Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Gut ; 72(10): 1838-1847, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36788014

RESUMEN

OBJECTIVES: Ulcerative colitis (UC) is a chronic inflammatory disorder of unknown aetiology. Gut virome dysbiosis is fundamental in UC progression, although its role in the early phases of the disease is far from fully understood. Therefore, we sought to investigate the role of a virome-associated protein encoded by the Orthohepadnavirus genus, the hepatitis B virus X protein (HBx), in UC aetiopathogenesis. DESIGN: HBx positivity of UC patient-derived blood and gut mucosa was assessed by RT-PCR and Sanger sequencing and correlated with clinical characteristics by multivariate analysis. Transcriptomics was performed on HBx-overexpressing endoscopic biopsies from healthy donors.C57BL/6 mice underwent intramucosal injections of liposome-conjugated HBx-encoding plasmids or the control, with or without antibiotic treatment. Multidimensional flow cytometry analysis was performed on colonic samples from HBx-treated and control animals. Transepithelial electrical resistance measurement, proliferation assay, chromatin immunoprecipitation assay with sequencing and RNA-sequencing were performed on in vitro models of the gut barrier. HBx-silencing experiments were performed in vitro and in vivo. RESULTS: HBx was detected in about 45% of patients with UC and found to induce colonic inflammation in mice, while its silencing reverted the colitis phenotype in vivo. HBx acted as a transcriptional regulator in epithelial cells, provoking barrier leakage and altering both innate and adaptive mucosal immunity ex vivo and in vivo. CONCLUSION: This study described HBx as a contributor to the UC pathogenesis and provides a new perspective on the virome as a target for tailored treatments.


Asunto(s)
Colitis Ulcerosa , Colitis , Animales , Ratones , Colitis Ulcerosa/patología , Viroma , Ratones Endogámicos C57BL , Colon/patología , Colitis/metabolismo , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Modelos Animales de Enfermedad , Sulfato de Dextran
2.
J Ultrasound Med ; 42(8): 1717-1728, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36744584

RESUMEN

OBJECTIVES: Recognition of intestinal lesions with substantial fibrosis is strategic for optimal management of patients with Crohn's disease (CD). We aimed to assess the relationships between intestinal ultrasound parameters and histopathologic findings in a prospective cohort of patients with CD undergoing surgery. METHODS: Seventeen consecutive adult CD patients with involvement of the terminal ileum or the sigmoid colon who underwent bowel resective surgeries were enrolled and performed intestinal ultrasound (IUS) within 30 days prior to surgery. Uni- and multivariable analyses were used to assess the relationships between IUS parameters and histopathological elements of lesions. RESULTS: Sensitivity, specificity, accuracy, PPV and NPV (95% CI) of IUS in detecting stricturing and penetrating complications (surgical specimen as reference standard) were 93% (68-100), 86% (42-100), 91% (71-99), 93% (68-100) and 86% (42-100), and 78% (40-97), 92% (64-100), 86% (65-97), 88% (47-100) and 86% (57-98), respectively. Only the presence of hyperechogenic spiculates was statistically significantly associated with collagen content (b = 7.29, 95% CI = 1.88/12.69, P = .012), while only the presence of vascular signals at color Doppler (Limberg score 3 or 4) was significantly associated with active inflammation (OR = 10.0, 95% CI = 0.9/108.6, P = .037). There was a strong correlation between IUS and histological measurements of the wall thickness (r = 0.67, P = .01). CONCLUSIONS: The presence of hyperechogenic spiculates was associated with the presence of fibrosis, while the presence of marked vascular signals was associated with the presence of inflammation. Wall thickness measured by IUS was reliable and reproducible in comparison with histological measurement.


Asunto(s)
Enfermedad de Crohn , Adulto , Humanos , Enfermedad de Crohn/diagnóstico por imagen , Enfermedad de Crohn/cirugía , Estudios Prospectivos , Inflamación , Fibrosis , Colon Sigmoide
3.
Stem Cells ; 37(7): 973-987, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30942926

RESUMEN

Mesenchymal stem cells (MSCs) are well established to have promising therapeutic properties. TNF-stimulated gene-6 (TSG-6), a potent tissue-protective and anti-inflammatory factor, has been demonstrated to be responsible for a significant part of the tissue-protecting properties mediated by MSCs. Nevertheless, current knowledge about the biological function of TSG-6 in MSCs is limited. Here, we demonstrated that TSG-6 is a crucial factor that influences many functional properties of MSCs. The transcriptomic sequencing analysis of wild-type (WT) and TSG-6-/- -MSCs shows that the loss of TSG-6 expression leads to the perturbation of several transcription factors, cytokines, and other key biological pathways. TSG-6-/- -MSCs appeared morphologically different with dissimilar cytoskeleton organization, significantly reduced size of extracellular vesicles, decreased cell proliferative rate, and loss of differentiation abilities compared with the WT cells. These cellular effects may be due to TSG-6-mediated changes in the extracellular matrix (ECM) environment. The supplementation of ECM with exogenous TSG-6, in fact, rescued cell proliferation and changes in morphology. Importantly, TSG-6-deficient MSCs displayed an increased capacity to release interleukin-6 conferring pro-inflammatory and pro-tumorigenic properties to the MSCs. Overall, our data provide strong evidence that TSG-6 is crucial for the maintenance of stemness and other biological properties of murine MSCs.


Asunto(s)
Moléculas de Adhesión Celular/genética , Transformación Celular Neoplásica/genética , Interleucina-6/genética , Células Madre Mesenquimatosas/metabolismo , Transcriptoma , Animales , Comunicación Autocrina/genética , Moléculas de Adhesión Celular/deficiencia , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Citocinas/genética , Citocinas/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Matriz Extracelular/química , Matriz Extracelular/genética , Vesículas Extracelulares/química , Vesículas Extracelulares/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Interleucina-6/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Redes y Vías Metabólicas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Int J Cancer ; 145(7): 1913-1920, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30889293

RESUMEN

Colorectal cancer (CRC) is one of the most malignant tumors worldwide. Stromal cells residing in the tumor microenvironment strongly contribute to cancer progression through their crosstalk with cancer cells and extracellular matrix. Here we provide the first evidence that CRC-associated lymphatic endothelium displays a distinct matrisome-associated transcriptomic signature, which distinguishes them from healthy intestinal lymphatics. We also demonstrate that CRC-associated human intestinal lymphatic endothelial cells regulate tumor cell growth via growth differentiation factor 11, a soluble matrisome component which in CRC patients was found to be associated with tumor progression. Our data provide new insights into lymphatic contribution to CRC growth, aside from their conventional role as conduits of metastasis.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Neoplasias Colorrectales/genética , Endotelio Linfático/citología , Matriz Extracelular/genética , Factores de Diferenciación de Crecimiento/genética , Animales , Células CACO-2 , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Células Endoteliales/química , Células Endoteliales/citología , Endotelio Linfático/química , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Trasplante de Neoplasias , Microambiente Tumoral
5.
Gastroenterology ; 153(5): 1363-1377.e6, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28827082

RESUMEN

BACKGROUND & AIMS: Alterations in signaling pathways that regulate resolution of inflammation (resolving pathways) contribute to pathogenesis of ulcerative colitis (UC). The resolution process is regulated by lipid mediators, such as those derived from the ω-3 docosahexaenoic acid (DHA), whose esterified form is transported by the major facilitator superfamily domain containing 2A (MFSD2A) through the endothelium of brain, retina, and placenta. We investigated if and how MFSD2A regulates lipid metabolism of gut endothelial cells to promote resolution of intestinal inflammation. METHODS: We performed lipidomic and functional analyses of MFSD2A in mucosal biopsies and primary human intestinal microvascular endothelial cells (HIMECs) isolated from surgical specimens from patients with active, resolving UC and healthy individuals without UC (controls). MFSD2A was knocked down in HIMECs with small hairpin RNAs or overexpressed from a lentiviral vector. Human circulating endothelial progenitor cells that overexpress MFSD2A were transferred to CD1 nude mice with dextran sodium sulfate-induced colitis, with or without oral administration of DHA. RESULTS: Colonic biopsies from patients with UC had reduced levels of inflammation-resolving DHA-derived epoxy metabolites compared to healthy colon tissues or tissues with resolution of inflammation. Production of these metabolites by HIMECs required MFSD2A, which is required for DHA retention and metabolism in the gut vasculature. In mice with colitis, transplanted endothelial progenitor cells that overexpressed MFSD2A not only localized to the inflamed mucosa but also restored the ability of the endothelium to resolve intestinal inflammation, compared with mice with colitis that did not receive MFSD2A-overexpressing endothelial progenitors. CONCLUSIONS: Levels of DHA-derived epoxides are lower in colon tissues from patients with UC than healthy and resolving mucosa. Production of these metabolites by gut endothelium requires MFSD2A; endothelial progenitor cells that overexpress MFSD2A reduce colitis in mice. This pathway might be induced to resolve intestinal inflammation in patients with colitis.


Asunto(s)
Colitis/prevención & control , Colon/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Células Progenitoras Endoteliales/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Colon/efectos de los fármacos , Colon/patología , Sistema Enzimático del Citocromo P-450/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Compuestos Epoxi/metabolismo , Humanos , Proteínas de Transporte de Membrana/genética , Ratones Desnudos , Oxilipinas/metabolismo , Interferencia de ARN , Transducción de Señal , Simportadores , Transfección , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Supresoras de Tumor/genética
6.
J Pathol ; 241(4): 547-558, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27981571

RESUMEN

Prolyl hydroxylase domain-containing proteins (PHDs) regulate the adaptation of cells to hypoxia. Pan-hydroxylase inhibition is protective in experimental colitis, in which PHD1 plays a prominent role. However, it is currently unknown how PHD1 targeting regulates this protection and which cell type(s) are involved. Here, we demonstrated that Phd1 deletion in endothelial and haematopoietic cells (Phd1f/f Tie2:cre) protected mice from dextran sulphate sodium (DSS)-induced colitis, with reduced epithelial erosions, immune cell infiltration, and colonic microvascular dysfunction, whereas the response of Phd2f/+ Tie2:cre and Phd3f/f Tie2:cre mice to DSS was similar to that of their littermate controls. Using bone marrow chimeras and cell-specific cre mice, we demonstrated that ablation of Phd1 in haematopoietic cells but not in endothelial cells was both necessary and sufficient to inhibit experimental colitis. This effect relied, at least in part, on skewing of Phd1-deficient bone marrow-derived macrophages towards an anti-inflammatory M2 phenotype. These cells showed an attenuated nuclear factor-κB-dependent response to lipopolysaccharide (LPS), which in turn diminished endothelial chemokine expression. In addition, Phd1 deficiency in dendritic cells significantly reduced interleukin-1ß production in response to LPS. Taken together, our results further support the development of selective PHD1 inhibitors for ulcerative colitis, and identify haematopoietic cells as their primary target. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Macrófagos/metabolismo , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/efectos de los fármacos , Colon/patología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Eliminación de Gen , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/genética
7.
Int J Mol Sci ; 18(6)2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28617319

RESUMEN

BACKGROUND: In the last few decades, a better knowledge of the inflammatory pathways involved in the pathogenesis of Inflammatory Bowel Disease (IBD) has promoted biological therapy as an important tool to treat IBD patients. However, in spite of a wider spectrum of biological drugs, a significant proportion of patients is unaffected by or lose their response to these compounds, along with increased risks of infections and malignancies. For these reasons there is an urgent need to look for new pharmacological targets. The novel Phosphodiesterase 4 (PDE4) inhibitors have been recently introduced as new modulators of intracellular signals and gene transcription for the treatment of IBD. AIM: To discuss and describe the state of the art of this new class of compounds in the IBD field, with particular attention to apremilast. METHODS: Published articles selected from PubMed were comprehensively reviewed, with key words including apremilast, inflammatory disease, IBD, psoriasis, psoriatic arthritis, pathogenesis, therapies, and treatment. RESULTS: PDE4 inhibitors generate elevated intracellular levels of cyclic Adenosine Monophosphate (cAMP), that consequently down-regulate the release of pro-inflammatory cytokines in the mucosa of IBD patients. The newly developed apremilast is one of these drugs and has already been approved for the treatment of dermatologic/rheumatologic inflammatory conditions; studies in psoriasis and psoriatic arthritis have in fact demonstrated its clinical activity. However, no clinical trials have yet been published on the use of apremilast in IBD. CONCLUSION: In light of the similarity of pro-inflammatory signaling pathways across the gut, the skin, and joints, apremilast is likely supposed to show its efficacy also in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Talidomida/análogos & derivados , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/análisis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Enfermedades Inflamatorias del Intestino/enzimología , Terapia Molecular Dirigida/métodos , Inhibidores de Fosfodiesterasa 4/farmacología , Talidomida/farmacología , Talidomida/uso terapéutico
8.
Gastroenterology ; 148(7): 1438-51.e8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25754161

RESUMEN

BACKGROUND & AIMS: Colorectal cancer (CRC) is highly metastatic. Metastases spread directly into local tissue or invade distant organs via blood and lymphatic vessels, but the role of lymphangiogenesis in CRC progression has not been determined. Lymphangiogenesis is induced via vascular endothelial growth factor C (VEGFC) activation of its receptor, VEGFR3; high levels of VEGFC have been measured in colorectal tumors undergoing lymphangiogenesis and correlated with metastasis. We investigated VEGFC signaling and lymphatic barriers in human tumor tissues and mice with orthotopic colorectal tumors. METHODS: We performed immunohistochemical, immunoblot, and real-time polymerase chain reaction analyses of colorectal tumor specimens collected from patients; healthy intestinal tissues collected during operations of patients without CRC were used as controls. CT26 CRC cells were injected into the distal posterior rectum of BALB/c-nude mice. Mice were given injections of an antibody against VEGFR3 or an adenovirus encoding human VEGFC before orthotopic tumors and metastases formed. Lymph node, lung, and liver tissues were collected and evaluated by flow cytometry. We measured expression of vascular endothelial cadherin (CDH5) on lymphatic vessels in mice and in human intestinal lymphatic endothelial cells. RESULTS: Levels of podoplanin (a marker of lymphatic vessels), VEGFC, and VEGFR3 were increased in colorectal tumor tissues, compared with controls. Mice that expressed VEGFC from the adenoviral vector had increased lymphatic vessel density and more metastases in lymph nodes, lungs, and livers, compared with control mice. Anti-VEGFR3 antibody reduced numbers of lymphatic vessels in colons and prevented metastasis. Expression of VEGFC compromised the lymphatic endothelial barrier in mice and endothelial cells, reducing expression of CDH5, increasing permeability, and increasing trans-endothelial migration by CRC cells. Opposite effects were observed in mice and cells when VEGFR3 was blocked. CONCLUSIONS: VEGFC signaling via VEGFR3 promotes lymphangiogenesis and metastasis by orthotopic colorectal tumors in mice and reduces lymphatic endothelial barrier integrity. Levels of VEGFC and markers of lymphatic vessels are increased in CRC tissues from patients, compared with healthy intestine. Strategies to block VEGFR3 might be developed to prevent CRC metastasis in patients.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Migración Transendotelial y Transepitelial , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células Endoteliales/patología , Endotelio Linfático/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Linfangiogénesis , Metástasis Linfática , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Transducción de Señal , Factores de Tiempo , Transfección , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Gastroenterology ; 149(1): 163-176.e20, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25790743

RESUMEN

BACKGROUND & AIMS: Mesenchymal stem cells (MSCs) are pluripotent cells that can promote expansion of immune regulatory cells and might be developed for the treatment of immune disorders, including inflammatory bowel diseases. MSCs were reported to reduce colitis in mice; we investigated whether MSC localization to the intestine and production of paracrine factors, including tumor necrosis factor-induced protein 6 (TSG6), were required for these effects. METHODS: MSCs were isolated from bone marrow (BM-MSCs) of 4- to 6-week-old C57BL/6, C57BL/6-green fluorescent protein, or Balb/c Tsg6-/- male mice. Colitis was induced by ad libitum administration of dextran sulfate sodium for 10 days; after 5 days the mice were given intraperitoneal injections of BM-MSCs or saline (controls). Blood samples and intestinal tissues were collected 24, 48, 96, and 120 hours later; histologic and flow cytometry analyses were performed. RESULTS: Injection of BM-MSCs reduced colitis in mice, increasing body weight and reducing markers of intestinal inflammation, compared with control mice. However, fewer than 1% of MSCs reached the inflamed colon. Most of the BM-MSCs formed aggregates in the peritoneal cavity. The aggregates contained macrophages and B and T cells, and produced immune-regulatory molecules including FOXP3, interleukin (IL)10, transforming growth factor-ß, arginase type II, chemokine (C-C motif) ligand 22 (CCL22), heme oxygenase-1, and TSG6. Serum from mice given BM-MSCs, compared with mice given saline, had increased levels of TSG6. Injection of TSG6 reduced the severity of colitis in mice, along with the numbers of CD45+ cells, neutrophils and metalloproteinase activity in the mucosa, while increasing the percentage of Foxp3CD45+ cells. TSG6 injection also promoted the expansion of regulatory macrophages that expressed IL10 and inducible nitric oxide synthase, and reduced serum levels of interferon-γ, IL6, and tumor necrosis factor. Tsg6-/- MSCs did not suppress the mucosal inflammatory response in mice with colitis. CONCLUSIONS: BM-MSCs injected into mice with colitis do not localize to the intestine but instead form aggregates in the peritoneum where they produce immunoregulatory molecules, including TSG6, that reduce intestinal inflammation. TSG6 is sufficient to reduce intestinal inflammation in mice with colitis.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Colitis/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Citocinas/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Intestinos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento
10.
Gut ; 64(4): 589-600, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24848264

RESUMEN

OBJECTIVE: Inflammation plays crucial roles in the pathogenesis of several chronic inflammatory disorders, including Crohn's disease (CD) and UC, the two major forms of IBD. The urokinase plasminogen activator receptor (uPAR) exerts pleiotropic functions over the course of both physiological and pathological processes. uPAR not only has a key role in fibrinolysis but also modulates the development of protective immunity. Additionally, uPAR supports extracellular matrix degradation and regulates cell migration, adhesion and proliferation, thus influencing the development of inflammatory and immune responses. This study aimed to evaluate the role of uPAR in the pathogenesis of IBD. DESIGN: The functional role of uPAR was assessed in established experimental models of colitis. uPAR deficiency effects on cytokine release, polarisation and bacterial phagocytosis were analysed in colonic macrophages. uPAR expression was analysed in surgical specimens collected from normal subjects and patients with IBD. RESULTS: In mice, uPAR expression is positively regulated as colitis progresses. uPAR-KO mice displayed severe inflammation compared with wild-type littermates, as indicated by clinical assessment, endoscopy and colon histology. The absence of uPAR led to an increased production of inflammatory cytokines by macrophages that showed an M1 polarisation and impaired phagocytosis. In human IBD, CD68(+) macrophages derived from the inflamed mucosa expressed low levels of uPAR. CONCLUSIONS: These findings point to uPAR as an essential component of intestinal macrophage functions and unravel a new potential target to control mucosal inflammation in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Macrófagos/fisiología , Fagocitosis/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Curr Opin Gastroenterol ; 29(6): 608-13, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24100721

RESUMEN

PURPOSE OF REVIEW: The review summarizes the current knowledge of the roles played by the vascular and lymphatic endothelium throughout the gut in the pathogenesis of inflammatory bowel disease (IBD) and gives an update on emerging strategies targeting both vasculatures. RECENT FINDINGS: Enormous efforts have been made to understand the mechanisms underlining the origin, development and maintenance of intestinal chronic inflammation. In particular, new studies focused their attention on the role played by the microvascular and lymphatic endothelium in the pathogenesis of IBD. During inflammation, whereas the microvasculature is responsible for the entry and distribution of immune cells in the mucosa, the lymphatic system controls leukocyte exit, bacterial clearance and edema absorption. The study of these events, which are aberrant during chronic inflammation, has resulted in the identification and validation of several targets for the treatment of experimental colitis, some of which have translated into effective treatments for patients with IBD. SUMMARY: Although much attention has been paid to the microvascular endothelium and to antiangiogenic therapies, specific studies on the lymphatic vasculature and its functions in IBD are still at the initial stage, and other molecular mechanisms, genes, molecules and new pathways must definitely be explored.


Asunto(s)
Endotelio Linfático/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Endotelio Linfático/fisiopatología , Endotelio Vascular/fisiopatología , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/fisiopatología , Intestinos/irrigación sanguínea , Linfangiogénesis/efectos de los fármacos , Terapia Molecular Dirigida/métodos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/etiología
12.
Cell Mol Life Sci ; 69(16): 2791-803, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22415324

RESUMEN

Chromogranin A (CgA), a secretory protein expressed by many neuroendocrine cells, neurons, cardiomyocytes, and keratinocytes, is the precursor of various peptides that regulate the carbohydrate/lipid metabolism and the cardiovascular system. We have found that CgA, locally administered to injured mice, can accelerate keratinocyte proliferation and wound healing. This biological activity was abolished by the Asp(45)Glu mutation. CgA and its N-terminal fragments, but not the corresponding Asp(45)Glu mutants, could selectively recognize the αvß6-integrin on keratinocytes (a cell-adhesion receptor that is up-regulated during wound healing) and regulate keratinocyte adhesion, proliferation, and migration. No binding was observed to other integrins such as αvß3, αvß5, αvß8, α5ß1, α1ß1, α3ß1, α6ß4, α6ß7 and α9ß1. Structure-activity studies showed that the entire CgA(39-63) region is crucial for αvß6 recognition (K(i) = 7 nM). This region contains an RGD site (residues CgA(43-45)) followed by an amphipathic α-helix (residues CgA(47-63)), both crucial for binding affinity and selectivity. These results suggest that the interaction of the RGD/α-helix motif of CgA with αvß6 regulates keratinocyte physiology in wound healing.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Cromogranina A/metabolismo , Fibroblastos/metabolismo , Integrinas/metabolismo , Queratinocitos/metabolismo , Oligopéptidos/metabolismo , Piel/metabolismo , Cicatrización de Heridas/fisiología , Secuencia de Aminoácidos , Animales , Unión Competitiva , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Fibroblastos/citología , Humanos , Queratinocitos/citología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Secretina/análogos & derivados , Secretina/metabolismo , Homología de Secuencia de Aminoácido , Piel/citología
14.
Nat Rev Gastroenterol Hepatol ; 19(3): 169-184, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34876680

RESUMEN

Intestinal fibrosis, which is usually the consequence of chronic inflammation, is a common complication of inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis. In the past few years, substantial advances have been made in the areas of pathogenesis, diagnosis and management of intestinal fibrosis. Of particular interest have been inflammation-independent mechanisms behind the gut fibrotic process, genetic and environmental risk factors (such as the role of the microbiota), and the generation of new in vitro and in vivo systems to study fibrogenesis in the gut. A huge amount of work has also been done in the area of biomarkers to predict or detect intestinal fibrosis, including novel cross-sectional imaging techniques. In parallel, researchers are embarking on developing and validating clinical trial end points and protocols to test novel antifibrotic agents, although no antifibrotic therapies are currently available. This Review presents the state of the art on the most recently identified pathogenic mechanisms of this serious IBD-related complication, focusing on possible targets of antifibrotic therapies, management strategies, and factors that might predict fibrosis progression or response to treatment.


Asunto(s)
Colitis Ulcerosa , Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Enfermedad Crónica , Enfermedad de Crohn/tratamiento farmacológico , Fibrosis , Humanos , Inflamación/complicaciones , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/terapia
15.
Nat Comput Sci ; 1(8): 511-515, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38217242

RESUMEN

Inflammatory bowel disease (IBD) is a class of chronic disorders whose etiogenesis is still unknown. Despite the high number of IBD-related omics studies, the RNA-sequencing data produced results that are hard to compare because of the experimental variability and different data analysis approaches. We here introduce the IBD Transcriptome and Metatranscriptome Meta-Analysis (TaMMA) framework, a comprehensive survey of publicly available IBD RNA-sequencing datasets. IBD TaMMA is an open-source platform where scientists can explore simultaneously the freely available IBD-associated transcriptomics and microbial profiles thanks to its interactive interface, resulting in a useful tool to the IBD community.

16.
Cancers (Basel) ; 12(8)2020 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-32722560

RESUMEN

Inflammation is a recognized hallmark of cancer that contributes to the development and progression of colorectal cancer (CRC). Anti-inflammatory drugs currently used for the treatment of CRC show many adverse side effects that prompted researchers to propose the polyunsaturated fatty acids-derived specialized pro-resolving mediators (SPMs) as promoters of resolution of cancer-associated inflammation. SPMs were found to inhibit the CRC-associated pro-inflammatory milieu via specific G-coupled protein receptors, although clinical data are still lacking. This review aims to summarize the state-of-the-art in this field, ultimately providing insights for the development of innovative anti-CRC therapies that promote the endogenous lipid-mediated resolution of CRC-associated inflammation.

17.
J Exp Clin Cancer Res ; 39(1): 253, 2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-33225975

RESUMEN

BACKGROUND: Sphingosine-1-phosphate receptor 2 (S1PR2) mediates pleiotropic functions encompassing cell proliferation, survival, and migration, which become collectively de-regulated in cancer. Information on whether S1PR2 participates in colorectal carcinogenesis/cancer is scanty, and we set out to fill the gap. METHODS: We screened expression changes of S1PR2 in human CRC and matched normal mucosa specimens [N = 76]. We compared CRC arising in inflammation-driven and genetically engineered models in wild-type (S1PR2+/+) and S1PR2 deficient (S1PR2-/-) mice. We reconstituted S1PR2 expression in RKO cells and assessed their growth in xenografts. Functionally, we mimicked the ablation of S1PR2 in normal mucosa by treating S1PR2+/+ organoids with JTE013 and characterized intestinal epithelial stem cells isolated from S1PR2-/-Lgr5-EGFP- mice. RESULTS: S1PR2 expression was lost in 33% of CRC; in 55%, it was significantly decreased, only 12% retaining expression comparable to normal mucosa. Both colitis-induced and genetic Apc+/min mouse models of CRC showed a higher incidence in size and number of carcinomas and/or high-grade adenomas, with increased cell proliferation in S1PR2-/- mice compared to S1PR2+/+ controls. Loss of S1PR2 impaired mucosal regeneration, ultimately promoting the expansion of intestinal stem cells. Whereas its overexpression attenuated cell cycle progression, it reduced the phosphorylation of AKT and augmented the levels of PTEN. CONCLUSIONS: In normal colonic crypts, S1PR2 gains expression along with intestinal epithelial cells differentiation, but not in intestinal stem cells, and contrasts intestinal tumorigenesis by promoting epithelial differentiation, preventing the expansion of stem cells and braking their malignant transformation. Targeting of S1PR2 may be of therapeutic benefit for CRC expressing high Lgr5.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Células Epiteliales/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Células Madre/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proliferación Celular/fisiología , Neoplasias Colorrectales/patología , Femenino , Genes Supresores de Tumor , Humanos , Masculino , Ratones , Persona de Mediana Edad
18.
Mol Biol Cell ; 17(1): 367-78, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16267271

RESUMEN

Urokinase-type plasminogen activator (uPA) binding to uPAR induces migration, adhesion, and proliferation through multiple interactions with G proteins-coupled receptor FPRL1, integrins, or the epidermal growth factor (EGF) receptor (EGFR). At least two forms of uPAR are present on the cell surface: full-length and cleaved uPAR, each specifically interacting with one or more transmembrane proteins. The connection between these interactions and the effects on the signaling pathways activation is not clear. We have exploited an uPAR mutant (hcr, human cleavage resistant) to dissect the pathways involved in uPA-induced cell migration. This mutant is not cleaved by proteases, is glycosylphosphatidylinositol anchored, and binds uPA with a normal K(d). Both wild-type (wt) and hcr-uPAR are able to mediate uPA-induced migration, are constitutively associated with the EGFR, and associate with alpha3beta1 integrin upon uPA binding. However, they engage different pathways in response to uPA. wt-uPAR requires both integrins and FPRL1 to mediate uPA-induced migration, and association of wt-uPAR to alpha3beta1 results in uPAR cleavage and extracellular signal-regulated kinase (ERK) activation. On the contrary, hcr-uPAR does not activate ERK and does not engage FPRL1 or any other G protein-coupled receptor, but it activates an alternative pathway initiated by the formation of a triple complex (uPAR-alpha3beta1-EGFR) and resulting in the autotyrosine phosphorylation of EGFR.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Mutación/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Secuencia de Aminoácidos , Animales , Línea Celular , Activación Enzimática , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Integrinas/metabolismo , Ratones , Datos de Secuencia Molecular , Péptido Hidrolasas/metabolismo , Receptores de Superficie Celular/química , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Factores de Tiempo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
19.
United European Gastroenterol J ; 7(8): 999-1007, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31662858

RESUMEN

The association of intestinal dysbiosis with the pathogenesis of inflammatory bowel disease has been well established. Besides bacteria, microbiota comprises yeasts, archaea, protists and viruses, neglected actors in inflammatory bowel disease-associated microbiota. In the past, a great limitation in studying microbiota composition was the low sensitivity of sequencing technologies and that few computational approaches were sufficient to thoroughly analyse the whole microbiome. However, new cutting-edge technologies in nucleic acid sequencing, -omics analysis and the innovative statistics and bioinformatics pipelines made possible more sensitive and accurate metagenomics, ultimately identifying novel players in intestinal inflammation, including prokaryotic and eukaryotic viruses, that together form the gut virome. The discovery of peculiar inflammatory bowel disease-associated microbial strains will not only shed new light on inflammatory bowel disease aetiogenesis, they may also support the development of novel therapeutic strategies not merely treating symptoms, but precisely counteracting the primary cause of chronic intestinal inflammation.


Asunto(s)
Bacterias/clasificación , Disbiosis/complicaciones , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/virología , Intestinos/virología , Metagenómica/métodos , Animales , Infecciones por Caliciviridae/complicaciones , Biología Computacional/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Inflamación/virología , Enfermedades Inflamatorias del Intestino/terapia , Microbiota/genética , Modelos Animales , Terapia Molecular Dirigida/métodos , Norovirus/genética , Filogenia , Virus/genética
20.
Gut Microbes ; 10(2): 149-158, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30252582

RESUMEN

Intestinal dysbiosis is one of the causes underlying the pathogenesis of inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD). Besides bacteria, microbiota comprises both prokaryotic and eukaryotic viruses, that together compose the gut virome. Few works have defined the viral composition of stools, while the virome populating intestinal mucosae from early-diagnosed IBD patients has never been studied. Here we show that, by in-depth metagenomic analysis of RNA-Seq data obtained from gut mucosae of young treatment-naïve patients, early-diagnosed for CD and UC, and from healthy subjects (Ctrl), UC patients display significantly higher levels of eukaryotic Hepadnaviridae transcripts by comparison with both Ctrl and CD patients, whereas CD patients show increased abundance of Hepeviridae versus Ctrl. Moreover, we found that UC gut mucosa is characterized by lower levels of Polydnaviridae and Tymoviridae, whereas the mucosa of patients with CD showed a reduced abundance of Virgaviridae. Our findings support the idea that certain eukaryotic viruses might trigger intestinal inflammation and contribute to IBD pathogenesis and pave the way not only for the discovery of novel diagnostic biomarkers but also for the development of anti-viral drugs for the treatment of IBD.


Asunto(s)
Microbioma Gastrointestinal/genética , Enfermedades Inflamatorias del Intestino/virología , Mucosa Intestinal/virología , Adolescente , Niño , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/patología , Colitis Ulcerosa/virología , Enfermedad de Crohn/diagnóstico , Enfermedad de Crohn/patología , Enfermedad de Crohn/virología , Bases de Datos Factuales , Heces/virología , Femenino , Humanos , Inflamación/patología , Inflamación/virología , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Masculino , Metagenómica , Virus/clasificación , Virus/genética , Virus/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA