec ligand, di(α2→8) N-acetylneuraminic acid (α2,8 NANA-NP). This nanoparticle induced enhanced oligomerization of the murine Siglec-E receptor on the surface of macrophages, unlike the free α2,8 NANA ligand. Furthermore, treatment of murine macrophages with these nanoparticles blocked the production of lipopolysaccharide-induced inflammatory cytokines in a Siglec-E-dependent manner. The nanoparticles were also therapeutically beneficial in vivo in both systemic and pulmonary murine models replicating inflammatory features of sepsis and ARDS. Moreover, we confirmed the anti-inflammatory effect of these nanoparticles on human monocytes and macrophages in vitro and in a human ex vivo lung perfusion (EVLP) model of lung injury. We also established that interleukin-10 (IL-10) induced Siglec-E expression and α2,8 NANA-NP further augmented the expression of IL-10. Indeed, the effectiveness of the nanoparticle depended on IL-10. Collectively, these results demonstrated a therapeutic effect of targeting Siglec receptors with a nanoparticle-based platform under inflammatory conditions.


Asunto(s)
Inflamación/prevención & control , Ácido N-Acetilneuramínico/química , Nanopartículas , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/efectos de los fármacos , Animales , Humanos , Interleucina-10/fisiología , Ratones , Ratones Endogámicos C57BL , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Regulación hacia Arriba
11.
Oncotarget ; 5(16): 6964-75, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-25026282

RESUMEN

Previously we have shown that expression of the deubiquitinating enzyme USP17 is required for cell proliferation and motility. More recently we reported that USP17 deubiquitinates RCE1 isoform 2 and thus regulates the processing of 'CaaX' motif proteins. Here we now show that USP17 expression is induced by epidermal growth factor and that USP17 expression is required for clathrin mediated endocytosis of epidermal growth factor receptor. In addition, we show that USP17 is required for the endocytosis of transferrin, an archetypal substrate for clathrin mediated endocytosis, and that USP17 depletion impedes plasma membrane recruitment of the machinery required for clathrin mediated endocytosis. Thus, our data reveal that USP17 is necessary for epidermal growth factor receptor and transferrin endocytosis via clathrin coated pits, indicate this is mediated via the regulation of the recruitment of the components of the endocytosis machinery and suggest USP17 may play a general role in receptor endocytosis.


Asunto(s)
Clatrina/metabolismo , Endopeptidasas/metabolismo , Receptores ErbB/metabolismo , Ubiquitina/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Endocitosis , Endopeptidasas/genética , Células HeLa , Humanos , Transfección
12.
J Leukoc Biol ; 96(2): 255-63, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24659790

RESUMEN

TNF-α is a proinflammatory cytokine, dramatically elevated during pathogenic infection and often responsible for inflammation-induced disease pathology. SOCS proteins are inhibitors of cytokine signaling and regulators of inflammation. In this study, we found that both SOCS1 and SOCS3 were transiently induced by TNF-α and negatively regulate its NF-κB-mediated signal transduction. We discovered that PBMCs from HCV-infected patients have elevated endogenous SOCS3 expression but less TNF-α-mediated IκB degradation and proinflammatory cytokine production than healthy controls. HCV protein expression in Huh7 hepatocytes also induced SOCS3 and directly inhibited TNF-α-mediated IL-8 production. Furthermore, we found that SOCS3 associates with TRAF2 and inhibits TRAF2-mediated NF-κB promoter activity, suggesting a mechanism by which SOCS3 inhibits TNF-α-mediated signaling. These results demonstrate a role for SOCS3 in regulating proinflammatory TNF-α signal transduction and reveal a novel immune-modulatory mechanism by which HCV suppresses inflammatory responses in primary immune cells and hepatocytes, perhaps explaining mild pathology often associated with acute HCV infection.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatocitos/inmunología , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Línea Celular , Femenino , Hepatitis C/patología , Hepatocitos/patología , Hepatocitos/virología , Humanos , Inflamación/inmunología , Inflamación/patología , Interleucina-8/inmunología , Masculino , FN-kappa B/inmunología , Transducción de Señal/inmunología , Proteína 3 Supresora de la Señalización de Citocinas , Factor 2 Asociado a Receptor de TNF/inmunología
13.
PLoS One ; 9(1): e85834, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465735

RESUMEN

Bacterial Lipopolysaccharide (LPS) is a strong inducer of inflammation and does so by inducing polarization of macrophages to the classic inflammatory M1 population. Given the role of Btk as a critical signal transducer downstream of TLR4, we investigated its role in M1/M2 induction. In Btk deficient (Btk (-\-)) mice we observed markedly reduced recruitment of M1 macrophages following intraperitoneal administration of LPS. Ex vivo analysis demonstrated an impaired ability of Btk(-/-) macrophages to polarize into M1 macrophages, instead showing enhanced induction of immunosuppressive M2-associated markers in response to M1 polarizing stimuli, a finding accompanied by reduced phosphorylation of STAT1 and enhanced STAT6 phosphorylation. In addition to STAT activation, M1 and M2 polarizing signals modulate the expression of inflammatory genes via differential activation of transcription factors and regulatory proteins, including NF-κB and SHIP1. In keeping with a critical role for Btk in macrophage polarization, we observed reduced levels of NF-κB p65 and Akt phosphorylation, as well as reduced induction of the M1 associated marker iNOS in Btk(-/-) macrophages in response to M1 polarizing stimuli. Additionally enhanced expression of SHIP1, a key negative regulator of macrophage polarisation, was observed in Btk(-/-) macrophages in response to M2 polarizing stimuli. Employing classic models of allergic M2 inflammation, treatment of Btk (-/-) mice with either Schistosoma mansoni eggs or chitin resulted in increased recruitment of M2 macrophages and induction of M2-associated genes. This demonstrates an enhanced M2 skew in the absence of Btk, thus promoting the development of allergic inflammation.


Asunto(s)
Polaridad Celular/efectos de los fármacos , Macrófagos/citología , Macrófagos/enzimología , Proteínas Tirosina Quinasas/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Hipersensibilidad/complicaciones , Hipersensibilidad/enzimología , Hipersensibilidad/patología , Inflamación/complicaciones , Inflamación/enzimología , Inflamación/patología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Fenotipo , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/deficiencia , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Transcripción Genética/efectos de los fármacos
14.
Biochem J ; 457(2): 289-300, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24134311

RESUMEN

Processing of the 'CaaX' motif found on the C-termini of many proteins, including the proto-oncogene Ras, requires the ER (endoplasmic reticulum)-resident protease RCE1 (Ras-converting enzyme 1) and is necessary for the proper localization and function of many of these 'CaaX' proteins. In the present paper, we report that several mammalian species have a novel isoform (isoform 2) of RCE1 resulting from an alternate splice site and producing an N-terminally truncated protein. We demonstrate that both RCE1 isoform 1 and the newly identified isoform 2 are required to reinstate proper H-Ras processing and thus plasma membrane localization in RCE1-null cells. In addition, we show that the deubiquitinating enzyme USP17 (ubiquitin-specific protease 17), previously shown to modulate RCE1 activity, can regulate the abundance and localization of isoform 2. Furthermore, we show that isoform 2 is ubiquitinated on Lys43 and deubiquitinated by USP17. Collectively, the findings of the present study indicate that RCE1 isoform 2 is required for proper 'CaaX' processing and that USP17 can regulate this via its modulation of RCE1 isoform 2 ubiquitination.


Asunto(s)
Membrana Celular/metabolismo , Endopeptidasas/metabolismo , Endopeptidasas/fisiología , Genes ras/fisiología , Membrana Celular/química , Células HEK293 , Células HeLa , Humanos , Isoformas de Proteínas/metabolismo , Proto-Oncogenes Mas
15.
Oncotarget ; 4(10): 1836-43, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24123619

RESUMEN

USP17 is a cell cycle regulated deubiquitinating enzyme that is highly expressed in tumor-derived cell lines and has an established role in cell proliferation and chemotaxis. This is the first study to examine the clinical significance of USP17 expression in non-small cell lung cancer (NSCLC). USP17 was overexpressed in both squamous and adenocarcinoma NSCLC tissue. Patients with USP17 positive tumors had significantly reduced recurrence-free survival than patients with USP17 negative tumors. Moreover, USP17 was more highly expressed in patients with recurrence of disease at distant sites, suggesting that USP17 levels may correlate with NSCLC distant metastases. Overall, these findings establish USP17 as a potentially valuable novel biomarker for metastatic lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Endopeptidasas/metabolismo , Neoplasias Pulmonares/enzimología , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Estudios de Cohortes , Supervivencia sin Enfermedad , Endopeptidasas/genética , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Análisis de Supervivencia , Ubiquitinación
16.
Int J Cancer ; 133(9): 2102-12, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23629809

RESUMEN

Recent murine studies have demonstrated that tumor-associated macrophages in the tumor microenvironment are a key source of the pro-tumorigenic cysteine protease, cathepsin S. We now show in a syngeneic colorectal carcinoma murine model that both tumor and tumor-associated cells contribute cathepsin S to promote neovascularization and tumor growth. Cathepsin S depleted and control colorectal MC38 tumor cell lines were propagated in both wild type C57Bl/6 and cathepsin S null mice to provide stratified depletion of the protease from either the tumor, tumor-associated host cells, or both. Parallel analysis of these conditions showed that deletion of cathepsin S inhibited tumor growth and development, and revealed a clear contribution of both tumor and tumor-associated cell derived cathepsin S. The most significant impact on tumor development was obtained when the protease was depleted from both sources. Further characterization revealed that the loss of cathepsin S led to impaired tumor vascularization, which was complemented by a reduction in proliferation and increased apoptosis, consistent with reduced tumor growth. Analysis of cell types showed that in addition to the tumor cells, tumor-associated macrophages and endothelial cells can produce cathepsin S within the microenvironment. Taken together, these findings clearly highlight a manner by which tumor-associated cells can positively contribute to developing tumors and highlight cathepsin S as a therapeutic target in cancer.


Asunto(s)
Carcinoma Pulmonar de Lewis/patología , Catepsinas/fisiología , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/patología , Melanoma Experimental/patología , Neovascularización Patológica , Animales , Apoptosis , Western Blotting , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/genética , Adhesión Celular , Ciclo Celular , Células Cultivadas , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/genética , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Macrófagos/citología , Macrófagos/metabolismo , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microambiente Tumoral
17.
FEBS Lett ; 587(10): 1571-8, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23587486

RESUMEN

JAK/STAT signalling is essential for anti-viral immunity, making IFN-α an obvious anti-viral therapeutic. However, many HCV+ patients fail treatment, indicating that the virus blocks successful IFN-α signalling. We found that STAT1 and STAT3 proteins, key components of the IFN-α signalling pathway were reduced in immune cells and hepatocytes from HCV infected patients, and upon HCV expression in Huh7 hepatocytes. However, STAT1 and STAT3 mRNA levels were normal. Mechanistic analysis revealed that in the presence of HCV, STAT3 protein was preferentially ubiquitinated, and degradation was blocked by the proteasomal inhibitor MG132. These findings show that HCV inhibits IFN-α responses in a broad spectrum of cells via proteasomal degradation of JAK/STAT pathway components.


Asunto(s)
Hepacivirus/fisiología , Hepatocitos/metabolismo , Interferón-alfa/metabolismo , Quinasas Janus/metabolismo , Linfocitos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Factores de Transcripción STAT/metabolismo , Adulto , Anciano , Células Cultivadas , Regulación hacia Abajo/inmunología , Femenino , Hepatocitos/virología , Humanos , Inmunidad Celular/fisiología , Linfocitos/virología , Masculino , Persona de Mediana Edad , Transducción de Señal/fisiología
18.
J Immunol ; 190(7): 3235-45, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23455506

RESUMEN

Suppressor of cytokine signaling (SOCS) proteins are key regulators of CD4(+) T cell differentiation, and in particular, we have recently shown that SOCS2 inhibits the development of Th2 cells and allergic immune responses. Interestingly, transcriptome analyses have identified SOCS2 as being preferentially expressed in both natural regulatory T cells (Tregs) and inducible Tregs (iTregs); however, the role of SOCS2 in Foxp3(+) Treg function or development has not been fully elucidated. In this study, we show that despite having no effect on natural Treg development or function, SOCS2 is highly expressed in iTregs and required for the stable expression of Foxp3 in iTregs in vitro and in vivo. Indeed, SOCS2-deficient CD4(+) T cells upregulated Foxp3 following in vitro TGF-ß stimulation, but failed to maintain stable expression of Foxp3. Moreover, in vivo generation of iTregs following OVA feeding was impaired in the absence of SOCS2 and could be rescued in the presence of IL-4 neutralizing Ab. Following IL-4 stimulation, SOCS2-deficient Foxp3(+) iTregs secreted elevated IFN-γ and IL-13 levels and displayed enhanced STAT6 phosphorylation. Therefore, we propose that SOCS2 regulates iTreg stability by downregulating IL-4 signaling. Moreover, SOCS2 is essential to maintain the anti-inflammatory phenotype of iTregs by preventing the secretion of proinflammatory cytokines. Collectively, these results suggest that SOCS2 may prevent IL-4-induced Foxp3(+) iTreg instability. Foxp3(+) iTregs are key regulators of immune responses at mucosal surfaces; therefore, this dual role of SOCS2 in both Th2 and Foxp3(+) iTregs reinforces SOCS2 as a potential therapeutic target for Th2-biased diseases.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Interleucina-4/farmacología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Factor de Transcripción STAT6/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Proteínas Supresoras de la Señalización de Citocinas/genética , Linfocitos T Reguladores/efectos de los fármacos
19.
Immunity ; 38(1): 66-78, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23177319

RESUMEN

Suppressors of cytokine signaling (SOCS) are important regulators of lipopolysaccharide (LPS) and cytokine responses but their role in macrophage polarization is unknown. We have shown here that myeloid-restricted Socs3 deletion (Socs3(Lyz2cre)) resulted in resistance to LPS-induced endotoxic shock, whereas Socs2(-/-) mice were highly susceptible. We observed striking bias toward M2-like macrophages in Socs3(Lyz2cre) mice, whereas the M1-like population was enriched in Socs2(-/-) mice. Adoptive transfer experiments showed that responses to endotoxic shock and polymicrobial sepsis were transferable and macrophage dependent. Critically, this dichotomous response was associated with enhanced regulatory T (Treg) cell recruitment by Socs3(Lyz2cre) cells, whereas Treg cell recruitment was absent in the presence of Socs2(-/-) macrophages. In addition, altered polarization coincided with enhanced interferon-gamma (IFN-γ)-induced signal transducer and activator of transcription-1 (STAT1) activation in Socs2(-/-) macrophages and enhanced interleukin-4 (IL-4) plus IL-13-induced STAT6 phosphorylation in Socs3(Lyz2cre) macrophages. SOCS, therefore, are essential controllers of macrophage polarization, regulating inflammatory responses.


Asunto(s)
Polaridad Celular/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Traslado Adoptivo , Animales , Regulación de la Expresión Génica , Interleucina-10/inmunología , Interleucina-10/metabolismo , Macrófagos/trasplante , Ratones , Factores de Transcripción STAT/metabolismo , Sepsis/genética , Sepsis/inmunología , Sepsis/prevención & control , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trasplante Isogénico
20.
J Immunol ; 189(5): 2079-83, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22837488

RESUMEN

GM-CSF is a potent proinflammatory cytokine that plays a pathogenic role in the CNS inflammatory disease experimental autoimmune encephalomyelitis. As IL-27 alleviates experimental autoimmune encephalomyelitis, we hypothesized that IL-27 suppresses GM-CSF expression by T cells. We found that IL-27 suppressed GM-CSF expression in CD4+ and CD8+ T cells in splenocyte and purified T cell cultures. IL-27 suppressed GM-CSF in Th1, but not Th17, cells. IL-27 also suppressed GM-CSF expression by human T cells in nonpolarized and Th1- but not Th17-polarized PBMC cultures. In vivo, IL-27p28 deficiency resulted in increased GM-CSF expression by CNS-infiltrating T cells during Toxoplasma gondii infection. Although in vitro suppression of GM-CSF by IL-27 was independent of IL-2 suppression, IL-10 upregulation, or SOCS3 signaling, we observed that IL-27-driven suppression of GM-CSF was STAT1 dependent. Our findings demonstrate that IL-27 is a robust negative regulator of GM-CSF expression in T cells, which likely inhibits T cell pathogenicity in CNS inflammation.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Tolerancia Inmunológica , Interleucina-17/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Polaridad Celular/genética , Polaridad Celular/inmunología , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Tolerancia Inmunológica/genética , Mediadores de Inflamación/farmacología , Mediadores de Inflamación/fisiología , Interleucina-17/farmacología , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Toxoplasmosis/inmunología , Toxoplasmosis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...