Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Immunol ; 53(11): e2350460, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37611637

RESUMEN

Transforming growth factor (TGF)-ß1, a member of the TGF-ß superfamily, is produced by many immune and nonimmune cells and has pleiotropic effects on both innate and adaptive immunity, especially in the control of T-cell differentiation and function. Consistently, loss of TGF-ß1 function is associated with exacerbated T-cell-dependent inflammatory responses that culminate in pathological processes in allergic and immune-mediated diseases. In this review, we highlight the roles of TGF-ß1 in immunity, focusing mainly on its ability to promote differentiation of regulatory T cells, T helper (Th)-17, and Th9 cells, thus contributing to amplifying or restricting T-cell responses in health and human diseases (e.g., inflammatory bowel diseases, type 1 diabetes, asthma, and MS). In addition, we discuss the involvement of Smad7, an inhibitor of TGF-ß1 signaling, in immune-mediated disorders (e.g., psoriasis, rheumatoid arthritis, MS, and inflammatory bowel diseases), as well as the discordant results of clinical trials with mongersen, an oral pharmaceutical compound containing a Smad7 antisense oligonucleotide, in patients with Crohn's disease. Further work is needed to ascertain the reasons for such a discrepancy as well as to identify better candidates for treatment with Smad7 inhibitors.


Asunto(s)
Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Humanos , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Proteína smad7/genética , Proteína smad7/metabolismo , Proteína smad7/uso terapéutico , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
2.
J Pathol ; 259(3): 233-235, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36541916

RESUMEN

Colorectal carcinoma (CRC) is one of the most common forms of malignancy in the Western world. Recent decades have witnessed enormous progress in our understanding of the mechanisms that sustain CRC, even though the factors implicated in the initiation and progression of this neoplasia are not fully understood. A recent study published in The Journal of Pathology looked at the consequences of hyperactivity of chromatin licensing and DNA replication factor 1 (CDT1), a regulator of DNA replication that is produced in excess in CRC, on the course of intestinal tumorigenesis. Mice engineered to selectively overexpress CDT1 and/or lack Geminin, an inhibitor of CDT1, in the intestinal epithelium were more susceptible to experimental intestinal tumorigenesis compared to wild-type mice. This work supports the pro-tumorigenic role of CDT1 and suggests the potential prognostic value of this protein in CRC. © 2022 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Proteínas de Ciclo Celular , Replicación del ADN , Proteínas de Unión al ADN , Animales , Ratones , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina , Proteínas de Unión al ADN/genética , Reino Unido
3.
Int J Mol Sci ; 25(6)2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38542229

RESUMEN

Human cancers are products of multistep processes resulting in abnormal cell growth and differentiation, along with a loss of apoptotic function, leading to the uncontrolled expansion of neoplastic cells and their spread to surrounding tissues and, ultimately, distant parts of the body [...].


Asunto(s)
Neoplasias , Humanos , Diferenciación Celular , Proliferación Celular , Neoplasias/genética , Literatura de Revisión como Asunto
4.
Int J Mol Sci ; 24(10)2023 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-37240286

RESUMEN

Cancer remains one of the most common causes of death worldwide, mainly due to late diagnosis and the lack of efficient therapeutic options for patients with advanced diseases [...].


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Inmunoterapia , Italia
5.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36769214

RESUMEN

Gastric cancer is the sixth most commonly diagnosed cancer and the fourth leading cause of cancer death worldwide. Helicobacter pylori (H. pylori) is one of the main risk factors for this type of neoplasia. Carcinogenetic mechanisms associated with H. pylori are based, on the one hand, on the onset of chronic inflammation and, on the other hand, on bacterial-specific virulence factors that can damage the DNA of gastric epithelial cells and promote genomic instability. Here, we review and discuss the major pathogenetic mechanisms by which H. pylori infection contributes to the onset and development of gastric cancer.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Humanos , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Infecciones por Helicobacter/genética , Mucosa Gástrica/patología
6.
Int J Mol Sci ; 24(7)2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-37047419

RESUMEN

Despite recent advances in treatment approaches, cancer is still one of the leading causes of death worldwide. Restoration of tumor immune surveillance represents a valid strategy to overcome the acquired resistance and cytotoxicity of conventional therapies in oncology and immunotherapeutic drugs, such as immune checkpoint inhibitors and immunogenic cell death inducers, and has substantially progressed the treatment of several malignancies and improved the clinical management of advanced disease. Unfortunately, because of tumor-intrinsic and/or -extrinsic mechanisms for escaping immune surveillance, only a fraction of patients clinically respond to and benefit from cancer immunotherapy. Accumulating evidence derived from studies of drug repositioning, that is, the strategy to identify new uses for approved or investigational drugs that are outside the scope of the original medical indication, has suggested that some anthelmintic drugs, in addition to their antineoplastic effects, exert important immunomodulatory actions on specific subsets of immune cell and related pathways. In this review, we report and discuss current knowledge on the impact of anthelmintic drugs on host immunity and their potential implication in cancer immunotherapy.


Asunto(s)
Antihelmínticos , Antineoplásicos , Neoplasias , Humanos , Receptor de Muerte Celular Programada 1 , Neoplasias/tratamiento farmacológico , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antihelmínticos/farmacología , Antihelmínticos/uso terapéutico , Inmunoterapia , Antígeno B7-H1
7.
Int J Mol Sci ; 24(23)2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-38068921

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.


Asunto(s)
Antineoplásicos , Flavonoides , Neoplasias , Animales , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Flavonoides/farmacología , Flavonoides/uso terapéutico , Ligandos , Neoplasias/tratamiento farmacológico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
8.
Int J Mol Sci ; 22(8)2021 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-33920230

RESUMEN

In physiological conditions, the human intestinal mucosa is massively infiltrated with various subsets of immune cells, the activity of which is tightly regulated by several counter-regulatory factors. One of these factors is transforming growth factor-ß1 (TGF-ß1), a cytokine produced by multiple cell types and targeting virtually all the intestinal mucosal cells. Binding of TGF-ß1 to its receptors triggers Smad2/3 signaling, thus culminating in the attenuation/suppression of immune-inflammatory responses. In patients with Crohn's disease and patients with ulcerative colitis, the major human inflammatory bowel diseases (IBD), and in mice with IBD-like colitis, there is defective TGF-ß1/Smad signaling due to high levels of the intracellular inhibitor Smad7. Pharmacological inhibition of Smad7 restores TGF-ß1 function, thereby reducing inflammatory pathways in patients with IBD and colitic mice. On the other hand, transgenic over-expression of Smad7 in T cells exacerbates colitis in various mouse models of IBD. Smad7 is also over-expressed in other inflammatory disorders of the gut, such as refractory celiac disease, necrotizing enterocolitis and cytomegalovirus-induced colitis, even though evidence is still scarce and mainly descriptive. Furthermore, Smad7 has been involved in colon carcinogenesis through complex and heterogeneous mechanisms, and Smad7 polymorphisms could influence cancer prognosis. In this article, we review the data about the expression and role of Smad7 in intestinal inflammation and cancer.


Asunto(s)
Neoplasias del Colon/genética , Enfermedades Inflamatorias del Intestino/genética , Proteína smad7/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones , Oligonucleótidos Antisentido/uso terapéutico , Transducción de Señal/inmunología , Proteína smad7/antagonistas & inhibidores
9.
Clin Sci (Lond) ; 134(7): 907-920, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32236445

