Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Gut ; 73(2): 282-297, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-37884352

RESUMO

OBJECTIVE: We sought to investigate the role of interleukin (IL)-20 in IBD and experimental colitis. DESIGN: Experimental colitis was induced in mice deficient in components of the IL-20 and signal transducer and activator of transcription (STAT)2 signalling pathways. In vivo imaging, high-resolution mini-endoscopy and histology were used to assess intestinal inflammation. We further used RNA-sequencing (RNA-Seq), RNAScope and Gene Ontology analysis, western blot analysis and co-immunoprecipitation, confocal microscopy and intestinal epithelial cell (IEC)-derived three-dimensional organoids to investigate the underlying molecular mechanisms. Results were validated using samples from patients with IBD and non-IBD control subjects by a combination of RNA-Seq, organoids and immunostainings. RESULTS: In IBD, IL20 levels were induced during remission and were significantly higher in antitumour necrosis factor responders versus non-responders. IL-20RA and IL-20RB were present on IECs from patients with IBD and IL-20-induced STAT3 and suppressed interferon (IFN)-STAT2 signalling in these cells. In IBD, experimental dextran sulfate sodium (DSS)-induced colitis and mucosal healing, IECs were the main producers of IL-20. Compared with wildtype controls, Il20-/-, Il20ra-/- and Il20rb-/- mice were more susceptible to experimental DSS-induced colitis. IL-20 deficiency was associated with increased IFN/STAT2 activity in mice and IFN/STAT2-induced necroptotic cell death in IEC-derived organoids could be markedly blocked by IL-20. Moreover, newly generated Stat2ΔIEC mice, lacking STAT2 in IECs, were less susceptible to experimental colitis compared with wildtype controls and the administration of IL-20 suppressed colitis activity in wildtype animals. CONCLUSION: IL-20 controls colitis and mucosal healing by interfering with the IFN/STAT2 death signalling pathway in IECs. These results indicate new directions for suppressing gut inflammation by modulating IL-20-controlled STAT2 signals.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Humanos , Animais , Camundongos , Mucosa Intestinal/metabolismo , Colite/metabolismo , Interleucinas/metabolismo , Inflamação/metabolismo , Células Epiteliais/metabolismo , Doenças Inflamatórias Intestinais/genética , Sulfato de Dextrana/farmacologia , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT2/metabolismo
2.
Gastroenterology ; 160(3): 925-928.e4, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33075345
3.
Gastroenterology ; 157(5): 1310-1322.e13, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31352002

RESUMO

BACKGROUND & AIMS: Interferon lambda (IFNL) is expressed at high levels by intestinal epithelial cells (IECs) and mucosal immune cells in response to infection and inflammation. We investigated whether IFNL might contribute to pathogenesis of Crohn's disease (CD). METHODS: We obtained serum samples and terminal ileum biopsies from 47 patients with CD and 16 healthy individuals (controls). We measured levels of IFNL by enzyme-linked immunosorbent assay and immunohistochemistry and location of expression by confocal microscopy. Activation of IFNL signaling via STAT1 was measured in areas of no, mild, moderate, and severe inflammation and correlated with Paneth cell homeostasis and inflammation. IFNL expression and function were studied in wild-type mice and mice with intestinal epithelial cell-specific (ΔIEC) disruption or full-body disruption of specific genes (Mlkl-/-, Stat1ΔIEC, Casp8ΔIEC, Casp8ΔIECRipk3-/-, Casp8ΔIECTnfr-/-, Casp8ΔIECMlkl-/-, and Nod2-/- mice). Some mice were given tail vein injections of a vector encoding a secreted form of IFNL. Intestinal tissues were collected from mice and analyzed by immunohistochemistry and immunoblots. We generated 3-dimensional small intestinal organoids from mice and studied the effects of IFNL and inhibitors of STAT-signaling pathway. RESULTS: Patients with CD had significant increases in serum and ileal levels of IFNL compared with controls. Levels of IFNL were highest in ileum tissues with severe inflammation. High levels of IFNL associated with a reduced number of Paneth cells and increased cell death at the crypt bottom in inflamed ileum samples. Intestinal tissues from the ileum of wild-type mice injected with a vector expressing IFNL had reduced numbers of Paneth cells. IFNL-induced death of Paneth cells in mice did not occur via apoptosis, but required Mixed Lineage Kinase Domain Like (MLKL) and activation of Signal transducer and activator of transcription 1 (STAT1). In organoids, inhibitors of Janus kinase (JAK) signaling via STAT1 (glucocorticoids, tofacitinib, or filgotinib) reduced expression of proteins that mediate cell death and prevented Paneth cell death. CONCLUSIONS: Levels of IFNL are increased in serum and inflamed ileal tissues from patients with CD and associated with a loss of Paneth cells. Expression of a secreted form of IFNL in mice results in loss of Paneth cells from intestinal tissues, via STAT1 and MLKL, controlled by caspase 8. Strategies to reduce IFNL or block its effects might be developed for treatment of patients with CD affecting the terminal ileum.


Assuntos
Doença de Crohn/metabolismo , Íleo/metabolismo , Interferons/metabolismo , Interleucinas/metabolismo , Celulas de Paneth/metabolismo , Fator de Transcrição STAT1/metabolismo , Animais , Caspase 8/genética , Caspase 8/metabolismo , Morte Celular , Doença de Crohn/imunologia , Doença de Crohn/patologia , Modelos Animais de Doenças , Humanos , Íleo/imunologia , Íleo/patologia , Interferons/genética , Interleucinas/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Celulas de Paneth/imunologia , Celulas de Paneth/patologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Transdução de Sinais , Técnicas de Cultura de Tecidos , Regulação para Cima
4.
Gut ; 64(5): 743-55, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-24957265

RESUMO

OBJECTIVE: Several pathogenic roles attributed over the past two decades to either T helper (Th)1 or Th2 cells are increasingly becoming associated with interleukin (IL)-17 and most recently IL-9 signalling. However, the implication of IL-9 in IBD has not been addressed so far. DESIGN: We investigated the expression of IL-9 and IL-9R by using peripheral blood, biopsies and surgical samples. We addressed the functional role of IL-9 signalling by analysis of downstream effector proteins. Using Caco-2 cell monolayers we followed the effect of IL-9 on wound healing. RESULTS: IL-9 mRNA expression was significantly increased in inflamed samples from patients with UC as compared with controls. CD3(+) T cells were major IL-9-expressing cells and some polymorphonuclear leucocytes (PMN) also expressed IL-9. IL-9 was co-localised with the key Th9 transcription factors interferon regulatory factor 4 and PU.1. Systemically, IL-9 was abundantly produced by activated peripheral blood lymphocytes, whereas its receptor was overexpressed on gut resident and circulating PMN. IL-9 stimulation of the latter induced IL-8 production in a dose-dependent manner and rendered PMN resistant to apoptosis suggesting a functional role for IL-9R signalling in the propagation of gut inflammation. Furthermore, IL-9R was overexpressed on gut epithelial cells and IL-9 induced STAT5 activation in these cells. Moreover, IL-9 inhibited the growth of Caco-2 epithelial cell monolayers in wound healing experiments. CONCLUSIONS: Our results provide evidence that IL-9 is predominantly involved in the pathogenesis of UC suggesting that targeting IL-9 might become a therapeutic option for patients with UC.


Assuntos
Colite Ulcerativa/imunologia , Interleucina-9/imunologia , Receptores de Interleucina-9/imunologia , Adolescente , Adulto , Idoso , Apoptose/imunologia , Complexo CD3/metabolismo , Células CACO-2 , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Integrina alfa4/sangue , Cadeias beta de Integrinas/sangue , Fatores Reguladores de Interferon/biossíntese , Interleucina-9/biossíntese , Interleucina-9/genética , Mucosa Intestinal/imunologia , Masculino , Pessoa de Meia-Idade , Fosforilação/imunologia , Proteínas Proto-Oncogênicas/biossíntese , RNA Mensageiro/genética , Receptores de Interleucina-9/antagonistas & inibidores , Fator de Transcrição STAT5/metabolismo , Subpopulações de Linfócitos T/imunologia , Transativadores/biossíntese , Regulação para Cima/imunologia , Cicatrização/imunologia , Adulto Jovem
5.
J Cell Mol Med ; 18(9): 1727-39, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25091020

RESUMO

Epidermolysis bullosa acquisita (EBA) is an autoimmune subepidermal blistering disease of mucous membranes and the skin caused by autoantibodies against collagen VII. In silico and wet laboratory epitope mapping studies revealed numerous distinct epitopes recognized by EBA patients' autoantibodies within the non-collagenous (NC)1 and NC2 domains of collagen VII. However, the distribution of pathogenic epitopes on collagen VII has not yet been described. In this study, we therefore performed an in vivo functional epitope mapping of pathogenic autoantibodies in experimental EBA. Animals (n = 10/group) immunized against fragments of the NC1 and NC2 domains of collagen VII or injected with antibodies generated against the same fragments developed to different extent experimental EBA. Our results demonstrate that antibodies targeting multiple, distinct epitopes distributed over the entire NC1, but not NC2 domain of collagen VII induce blistering skin disease in vivo. Our present findings have crucial implications for the development of antigen-specific B- and T cell-targeted therapies in EBA.


Assuntos
Colágeno Tipo VII/imunologia , Epidermólise Bolhosa Adquirida/imunologia , Epitopos/imunologia , Animais , Mapeamento de Epitopos , Feminino , Masculino , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Fragmentos de Peptídeos/imunologia , Estrutura Terciária de Proteína , Coelhos , Pele/imunologia , Pele/patologia
6.
Hum Immunol ; 75(4): 354-63, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24468586

RESUMO

Bullous pemphigoid, the most common autoimmune blistering disease in Western Europe and the USA is characterized by the presence of circulating and tissue-bound autoantibodies against the hemidesmosomal proteins BP230 and BP180/collagen XVII. After binding to their target antigens at the basement membrane of the dermal-epidermal junction these autoantibodies are thought to trigger an inflammatory cascade comprising complement- and granulocyte-dependent reactions that result in tissue damage. Whereas the role of anti-BP180 antibodies has been extensively characterized, few and conflicting data is available on the contribution of anti-BP230 antibodies to bullous pemphigoid pathogenesis. Therefore, we addressed in the present study the role of autoantibodies to BP230 in experimental bullous pemphigoid. Rabbit polyclonal antibodies generated against epitopes of the C-terminal fragment of murine BP230 bound to the basement membrane and activated the complement system ex vivo. Affinity-purified antibodies were subsequently subcutaneously transferred into neonatal and adult BALB/c mice. In vivo, we observed a dose-dependent binding of transferred antibodies in the murine skin; however, there was no complement activation and these mice showed no clinical or histological signs of inflammatory disease, in contrast to mice receiving anti-BP180 antibodies. We further conducted ex vivo experiments and demonstrated that rabbit IgG anti-BP230-specific antibodies, in contrast to antibodies from bullous pemphigoid patients or rabbit IgG anti-BP180 antibodies used as positive controls, did not activate human granulocytes to induce dermal-epidermal separation in skin cryosections. Our present findings demonstrate that antibodies against BP230 are non-pathogenic in experimental models of bullous pemphigoid and suggest that proper activation of the complement and granulocytes represent prerequisites for conferring bullous pemphigoid autoantibodies their tissue destructive potential.


Assuntos
Epitopos Imunodominantes/imunologia , Imunoglobulina G/imunologia , Glicoproteínas de Membrana/imunologia , Penfigoide Bolhoso/imunologia , Domínios e Motivos de Interação entre Proteínas/imunologia , Fatores Etários , Animais , Animais Recém-Nascidos , Autoantígenos/imunologia , Proteínas de Transporte , Proteínas do Citoesqueleto , Derme/imunologia , Derme/metabolismo , Derme/patologia , Modelos Animais de Doenças , Distonina , Epiderme/imunologia , Epiderme/metabolismo , Epiderme/patologia , Imunofluorescência , Expressão Gênica , Humanos , Epitopos Imunodominantes/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso , Colágenos não Fibrilares/imunologia , Fenótipo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Colágeno Tipo XVII
7.
Orphanet J Rare Dis ; 8: 17, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23360583

RESUMO

BACKGROUND: Bullous pemphigoid is a subepidermal blistering disorder associated with tissue-bound and circulating autoantibodies directed mainly to the hemidesmosomal component collagen XVII. While recapitulating the main immunopathological features of the human disease, frank skin blistering does not develop in the absence of skin rubbing in experimental pemphigoid models that have been established in neonatal mice. Moreover, due to their experimental design they only allow for short-term disease observation. In the present study we aimed to establish a model that reproduces the frank skin blistering seen in patients and allows for longer observation times. METHODS: Rabbit and sheep antibodies specific to several fragments of collagen XVII were generated and the purified antibodies were passively transferred into adult mice. RESULTS: Collagen XVII-specific IgG bound to the basal membrane of the skin and mucous membranes activating murine complement in vivo. Mice injected with collagen XVII-specific antibodies, in contrast to mice receiving control antibodies, developed frank skin blistering disease, reproducing human bullous pemphigoid at the clinical, histological and immunopathological levels. Titres of circulating IgG in the serum of mice correlated with the extent of the clinical disease. Mice receiving sheep antibodies specific to murine collagen XVII showed an early onset and a more active disease when compared to litter mates receiving specific rabbit antibodies. CONCLUSION: This novel animal model for bullous pemphigoid should facilitate further investigations of the pathogenesis of bullous pemphigoid and the development of innovative therapies for this disease.


Assuntos
Autoanticorpos/imunologia , Autoantígenos/imunologia , Vesícula/imunologia , Colágenos não Fibrilares/imunologia , Animais , Camundongos , Camundongos Endogâmicos BALB C , Colágeno Tipo XVII
8.
Methods Mol Biol ; 961: 227-42, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23325647

RESUMO

Disease models represent powerful tools used by biomedical researchers to address various questions related to the pathogenesis and the possible therapeutic targets in different diseases. To get a complete picture of the process one needs complementary animal and ex vivo disease models. While subsequent chapters dwell upon animal models, this chapter describes an ex vivo model for granulocyte-dependent dermal-epidermal separation induced by autoantibodies to the basal membrane. The strength of the described ex vivo model resides in the fact that it uses human material (the skin, autoantibodies, and cells), thus better mimicking the pathogenesis as it occurs in patients. The reproducibility rate of the protocol is about 85-90% when using a good combination of the three main ingredients. This rate can drop dramatically when changing one of the three. Nevertheless, being a one-day model, this procedure enables investigators to have a quick readout system when investigating multiple sera for their potential to activate granulocytes and induce dermal-epidermal separation.


Assuntos
Autoanticorpos/imunologia , Derme/patologia , Epiderme/patologia , Granulócitos/patologia , Separação Celular/métodos , Crioultramicrotomia/métodos , Derme/imunologia , Epiderme/imunologia , Granulócitos/imunologia , Humanos , Microscopia/métodos , Modelos Biológicos , Coloração e Rotulagem/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA