Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Transl Oncol ; 14(1): 100883, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33074129

RESUMEN

Rovalpituzumab tesirine (Rova-T) offers a targeted therapy for ~85% of SCLC patients whose tumors express DLL3, but clinical dosing is limited due to off-target toxicities. We hypothesized that a sub-efficacious dose of Rova-T combined with anti-PD1, which alone shows a clinical benefit to ~15% of SCLC patients, might elicit a novel mechanism of action and extend clinical utility. Using a pre-clinical murine SCLC tumor model that expresses Dll3 and has an intact murine immune system, we found that sub-efficacious doses of Rova-T with anti-PD1 resulted in enhanced anti-tumor activity, compared to either monotherapy. Multiplex immunohistochemistry (IHC) showed CD4 and CD8 T-cells primarily in normal tissue immediately adjacent to the tumor. Combination treatment, but not anti-PD1 alone, increased Ki67+/CD8 T-cells and Granzyme B+/CD8 in tumors by flow cytometry and IHC. Antibody depletion of T-cell populations showed CD8+ T-cells are required for in vivo anti-tumor efficacy. Whole transcriptome analysis as well as flow cytometry and IHC showed that Rova-T activates dendritic cells and increases Ccl5, Il-12, and Icam more than anti-PD1 alone. Increased tumor expression of PDL1 and MHC1 following Rova-T treatment also supports combination with anti-PD1. Mice previously treated with Rova-T + anti-PD1 withstood tumor re-challenge, demonstrating sustained anti-tumor immunity. Collectively our pre-clinical data support clinical combination of sub-efficacious Rova-T with anti-PD1 to extend the benefit of immune checkpoint inhibitors to more SCLC patients.

2.
Clin Exp Allergy ; 50(12): 1342-1351, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32909660

RESUMEN

BACKGROUND: The anti-interleukin 13 (IL-13) monoclonal antibody lebrikizumab improves lung function in patients with moderate-to-severe uncontrolled asthma, but its effects on airway inflammation and remodelling are unknown. CLAVIER was designed to assess lebrikizumab's effect on eosinophilic inflammation and remodelling. OBJECTIVE: To report safety and efficacy results from enrolled participants with available data from CLAVIER. METHODS: We performed bronchoscopy on patients with uncontrolled asthma before and after 12 weeks of randomized double-blinded treatment with lebrikizumab (n = 31) or placebo (n = 33). The pre-specified primary end-point was relative change in airway subepithelial eosinophils per mm2 of basement membrane (cells/mm2 ). Pre-specified secondary and exploratory outcomes included change in IL-13-associated biomarkers and measures of airway remodelling. RESULTS: There was a baseline imbalance in tissue eosinophils and high variability between treatment groups. There was no discernible change in adjusted mean subepithelial eosinophils/mm2 in response to lebrikizumab (95% CI, -82.5%, 97.5%). As previously observed, FEV1 increased after lebrikizumab treatment. Moreover, subepithelial collagen thickness decreased 21.5% after lebrikizumab treatment (95% CI, -32.9%, -10.2%), and fractional exhaled nitric oxide, CCL26 and SERPINB2 mRNA expression in bronchial tissues also reduced. Lebrikizumab was well tolerated, with a safety profile consistent with other lebrikizumab asthma studies. CONCLUSIONS & CLINICAL RELEVANCE: We did not observe reduced tissue eosinophil numbers in association with lebrikizumab treatment. However, in pre-specified exploratory analyses, lebrikizumab treatment was associated with reduced degree of subepithelial fibrosis, a feature of airway remodelling, as well as improved lung function and reduced key pharmacodynamic biomarkers in bronchial tissues. These results reinforce the importance of IL-13 in airway pathobiology and suggest that neutralization of IL-13 may reduce asthmatic airway remodelling. CLINICAL TRIAL REGISTRATION: NCT02099656.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Antiasmáticos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Asma/tratamiento farmacológico , Eosinófilos/efectos de los fármacos , Interleucina-13/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Adolescente , Adulto , Anciano , Antiasmáticos/efectos adversos , Antiasmáticos/farmacocinética , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Asma/inmunología , Asma/fisiopatología , Método Doble Ciego , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Humanos , Pulmón/inmunología , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Transducción de Señal , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
3.
Proc Natl Acad Sci U S A ; 115(44): 11244-11249, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30322923

RESUMEN

The E3 ubiquitin ligase CRL4COP1/DET1 is active in the absence of ERK signaling, modifying the transcription factors ETV1, ETV4, ETV5, and c-JUN with polyubiquitin that targets them for proteasomal degradation. Here we show that this posttranslational regulatory mechanism is active in neurons, with ETV5 and c-JUN accumulating within minutes of ERK activation. Mice with constitutive photomorphogenesis 1 (Cop1) deleted in neural stem cells showed abnormally elevated expression of ETV1, ETV4, ETV5, and c-JUN in the developing brain and spinal cord. Expression of c-JUN target genes Vimentin and Gfap was increased, whereas ETV5 and c-JUN both contributed to an expanded number of cells expressing genes associated with gliogenesis, including Olig1, Olig2, and Sox10. The mice had subtle morphological abnormalities in the cerebral cortex, hippocampus, and cerebellum by embryonic day 18 and died soon after birth. Elevated c-JUN, ETV5, and ETV1 contributed to the perinatal lethality, as several Cop1-deficient mice also lacking c-Jun and Etv5, or lacking Etv5 and heterozygous for Etv1, were viable.


Asunto(s)
Encéfalo/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al ADN/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factores de Transcripción/metabolismo
5.
J Histochem Cytochem ; 63(4): 244-55, 2015 04.
Artículo en Inglés | MEDLINE | ID: mdl-25575566

RESUMEN

With recent advances in immunohistochemical (IHC) techniques, immunohistochemistry now plays a more important role in research, especially in mouse models where characterization of cellular patterns of protein expression has become critical. Even with these recent advances, a paucity of IHC quality antibodies for some proteins still exists. To address this, we have developed a novel IHC assay that utilizes a commercially available goat anti-DDDDK peptide polyclonal antibody on paraffin-embedded tissues from knock-in mice expressing proteins of interest tagged with a 3 × FLAG epitope at physiologically relevant levels. Focusing on two 3 × FLAG-tagged proteins for which specific antibodies were available, USP48 and RIPK3, we were able to validate our anti-DDDDK assay by comparing the IHC directed against the actual proteins to the anti-DDDDK IHC assay, which recognizes the FLAG epitope. We were also able to detect a third 3 × FLAG-tagged protein, BAP1, for which quality reagents were not available. This universal IHC method will enable researchers to characterize the expression patterns of proteins of interest when specific antibodies are lacking.


Asunto(s)
Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Anticuerpos , Epítopos , Técnicas de Sustitución del Gen , Cabras , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Mutantes , Oligopéptidos/inmunología , Oligopéptidos/metabolismo , Especificidad de Órganos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Proteasas Ubiquitina-Específicas/genética
6.
Science ; 343(6177): 1357-60, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24557836

RESUMEN

Receptor-interacting protein kinase 1 (RIPK1) and RIPK3 trigger pro-inflammatory cell death termed "necroptosis." Studies with RIPK3-deficient mice or the RIPK1 inhibitor necrostatin-1 suggest that necroptosis exacerbates pathology in many disease models. We engineered mice expressing catalytically inactive RIPK3 D161N or RIPK1 D138N to determine the need for the active kinase in the whole animal. Unexpectedly, RIPK3 D161N promoted lethal RIPK1- and caspase-8-dependent apoptosis. In contrast, mice expressing RIPK1 D138N were viable and, like RIPK3-deficient mice, resistant to tumor necrosis factor (TNF)-induced hypothermia. Cells expressing RIPK1 D138N were resistant to TNF-induced necroptosis, whereas TNF-induced signaling pathways promoting gene transcription were unperturbed. Our data indicate that the kinase activity of RIPK3 is essential for necroptosis but also governs whether a cell activates caspase-8 and dies by apoptosis.


Asunto(s)
Apoptosis , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Caspasa 8/genética , Caspasa 8/metabolismo , Supervivencia Celular , Pérdida del Embrión , Desarrollo Embrionario , Enteritis/patología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Técnicas de Sustitución del Gen , Intestino Grueso/patología , Intestino Delgado/patología , Ratones , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factor de Necrosis Tumoral alfa/farmacología
7.
PLoS One ; 7(5): e36713, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615798

RESUMEN

The fibroblast growth factor (FGF)-FGF receptor (FGFR) signaling system plays critical roles in a variety of normal developmental and physiological processes. It is also well documented that dysregulation of FGF-FGFR signaling may have important roles in tumor development and progression. The FGFR4-FGF19 signaling axis has been implicated in the development of hepatocellular carcinomas (HCCs) in mice, and potentially in humans. In this study, we demonstrate that FGFR4 is required for hepatocarcinogenesis; the progeny of FGF19 transgenic mice, which have previously been shown to develop HCCs, bred with FGFR4 knockout mice fail to develop liver tumors. To further test the importance of FGFR4 in HCC, we developed a blocking anti-FGFR4 monoclonal antibody (LD1). LD1 inhibited: 1) FGF1 and FGF19 binding to FGFR4, 2) FGFR4-mediated signaling, colony formation, and proliferation in vitro, and 3) tumor growth in a preclinical model of liver cancer in vivo. Finally, we show that FGFR4 expression is elevated in several types of cancer, including liver cancer, as compared to normal tissues. These findings suggest a modulatory role for FGFR4 in the development and progression of hepatocellular carcinoma and that FGFR4 may be an important and novel therapeutic target in treating this disease.


Asunto(s)
Carcinoma Hepatocelular/prevención & control , Modelos Animales de Enfermedad , Neoplasias Hepáticas/prevención & control , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Animales , Anticuerpos Neutralizantes/inmunología , Carcinoma Hepatocelular/patología , División Celular , Neoplasias Hepáticas/patología , Ratones , Ratones Transgénicos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/inmunología
8.
Methods Mol Biol ; 832: 185-96, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22350886

RESUMEN

Antibodies that specifically recognize polyubiquitin chains containing ubiquitins linked at a particular lysine residue are powerful tools for interrogating endogenous protein modifications. Here, we describe protocols for revealing K11-, K48-, and K63-linked polyubiquitin chains by western blotting, immunoprecipitation, or immunostaining.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Poliubiquitina/química , Poliubiquitina/inmunología , Western Blotting/métodos , Inmunoprecipitación/métodos , Poliubiquitina/metabolismo , Procesamiento Proteico-Postraduccional , Enzimas Activadoras de Ubiquitina , Enzimas Ubiquitina-Conjugadoras , Ubiquitina-Proteína Ligasas , Ubiquitinación
9.
Nature ; 474(7351): 403-6, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21572435

RESUMEN

The proto-oncogenes ETV1, ETV4 and ETV5 encode transcription factors in the E26 transformation-specific (ETS) family, which includes the most frequently rearranged and overexpressed genes in prostate cancer. Despite being critical regulators of development, little is known about their post-translational regulation. Here we identify the ubiquitin ligase COP1 (also known as RFWD2) as a tumour suppressor that negatively regulates ETV1, ETV4 and ETV5. ETV1, which is mutated in prostate cancer more often, was degraded after being ubiquitinated by COP1. Truncated ETV1 encoded by prostate cancer translocation TMPRSS2:ETV1 lacks the critical COP1 binding motifs and was 50-fold more stable than wild-type ETV1. Almost all patient translocations render ETV1 insensitive to COP1, implying that this confers a selective advantage to prostate epithelial cells. Indeed, COP1 deficiency in mouse prostate elevated ETV1 and produced increased cell proliferation, hyperplasia, and early prostate intraepithelial neoplasia. Combined loss of COP1 and PTEN enhanced the invasiveness of mouse prostate adenocarcinomas. Finally, rare human prostate cancer samples showed hemizygous loss of the COP1 gene, loss of COP1 protein, and elevated ETV1 protein while lacking a translocation event. These findings identify COP1 as a tumour suppressor whose downregulation promotes prostatic epithelial cell proliferation and tumorigenesis.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Masculino , Ratones , Proteínas Nucleares/deficiencia , Fosfohidrolasa PTEN/deficiencia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
10.
Clin Cancer Res ; 17(5): 965-75, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245091

RESUMEN

PURPOSE: To identify and evaluate targets amenable to antibody therapy in melanoma. EXPERIMENTAL DESIGN: We searched for mRNA transcripts coding for cell-surface proteins with expression patterns similar to that of the melanoma oncogene MITF. One such candidate, the endothelin B receptor (EDNBR), was first analyzed for a functional contribution to tumor growth by conditional induction of shRNA. Second, antibodies were raised to the receptor, conjugated with monomethyl auristatin E, and tested for efficacy against melanoma tumor models generated from cell lines. RESULTS: Conditional knockdown of the receptor in tumor xenograft models resulted in only a modest impact on tumor growth. A monoclonal antibody reactive with the N-terminal tail of EDNBR was found to internalize rapidly into melanoma cells. When conjugated with monomethyl auristatin E, the antibody-drug conjugate (ADC) showed remarkable efficacy against human melanoma cell lines and xenograft tumor models that was commensurate with levels of receptor expression. Comparative immunohistochemistry revealed a range of EDNBR expression across a panel of human melanomas, with the majority expressing levels equivalent to or greater than that in the models responsive to the ADC. CONCLUSION: An ADC targeting the EDNBR is highly efficacious in preclinical models of melanoma.


Asunto(s)
Antineoplásicos/uso terapéutico , Antagonistas de los Receptores de la Endotelina B , Inmunoconjugados/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Receptor de Endotelina B/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Western Blotting , Línea Celular Tumoral , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Macaca fascicularis , Melanoma/genética , Melanoma/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción Asociado a Microftalmía/genética , Oligopéptidos/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Nat Chem Biol ; 7(1): 41-50, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21113169

RESUMEN

Bruton's tyrosine kinase (Btk) is a therapeutic target for rheumatoid arthritis, but the cellular and molecular mechanisms by which Btk mediates inflammation are poorly understood. Here we describe the discovery of CGI1746, a small-molecule Btk inhibitor chemotype with a new binding mode that stabilizes an inactive nonphosphorylated enzyme conformation. CGI1746 has exquisite selectivity for Btk and inhibits both auto- and transphosphorylation steps necessary for enzyme activation. Using CGI1746, we demonstrate that Btk regulates inflammatory arthritis by two distinct mechanisms. CGI1746 blocks B cell receptor-dependent B cell proliferation and in prophylactic regimens reduces autoantibody levels in collagen-induced arthritis. In macrophages, Btk inhibition abolishes FcγRIII-induced TNFα, IL-1ß and IL-6 production. Accordingly, in myeloid- and FcγR-dependent autoantibody-induced arthritis, CGI1746 decreases cytokine levels within joints and ameliorates disease. These results provide new understanding of the function of Btk in both B cell- or myeloid cell-driven disease processes and provide a compelling rationale for targeting Btk in rheumatoid arthritis.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Linfocitos B/efectos de los fármacos , Benzamidas/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Células Mieloides/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Agammaglobulinemia Tirosina Quinasa , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Benzamidas/química , Benzamidas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Ratones , Células Mieloides/inmunología , Células Mieloides/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/farmacología , Proteínas Tirosina Quinasas/uso terapéutico , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
12.
Nature ; 463(7277): 103-7, 2010 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20023629

RESUMEN

MCL1 is essential for the survival of stem and progenitor cells of multiple lineages, and is unique among pro-survival BCL2 family members in that it is rapidly turned over through the action of ubiquitin ligases. B- and mantle-cell lymphomas, chronic myeloid leukaemia, and multiple myeloma, however, express abnormally high levels of MCL1, contributing to chemoresistance and disease relapse. The mechanism of MCL1 overexpression in cancer is not well understood. Here we show that the deubiquitinase USP9X stabilizes MCL1 and thereby promotes cell survival. USP9X binds MCL1 and removes the Lys 48-linked polyubiquitin chains that normally mark MCL1 for proteasomal degradation. Increased USP9X expression correlates with increased MCL1 protein in human follicular lymphomas and diffuse large B-cell lymphomas. Moreover, patients with multiple myeloma overexpressing USP9X have a poor prognosis. Knockdown of USP9X increases MCL1 polyubiquitination, which enhances MCL1 turnover and cell killing by the BH3 mimetic ABT-737. These results identify USP9X as a prognostic and therapeutic target, and they show that deubiquitinases may stabilize labile oncoproteins in human malignancies.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Poliubiquitina/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN , Docetaxel , Etopósido/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Semivida , Humanos , Lisina/metabolismo , Ratones , Ratones SCID , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias/diagnóstico , Nitrofenoles/farmacología , Fosforilación/efectos de la radiación , Piperazinas/farmacología , Pronóstico , Unión Proteica/efectos de la radiación , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Interferencia de ARN , Sulfonamidas/farmacología , Taxoides/farmacología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/genética , Ubiquitinación , Rayos Ultravioleta , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Biol Chem ; 284(1): 218-228, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18948266

RESUMEN

The trypsin-like serine protease marapsin is a member of the large protease gene cluster at human chromosome 16p13.3, which also contains the structurally related proteases testisin, tryptase epsilon, tryptase gamma, and EOS. To gain insight into the biological functions of marapsin, we undertook a detailed gene expression analysis. It showed that marapsin expression was restricted to tissues containing stratified squamous epithelia and was absent or only weakly expressed in all other tissues, including the pancreas. Marapsin was constitutively expressed in nonkeratinizing stratified squamous epithelia of human esophagus, tonsil, cervix, larynx, and cornea. In the keratinizing stratified squamous epidermis of skin, however, its expression was induced only during epidermal hyperproliferation, such as in psoriasis and in murine wound healing. In fact, marapsin was the second most strongly up-regulated protease in psoriatic lesions, where expression was localized to the upper region of the hyperplastic epidermis. Similarly, in the hyperproliferative epithelium of regenerating murine skin wounds, marapsin localized to the suprabasal layers, where keratinocytes undergo squamous differentiation. The transient up-regulation of marapsin, which closely correlated with re-epithelialization, was virtually absent in a genetic mouse model of delayed wound closure. These results suggested a function during the process of re-epithelialization. Furthermore, in reconstituted human epidermis, a model system of epidermal differentiation, members of the IL-20 subfamily of cytokines, such as IL-22, induced marapsin expression. Consistent with a physiologic role in marapsin regulation, IL-22 was also strongly expressed in re-epithelializing skin wounds. Marapsin's restricted expression, localization, and cytokine-inducible expression suggest a role in the terminal differentiation of keratinocytes in hyperproliferating squamous epithelia.


Asunto(s)
Proliferación Celular , Epidermis/enzimología , Regulación Enzimológica de la Expresión Génica , Psoriasis/enzimología , Serina Endopeptidasas/biosíntesis , Cicatrización de Heridas , Animales , Células CHO , Cricetinae , Cricetulus , Epidermis/patología , Humanos , Interleucinas/metabolismo , Queratinocitos/enzimología , Queratinocitos/patología , Ratones , Especificidad de Órganos , Psoriasis/patología , Regulación hacia Arriba , Heridas Penetrantes/enzimología , Heridas Penetrantes/patología , Interleucina-22
14.
Cell ; 134(4): 668-78, 2008 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-18724939

RESUMEN

Posttranslational modification of proteins with polyubiquitin occurs in diverse signaling pathways and is tightly regulated to ensure cellular homeostasis. Studies employing ubiquitin mutants suggest that the fate of polyubiquitinated proteins is determined by which lysine within ubiquitin is linked to the C terminus of an adjacent ubiquitin. We have developed linkage-specific antibodies that recognize polyubiquitin chains joined through lysine 63 (K63) or 48 (K48). A cocrystal structure of an anti-K63 linkage Fab bound to K63-linked diubiquitin provides insight into the molecular basis for specificity. We use these antibodies to demonstrate that RIP1, which is essential for tumor necrosis factor-induced NF-kappaB activation, and IRAK1, which participates in signaling by interleukin-1beta and Toll-like receptors, both undergo polyubiquitin editing in stimulated cells. Both kinase adaptors initially acquire K63-linked polyubiquitin, while at later times K48-linked polyubiquitin targets them for proteasomal degradation. Polyubiquitin editing may therefore be a general mechanism for attenuating innate immune signaling.


Asunto(s)
Anticuerpos/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Ubiquitina/metabolismo , Animales , Línea Celular , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Proteínas de Complejo Poro Nuclear/química , Biblioteca de Péptidos , Proteínas de Unión al ARN/química , Saccharomyces cerevisiae , Schizosaccharomyces , Ubiquitina/química , Ubiquitinación
15.
Chest ; 130(4): 1102-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17035444

RESUMEN

BACKGROUND: Habitual cigarette smoking is associated with chronic mucus hypersecretion, but the relationship between mucus abnormalities and airflow obstruction in smokers is uncertain. METHODS: We collected bronchial biopsy samples and epithelial brushings from 24 smokers with and without airflow obstruction and 19 nonsmoking healthy control subjects. Epithelial mucin stores, mucin immunostains, and goblet cell morphology were quantified in bronchial biopsy samples using stereology, and mucin gene expression was quantified in epithelial brushings using real-time reverse transcriptase-polymerase chain reaction. RESULTS: Goblet cell size and number were higher than normal in smokers (both p < 0.05), leading to a 2.2-fold increase in the volume of stored mucin in the epithelium per surface area of basal lamina (1.94 +/- 0.31 microm(3)/microm(2) vs 4.32 +/- 0.55 microm(3)/microm(2) in control subjects vs smokers, p = 0.001). The increase in stored mucin occurred because of an increase in MUC5AC (p = 0.018) and despite a decrease in MUC5B (p < 0.0001). Stored mucin was significantly higher in the subgroup of smokers with airflow obstruction (p = 0.029) and correlated with FEV(1)/FVC even when controlling for diffusing capacity as a measure of emphysema (p = 0.034). CONCLUSIONS: Epithelial mucin stores are increased in habitual smokers because of goblet cell hypertrophy and hyperplasia, and the pattern of mucin gene expression is abnormal. The highest epithelial mucin stores are found in smokers with airflow obstruction, suggesting a mechanistic link between epithelial mucin dysregulation and airflow obstruction.


Asunto(s)
Mucinas/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/patología , Fumar/efectos adversos , Fumar/patología , Adulto , Biopsia , Bronquios/patología , Broncoscopía , Femenino , Volumen Espiratorio Forzado/fisiología , Expresión Génica/fisiología , Células Caliciformes/patología , Humanos , Hiperplasia/patología , Masculino , Persona de Mediana Edad , Mucina 5AC , Mucina 2 , Mucina 5B , Enfermedad Pulmonar Obstructiva Crónica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Capacidad Vital/fisiología
16.
Chest ; 125(6): 2328-35, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15189958

RESUMEN

BACKGROUND: Controlled human exposure studies have produced conflicting results regarding the effect of ozone on the early bronchoconstrictor response to inhaled allergen in specifically sensitized asthmatic subjects. Spirometric parameters do not necessarily reflect the airway inflammatory effects of inhaled ozone or allergen. OBJECTIVE: This study was designed to investigate whether exposure to ozone enhances the late airway inflammatory response, as well as the early bronchoconstrictor response, to inhaled house dust mite allergen in sensitized asthmatic subjects. DESIGN: Randomized, counter-balanced, cross-over study. SETTING: Human exposure laboratory. METHODS: Fourteen subjects were exposed to 0.2 ppm O(3) or filtered air, on separate days, for 1 h during exercise. After each exposure, the subjects were challenged with doubling doses of Dermatophagoides farinae (DF) allergen (provocative concentration of DF causing a 15% decrease in FEV(1) [PC(15)]). At 6 h after allergen challenge, bronchoscopy with BAL, proximal airway lavage (PAL), and endobronchial biopsy were performed. The second exposure/allergen challenge/bronchoscopy sequence was performed at least 4 weeks after the first sequence. RESULTS: No significant difference in cellular or biochemical markers of the late inflammatory response after allergen was found between the ozone and air exposures (although a trend toward increased neutrophils was noted after ozone exposure in the PAL fluid, p = 0.06). For the group as a whole, no significant difference in PC(15) was demonstrated after ozone exposure compared to air exposure. However, subjects with the greatest ozone-induced decrements in FEV(1) tended to have lower PC(15) values after ozone exposure. CONCLUSION: Exposure to a relatively low-level concentration of ozone does not enhance the late inflammatory or early bronchoconstrictor response to inhaled antigen in most allergic asthmatic subjects. Our results do suggest, however, that a subgroup of asthmatics may acquire increased sensitivity to aeroallergens after exposure to ozone.


Asunto(s)
Antígenos Dermatofagoides/efectos adversos , Asma/inmunología , Asma/fisiopatología , Hiperreactividad Bronquial/inmunología , Ozono/farmacología , Administración por Inhalación , Adolescente , Adulto , Asma/diagnóstico , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar/citología , Estudios Cruzados , Monitoreo del Ambiente , Humanos , Masculino , Compuestos Organotiofosforados , Probabilidad , Valores de Referencia , Pruebas de Función Respiratoria , Sensibilidad y Especificidad , Estadísticas no Paramétricas
17.
Am J Respir Crit Care Med ; 169(9): 1001-6, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-14726423

RESUMEN

Bronchial hyperresponsiveness in mild to moderate asthma may result from airway smooth muscle cell proliferation or acquisition of a hypercontractile phenotype. Because these cells have not been well characterized in mild to moderate asthma, we examined the morphometric and gene expression characteristics of smooth muscle cells in this subgroup of patients with asthma. Using bronchial biopsies from 14 subjects with mild to moderate asthma and 15 control subjects, we quantified smooth muscle cell morphology by stereology and the expression of a panel of genes related to a hypercontractile phenotype of airway smooth muscle, using laser microdissection and two-step real-time polymerase chain reaction. We found that airway smooth muscle cell size was similar in both groups, but cell number was nearly twofold higher in subjects with asthma (p = 0.03), and the amount of smooth muscle in the submucosa was increased 50-83% (p < 0.005). Gene expression profiling in smooth muscle cells showed no difference in the expression of genes encoding phenotypic markers in cells from healthy subjects and subjects with asthma (all p > 0.1). We conclude that airway smooth muscle proliferation is a pathologic characteristic of subjects with mild to moderate asthma. However, smooth muscle cells in mild to moderate asthma do not show hypertrophy or gene expression changes of a hypercontractile phenotype observed in vitro.


Asunto(s)
Asma , Expresión Génica/fisiología , Músculo Liso , Adulto , Asma/genética , Asma/patología , Asma/fisiopatología , Biopsia , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/fisiopatología , Estudios de Casos y Controles , División Celular , Tamaño de la Célula , Estudios Transversales , Femenino , Volumen Espiratorio Forzado , Perfilación de la Expresión Génica , Humanos , Hiperplasia , Hipertrofia , Masculino , Microdisección , Microscopía Confocal , Persona de Mediana Edad , Músculo Liso/patología , Músculo Liso/fisiopatología , Miocitos del Músculo Liso/patología , Fenotipo , Reacción en Cadena de la Polimerasa , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...