RESUMEN

BACKGROUND: Increased keratinocyte proliferation occurs in the skin of psoriatic patients and is supposed to play a role in the pathogenesis of this disorder. Compounds interfering with keratinocyte proliferation could be useful in the management of psoriatic patients. AIM: To investigate whether albendazole, an anti-helmintic drug that regulates epithelial cell function in various systems, inhibits keratinocyte proliferation in models of psoriasis. METHODS: Aldara-treated mice received daily topical application of albendazole. Keratinocyte proliferation and keratin (K) 6 and K16 expression were evaluated by immunohistochemistry and Western blotting and inflammatory cells/mediators were analysed by immunohistochemistry and real-time PCR. In human keratinocytes (HEKa and HaCaT) treated with albendazole, cell cycle and proliferation, keratins and cell cycle-associated factors were evaluated by flow cytometry, colorimetric assay and Western blotting respectively. RESULTS: Aldara-treated mice given albendazole exhibited reduced epidermal thickness, decreased number of proliferating keratinocytes and K6/K16 expression. Reduction of CD3- and Ly6G-positive cells in the skin of albendazole-treated mice associated with inhibition of IL-6, TNF-α, IL-1ß, IL-17A, IL-36, CCL17, CXCL1, CXCL2 and CXCL5 expression. Treatment of keratinocytes with albendazole reduced K6/K16 expression and reversibly inhibited cell growth by promoting accumulation of cells in S-phase. This phenomenon was accompanied by down-regulation of CDC25A, a phosphatase regulating progression of cell cycle through S-phase, and PKR-dependent hyper-phosphorylation of eIF2α, an inhibitor of CDC25 translation. In Aldara-treated mice, albendazole activated PKR, enhanced eIF2α phosphorylation and reduced CDC25A expression. CONCLUSIONS: Data show that albendazole inhibits keratinocyte proliferation and exerts therapeutic effect in a murine model of psoriasis.


Asunto(s)
Albendazol/farmacología , Proliferación Celular/efectos de los fármacos , Fármacos Dermatológicos/farmacología , Queratinocitos/efectos de los fármacos , Psoriasis/tratamiento farmacológico , Piel/efectos de los fármacos , Animales , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Imiquimod , Mediadores de Inflamación/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinas/genética , Queratinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Fosforilación , Psoriasis/inducido químicamente , Psoriasis/metabolismo , Psoriasis/patología , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Transducción de Señal , Piel/metabolismo , Piel/patología , Fosfatasas cdc25/metabolismo , eIF-2 Quinasa/metabolismo
10.
Int J Mol Sci ; 21(11)2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32532079

RESUMEN

Celiac disease (CD) is a chronic enteropathy that develops in genetically susceptible individuals after the ingestion of gluten. There has been a substantial increase in CD prevalence in the last 50 years, and it is now estimated that this disease affects approximately 1% of the population in the Western world. In the large majority of cases, CD is a benign disease, characterized by the complete resolution of symptoms and a normal life expectancy after the onset of a gluten-free diet (GFD). However, failure to adhere to a strict GFD bears the risk of adverse events and increases mortality. A considerable number of studies have considered the possible association between CD and neoplasms. In particular, an increased risk of malignancies, such as cancers of the gastrointestinal tract and intestinal lymphomas, has been reported. In this review, we summarize and discuss the current evidence on the possible association between CD and cancer.


Asunto(s)
Enfermedad Celíaca/complicaciones , Neoplasias/etiología , Enfermedad Celíaca/dietoterapia , Dieta Sin Gluten , Neoplasias Gastrointestinales/etiología , Humanos , Trastornos Linfoproliferativos/etiología
11.
Int J Mol Sci ; 21(14)2020 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-32668817

RESUMEN

Tumors of the digestive system, when combined together, account for more new cases and deaths per year than tumors arising in any other system of the body and their incidence continues to increase. Despite major efforts aimed at discovering and validating novel and effective drugs against these malignancies, the process of developing such drugs remains lengthy and costly, with high attrition rates. Drug repositioning (also known as drug repurposing), that is, the process of finding new uses for approved drugs, has been gaining popularity in oncological drug development as it provides the opportunity to expedite promising anti-cancer agents into clinical trials. Among the drugs considered for repurposing in oncology, compounds belonging to some classes of anthelmintics-a group of agents acting against infections caused by parasitic worms (helminths) that colonize the mammalian intestine-have shown pronounced anti-tumor activities and attracted particular attention due to their ability to target key oncogenic signal transduction pathways. In this review, we summarize and discuss the available experimental and clinical evidence about the use of anthelmintic drugs for the treatment of cancers of the digestive system.


Asunto(s)
Antihelmínticos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias del Sistema Digestivo/tratamiento farmacológico , Reposicionamiento de Medicamentos , Antihelmínticos/efectos adversos , Antihelmínticos/farmacología , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Bencimidazoles/efectos adversos , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Ensayos Clínicos como Asunto , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Salicilanilidas/efectos adversos , Salicilanilidas/farmacología , Salicilanilidas/uso terapéutico , Transducción de Señal/efectos de los fármacos
12.
Int J Mol Sci ; 21(21)2020 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-33143318

RESUMEN

The human virome, which is a collection of all the viruses that are present in the human body, is increasingly being recognized as an essential part of the human microbiota. The human gastrointestinal tract and related organs (e.g., liver, pancreas, and gallbladder)-composing the gastrointestinal (or digestive) system-contain a huge number of viral particles which contribute to maintaining tissue homeostasis and keeping our body healthy. However, perturbations of the virome steady-state may, both directly and indirectly, ignite/sustain oncogenic mechanisms contributing to the initiation of a dysplastic process and/or cancer progression. In this review, we summarize and discuss the available evidence on the association and role of viruses in the development of cancers of the digestive system.


Asunto(s)
Efecto Espectador , Neoplasias Gastrointestinales/patología , Virosis/complicaciones , Virus/patogenicidad , Animales , Neoplasias Gastrointestinales/virología , Humanos , Virosis/virología
13.
Clin Sci (Lond) ; 132(11): 1155-1168, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29540537

RESUMEN

Metformin, a hypoglycemic drug used for treatment of type 2 diabetes, regulates inflammatory pathways. By using several models of intestinal inflammation, we examined whether metformin exerts anti-inflammatory effects and investigated the basic mechanism by which metformin blocks pathologic signals. Colitic mice given metformin exhibited less colonic inflammation and increased expression of active AMP-activated protein kinase, a mediator of the metabolic effects of metformin, in both epithelial and lamina propria compartments. Pharmacological inhibition of AMP-activated protein kinase reduced but did not prevent metformin-induced therapeutic effect as well as treatment of colitic mice with a pharmacological activator of AMP-activated protein kinase attenuated but did not resolve colitis. These data suggest that the anti-inflammatory effect of metformin relies on the control of additional pathways other than AMP-activated protein kinase. Indeed, metformin down-regulated p38 MAP kinase activation in colitic mice through an AMP-activated protein kinase-independent mechanism. Expression of active form of AMP-activated protein kinase was reduced in inflammatory bowel disease patients and treatment of mucosal cells of such patients with metformin enhanced AMP-activated protein kinase activation and reduced p38 MAP kinase activation, thereby inhibiting interleukin-6 expression. Our findings indicate that metformin is a good candidate for inhibiting pathological inflammation in the gut.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Colitis Ulcerosa/enzimología , Hipoglucemiantes/farmacología , Metformina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Colitis Ulcerosa/tratamiento farmacológico , Colon/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Activación Enzimática/efectos de los fármacos , Femenino , Hipoglucemiantes/uso terapéutico , Mediadores de Inflamación/metabolismo , Interleucina-6/biosíntesis , Mucosa Intestinal/enzimología , Metformina/uso terapéutico , Ratones Endogámicos BALB C , Fosforilación/efectos de los fármacos , Receptor de Insulina/metabolismo
14.
Int J Mol Sci ; 19(6)2018 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-29914167

RESUMEN

Signal transducers and activators of transcription (STATs) mediate essential signaling pathways in different biological processes, including immune responses, hematopoiesis, and neurogenesis. Among the STAT members, STAT3 plays crucial roles in cell proliferation, survival, and differentiation. While STAT3 activation is transient in physiological conditions, STAT3 becomes persistently activated in a high percentage of solid and hematopoietic malignancies (e.g., melanoma, multiple myeloma, breast, prostate, ovarian, and colon cancers), thus contributing to malignant transformation and progression. This makes STAT3 an attractive therapeutic target for cancers. Initial strategies aimed at inhibiting STAT3 functions have focused on blocking the action of its activating kinases or sequestering its DNA binding ability. More recently, the diffusion of proteomic-based techniques, which have allowed for the identification and characterization of novel STAT3-interacting proteins able to modulate STAT3 activity via its subcellular localization, interact with upstream kinases, and recruit transcriptional machinery, has raised the possibility to target such cofactors to specifically restrain STAT3 oncogenic functions. In this article, we summarize the available data about the function of STAT3 interactors in malignant cells and discuss their role as potential therapeutic targets for cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Factor de Transcripción STAT3/metabolismo , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Transcripción STAT3/genética
15.
Carcinogenesis ; 35(7): 1536-46, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24480808

RESUMEN

Transforming growth factor-beta (TGF-ß) is deeply involved in colorectal cancer development and the disruption of the TGF-ß signaling in dysplastic cells is required for tumor to grow. Nevertheless, tumor cells express TGF-ß to escape the immune surveillance mediated by T cells. T-cell expression of Smad7, an intracellular inhibitor of the TGF-ß signaling, protects against colitis-associated colorectal cancer. However, whether Smad7 in T cells might influence colorectal cancer growth independently of chronic inflammation and which T-cell subset is involved in this process is unknown. To address this issue, T-cell-specific Smad7 transgenic mice and wild-type (WT) littermates were subcutaneously transplanted with syngenic MC38 colon carcinoma cells. Smad7Tg mice were resistant to tumor development compared with WT mice and protection was dependent on CD4(+) T cells. Smad7 expression in T cells increased the number of tumor-infiltrating Tbet/ROR-γ-t double-positive CD4 T cells characterized by the expression of tumor necrosis factor-alpha (TNF-α) and interferon-gamma but lower IL17A. The low expression of IL17A caused by the Smad7 expression in tumor-infiltrating CD4(+) T cells enabled the TNF-α-mediated killing of cancer cells both in vitro and in vivo, thus indicating that the Smad7-mediated plastic effect on T-cell phenotype induces protection against colorectal cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/patología , Interleucina-17/fisiología , Linfocitos Infiltrantes de Tumor/inmunología , Proteína smad7/fisiología , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Interferón gamma/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Th17/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
16.
J Crohns Colitis ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38953702

RESUMEN

BACKGROUND AND AIM: Bromodomain-containing protein 4 (BRD4), one of the components of the bromodomain and extraterminal domain (BET) family, is a transcriptional and epigenetic regulator of cellular proliferation and cytokine production. In this study, we assessed whether BRD4 regulates the cytokine response in inflammatory bowel diseases (IBD). MATERIALS AND METHODS: BRD4 expression was analyzed in intestinal mucosal samples of patients with ulcerative colitis (UC), patients with Crohn's disease (CD), normal controls (CTRs), and mice with chemically-induced colitis by real-time PCR, Western blotting, and confocal microscopy. Cytokine production was evaluated in lamina propria mononuclear cells (LPMCs) of IBD patients and mucosal tissues of colitic mice treated with BRD4 inhibitors. Finally, we evaluated the effect of JQ1, an inhibitor of the BRD4 signaling pathway, on the course of murine colitis. RESULTS: BRD4 RNA and protein expression was up-regulated in the inflamed mucosa of patients with UC and patients with CD as compared to the uninvolved areas of the same patients and CTRs, and in the inflamed colon of colitic mice. Knockdown of BRD4 with a specific antisense oligonucleotide in IBD LPMCs led to reduced expression of TNF-α, IL-6, IFN-γ, and IL-17A. Administration of JQ1 to colitic mice inhibited the inflammatory cytokine response and attenuated the ongoing colitis. CONCLUSIONS: This is the first study showing the up-regulation of BRD4 in IBD and suggesting the role of such a protein in the positive control of the inflammatory cytokine response in the gut.

17.
J Immunol ; 186(9): 5435-42, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21441456

RESUMEN

Psoriasis is a chronic inflammatory disorder of the skin characterized by epidermal hyperplasia and infiltration of leukocytes into the dermis and epidermis. T cell-derived cytokines, such as IFN-γ and IL-17A, play a major role in the psoriasis-associated epidermal hyperplasia, even though factors/mechanisms that regulate the production of these cytokines are not fully understood. We have recently shown that IL-21 is synthesized in excess in psoriatic skin lesions and causes epidermal hyperplasia when injected intradermally in mice. Moreover, in the human psoriasis SCID mouse model, neutralization of IL-21 reduces both skin thickening and expression of inflammatory molecules, thus supporting the pathogenic role of IL-21 in psoriasis. However, the basic mechanism by which IL-21 promotes skin pathology remains unknown. In this study, we show that CD4(+) cells accumulate early in the dermis of IL-21-treated mice and mediate the development of epidermal hyperplasia. Indeed, IL-21 fails to induce skin damage in RAG1-deficient mice and CD4(+) cell-depleted wild-type mice. The majority of CD4(+) cells infiltrating the dermis of IL-21-treated mice express IFN-γ and, to a lesser extent, IL-17A. Studies in cytokine knockout mice show that IFN-γ, but not IL-17A, is necessary for IL-21-induced epidermal hyperplasia. Finally, we demonstrate that IFN-γ-producing CD4(+) cells infiltrating the human psoriatic plaque express IL-21R, and abrogation of IL-21 signals reduces IFN-γ expression in cultures of psoriatic CD4(+) cells. Data indicate that IL-21 induces an IFN-γ-dependent pathogenic response in vivo, thus contributing to elucidate a mechanism by which IL-21 sustains skin-damaging inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interferón gamma/biosíntesis , Interleucinas/inmunología , Psoriasis/inmunología , Piel/patología , Animales , Linfocitos T CD4-Positivos/metabolismo , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Hiperplasia/patología , Interferón gamma/inmunología , Interleucinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Psoriasis/metabolismo , Psoriasis/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/inmunología , Piel/metabolismo
18.
Int J Mol Sci ; 14(9): 17972-85, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-24005861

RESUMEN

Colorectal cancer (CRC) is the third most common malignant neoplasm worldwide. Although conclusive evidence is still lacking, epidemiologic studies suggest that long-term use of non-steroidal anti-inflammatory drugs (NSAIDs) has chemopreventive properties against CRC. Similarly, regular consumption of mesalazine, a drug structurally related to NSAIDs, seems to reduce the risk of CRC in patients with ulcerative colitis. These observations are supported by a large body of experimental data showing the ability of such drugs to inhibit multiple pathways that sustain colon carcinogenesis. This review summarizes the current information on the molecular mechanisms by which NSAIDs and mesalazine could interfere with CRC cell growth and survival.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Quimioprevención/métodos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/prevención & control , Mesalamina/uso terapéutico , Animales , Humanos
19.
Int J Mol Sci ; 14(12): 23774-90, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24317436

RESUMEN

Smad7 was initially identified as an inhibitor of Transforming growth factor (TGF)-ß due mainly to its ability to bind TGF-ß receptor type I and prevent TGF-ß-associated Smad signaling. More recently, it has been demonstrated that Smad7 can interact with other intracellular proteins and regulate also TGF-ß-independent signaling pathways thus making a valid contribution to the neoplastic processes in various organs. In particular, data emerging from experimental studies indicate that Smad7 may differently modulate the course of various tumors depending on the context analyzed. These observations, together with the demonstration that Smad7 expression is deregulated in many cancers, suggest that therapeutic interventions around Smad7 can help interfere with the development/progression of human cancers. In this article we review and discuss the available data supporting the role of Smad7 in the modulation of cancer growth and progression.


Asunto(s)
Neoplasias/metabolismo , Proteína smad7/metabolismo , Humanos , Metástasis de la Neoplasia , Neoplasias/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
20.
Curr Res Immunol ; 4: 100055, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36714553

RESUMEN

In physiological conditions, the human gut contains more immune cells than the rest of the body, but no overt tissue damage occurs, because several regulatory mechanisms control the activity of such cells thus preventing excessive and detrimental responses. One such mechanism relies on the action of transforming growth factor (TGF)-ß1, a cytokine that targets both epithelial cells and many immune cell types. Loss of TGF-ß1 function leads to intestinal pathology in both mice and humans. For instance, disruption of TGF-ß1 signaling characterizes the destructive immune-inflammatory response in patients with Crohn's disease and patients with ulcerative colitis, the major human inflammatory bowel disease (IBD) entities. In these pathologies, the defective TGF-ß1-mediated anti-inflammatory response is associated with elevated intestinal levels of Smad7, an antagonist of TGF-ß1 signaling. Consistently, knockdown of Smad7 restores TGF-ß1 function thereby attenuating intestinal inflammation in patients with IBD as well as in mice with IBD-like colitis. Up-regulation of Smad7 and reduced TGF-ß1 signaling occurs also in necrotizing enterocolitis, environmental enteropathy, refractory celiac disease, and cytomegalovirus-induced colitis. In this article, we review the available data supporting the pathogenic role of Smad7 in the gastrointestinal tract and discuss whether and how targeting Smad7 can help attenuate detrimental immuno-inflammatory responses in the gut.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA