Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cell Rep ; 42(5): 112475, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37167967

RESUMEN

Immunosuppressive tumor microenvironments (TMEs) reduce the effectiveness of immune responses in cancer. Mesenchymal stromal cells (MSCs), precursors to cancer-associated fibroblasts (CAFs), promote tumor progression by enhancing immune cell suppression in colorectal cancer (CRC). Hyper-sialylation of glycans promotes immune evasion in cancer through binding of sialic acids to their receptors, Siglecs, expressed on immune cells, which results in inhibition of effector functions. The role of sialylation in shaping MSC/CAF immunosuppression in the TME is not well characterized. In this study, we show that tumor-conditioned stromal cells have increased sialyltransferase expression, α2,3/6-linked sialic acid, and Siglec ligands. Tumor-conditioned stromal cells and CAFs induce exhausted immunomodulatory CD8+ PD1+ and CD8+ Siglec-7+/Siglec-9+ T cell phenotypes. In vivo, targeting stromal cell sialylation reverses stromal cell-mediated immunosuppression, as shown by infiltration of CD25 and granzyme B-expressing CD8+ T cells in the tumor and draining lymph node. Targeting stromal cell sialylation may overcome immunosuppression in the CRC TME.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Humanos , Linfocitos T CD8-positivos , Microambiente Tumoral , Terapia de Inmunosupresión , Células del Estroma/metabolismo , Neoplasias/patología , Fibroblastos Asociados al Cáncer/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo
2.
BMC Gastroenterol ; 22(1): 219, 2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35508989

RESUMEN

Gastroesophageal reflux disease (GERD) is one the most common medical complaints in pregnant women. Some women continue to experience GERD symptoms after delivery. Effective management of GERD symptoms is important to improve productivity and quality of life. Management of heartburn in pregnant and breastfeeding women involves lifestyle modifications, dietary modifications, non-pharmaceutical remedies and pharmaceutical drugs. For most patients, lifestyle/dietary modifications are helpful in reducing GERD symptoms. For patients who require a more intense intervention, various types of pharmaceutical drugs are available. However, the suitability of each treatment for use during pregnancy and lactation must be taken into consideration. This article explores the reported efficacy and safety of these treatment options in pregnant and breastfeeding women. Recommended treatment algorithm in pregnant and breastfeeding women have been developed.


Asunto(s)
Reflujo Gastroesofágico , Complicaciones del Embarazo , Lactancia Materna , Femenino , Reflujo Gastroesofágico/tratamiento farmacológico , Pirosis/tratamiento farmacológico , Pirosis/terapia , Humanos , Preparaciones Farmacéuticas , Embarazo , Complicaciones del Embarazo/terapia , Calidad de Vida
3.
Biomaterials ; 281: 121364, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35066288

RESUMEN

Delivering drugs directly to the inflamed intestinal sites to treat inflammatory bowel disease (IBD), particularly Crohn's and ulcerative colitis, is highly challenging. Recent advances in colitis therapy medications are expanding opportunities for improving local on-site drug availability by minimising the associated systemic side-effects. Drug delivery with targeted carrier systems has shown the potential to increase site-specificity, stability, and therapeutic efficacy. Herein, we report the development of a strong anionic charged inflammation targeted nanocarriers (IT-NCs) loaded with an immunosuppressant model drug. This system showed preferential adhesion on a charge-modified surface in vitro, and in both dextran sulfate sodium (DSS) and TNBS colitis mice in vivo models. IT-NCs showed improved colitis phenotype therapeutic efficacy in both animal models compared to free drug. Furthermore, ex vivo study of colon tissue biopsies from patients with colitis revealed that IT-NCs adhered preferentially to inflamed biopsies compared to normal. Together, our results suggest that IT-NCs have promising therapeutic potential as delivery carriers' in colitis management.


Asunto(s)
Colitis Ulcerosa , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Portadores de Fármacos/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal , Ratones
4.
Frontline Gastroenterol ; 11(6): 441-447, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33104766

RESUMEN

INTRODUCTION: Accelerated dose infliximab (IFX) induction is associated with reduced short-term colectomy rate in acute severe ulcerative colitis (ASUC). Data on medium/long-term outcomes of this strategy are limited. AIMS: Evaluate medium/long-term outcomes in patients receiving IFX induction for ASUC, comparing accelerated dose (AD) and standard dose (SD) induction. METHODS: Retrospective study of consecutive patients admitted with corticosteroid-refractory ASUC in four tertiary referral centres within INITIative IBD research network (www.initiativeibd.ie). IFX rescue was given either as SD (weeks 0, 2, 6) or AD (<28 days) from January 2010 to September 2017. AD induction has been utilised in participating centres since 2014. Consequently SD patients were subdivided based on time period of IFX rescue: historical SD group (SD1) (2010-2013) and current SD group (SD2) (2014-2017). Primary endpoint was time to colectomy; secondary endpoint was time to IFX discontinuation if induction was complete. RESULTS: 145 patients received rescue IFX (AD=58, SD1=32, SD2=55). Disease severity at induction was comparable between AD and SD1 groups; however, SD2 group had less severe disease: median C-reactive protein (CRP) 39, 44 and 20 mg/L for AD, SD1 and SD2 groups, respectively (p=0.026, Kruskal-Wallis); median CRP: albumin ratio was 1.4, 1.8 and 0.6 (p=0.016). Median follow-up for AD, SD1 and SD2 groups was 1.6 (IQR 1.1-3.1), 4.9 (IQR 2.6-5.5) and 1.5 (IQR 0.9-2.3) years. Time to colectomy was shorter in SD1 (log rank p=0.0013); no significant difference in time to colectomy was observed comparing AD and SD2 groups (log rank p=0.32). 123 patients (84%) completed IFX induction and received maintenance therapy. Time to IFX discontinuation was shorter in SD1 (log rank p=0.009). CONCLUSION: Time to colectomy is significantly prolonged with use of AD IFX in selected ASUC patients with more severe disease. Historical use of standard IFX induction for all ASUC patients is associated with inferior long-term outcomes.

5.
Proc Natl Acad Sci U S A ; 117(30): 17808-17819, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32661168

RESUMEN

p53 is the most frequently mutated, well-studied tumor-suppressor gene, yet the molecular basis of the switch from p53-induced cell-cycle arrest to apoptosis remains poorly understood. Using a combination of transcriptomics and functional genomics, we unexpectedly identified a nodal role for the caspase-8 paralog and only human pseudo-caspase, FLIP(L), in regulating this switch. Moreover, we identify FLIP(L) as a direct p53 transcriptional target gene that is rapidly up-regulated in response to Nutlin-3A, an MDM2 inhibitor that potently activates p53. Genetically or pharmacologically inhibiting expression of FLIP(L) using siRNA or entinostat (a clinically relevant class-I HDAC inhibitor) efficiently promoted apoptosis in colorectal cancer cells in response to Nutlin-3A, which otherwise predominantly induced cell-cycle arrest. Enhanced apoptosis was also observed when entinostat was combined with clinically relevant, p53-activating chemotherapy in vitro, and this translated into enhanced in vivo efficacy. Mechanistically, FLIP(L) inhibited p53-induced apoptosis by blocking activation of caspase-8 by the TRAIL-R2/DR5 death receptor; notably, this activation was not dependent on receptor engagement by its ligand, TRAIL. In the absence of caspase-8, another of its paralogs, caspase-10 (also transcriptionally up-regulated by p53), induced apoptosis in Nutlin-3A-treated, FLIP(L)-depleted cells, albeit to a lesser extent than in caspase-8-proficient cells. FLIP(L) depletion also modulated transcription of canonical p53 target genes, suppressing p53-induced expression of the cell-cycle regulator p21 and enhancing p53-induced up-regulation of proapoptotic PUMA. Thus, even in the absence of caspase-8/10, FLIP(L) silencing promoted p53-induced apoptosis by enhancing PUMA expression. Thus, we report unexpected, therapeutically relevant roles for FLIP(L) in determining cell fate following p53 activation.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzamidas/farmacología , Caspasa 8/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Sinergismo Farmacológico , Regulación de la Expresión Génica , Humanos , Imidazoles/metabolismo , Modelos Biológicos , Piperazinas/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Piridinas/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteína p53 Supresora de Tumor/genética
6.
Immunopharmacol Immunotoxicol ; 41(5): 527-537, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31505962

RESUMEN

Background: Pattern recognition receptors form an essential part of the host defenses against pathogens, in particular in the intestinal epithelium. However, despite their importance relatively little is understood about the regulation of their expression. Increasing evidence suggesting that epigenetic mechanisms such as DNA methylation and histone acetylation have substantial effects on gene expression and regulation. Epigenetic modifying drugs are now used to treat certain cancers but not a lot is known about their effects on the innate immune system. Thus, we set out to examine the role of such drugs in the expression and function of Toll-like receptors. Methods: Using the HCT116 epithelial cell line, we determined the effects of genetic knockout of the DNA methyltransferases enzymes (DNMTs), as well as pharmacological inhibition of the DNMTs and histone deacetylase complexes (HDACs) on TLR responses to their ligands. Results: Our initial results showed that anti-viral responses were affected by changes in the epigenome, with TLR3 responses showing the most dramatic differences. We determined that inhibition of methylation and acetylation inhibited poly I:C induced increases in signaling protein phosphorylation, as well as increases in cytokine mRNA expression and release. We also observed that treatment with epigenetic modifying drugs were leading to large increases in IRF8 expression, a protein that is a known negative regulator of TLR3. When we overexpressed IRF8 in our WT cells we noticed inhibition of poly I:C responses. Conclusion: This research highlighted the potential immunoregulatory role of epigenetic modifying drugs specifically in response to viral stimulation.


Asunto(s)
Antivirales/farmacología , Epigénesis Genética/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Poli I-C/farmacología , Receptor Toll-Like 3/metabolismo , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Células Epiteliales/virología , Técnicas de Silenciamiento del Gen , Células HCT116 , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/virología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Receptor Toll-Like 3/genética , ADN Metiltransferasa 3B
7.
Cancer Immunol Res ; 6(11): 1426-1441, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30228206

RESUMEN

Stromal cells of mesenchymal origin reside below the epithelial compartment and provide structural support in the intestine. These intestinal stromal cells interact with both the epithelial cell compartments, as well as infiltrating hematopoietic immune cells. The importance of these cells in regulating immune homeostasis during inflammation is well recognized. However, little is known about their function and phenotype in the inflammatory tumor microenvironment. Using a syngeneic, immunogenic model of colorectal cancer, we showed that TNFα-initiated inflammatory signaling in CT26 colorectal cancer cells selectively induced PD-L1 expression in stromal cells. Using CD274 shRNA and antibody-mediated approaches, we showed that stromal cell PD-L1 potentiated enhanced immunosuppression, characterized by inhibition of activated CD8+ granzyme B-secreting T cells in vitro, and the inhibition of CD8+ effector cells was associated with enhanced tumor progression. Stromal cell immunosuppressive and tumor-promoting effects could be reversed with administration of anti-PD-1 in vivo We validated our findings of stromal cell CD274 expression in two cohorts of clinical samples and also observed PD-L1 induction on human stromal cells in response to exposure to the inflammatory secretome from human colon cancer cells, irrespective of microsatellite instability. Collectively, our data showed that tumor-associated stromal cells support T-cell suppression by PD-L1 induction, which is dependent on colon cancer inflammatory signaling. Our findings reveal a key role of mesenchymal stromal cells PD-L1 in suppression of CD8+ antitumor immune responses and potentiation of colorectal cancer progression. Cancer Immunol Res; 6(11); 1426-41. ©2018 AACR.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/inmunología , Células del Estroma/inmunología , Animales , Antígeno B7-H1/genética , Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Humanos , Ratones Endogámicos BALB C , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Epigenetics ; 12(8): 591-606, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28557546

RESUMEN

Aberrant DNA methylation patterns have been reported in inflamed tissues and may play a role in disease. We studied DNA methylation and gene expression profiles of purified intestinal epithelial cells from ulcerative colitis patients, comparing inflamed and non-inflamed areas of the colon. We identified 577 differentially methylated sites (false discovery rate <0.2) mapping to 210 genes. From gene expression data from the same epithelial cells, we identified 62 differentially expressed genes with increased expression in the presence of inflammation at prostate cancer susceptibility genes PRAC1 and PRAC2. Four genes showed inverse correlation between methylation and gene expression; ROR1, GXYLT2, FOXA2, and, notably, RARB, a gene previously identified as a tumor suppressor in colorectal adenocarcinoma as well as breast, lung and prostate cancer. We highlight targeted and specific patterns of DNA methylation and gene expression in epithelial cells from inflamed colon, while challenging the importance of epithelial cells in the pathogenesis of chronic inflammation.


Asunto(s)
Colitis Ulcerosa/genética , Metilación de ADN , Mucosa Intestinal/metabolismo , Adulto , Colitis Ulcerosa/metabolismo , Femenino , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Transcriptoma
10.
Inflamm Bowel Dis ; 22(12): 2863-2878, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27824651

RESUMEN

BACKGROUND: Infliximab (IFX), an anti-tumour necrosis factor alpha (TNFα) monoclonal antibody, provides clinical benefits in treating Crohn's disease (CD) but its mechanisms of action are not fully elucidated. This study investigated blood monocyte repertoires and the acute effects of IFX infusion on monocyte subset phenotype and function in IFX-treated patients with CD. METHODS: Monocytes and monocyte subsets were enumerated and phenotypically characterized by multicolor flow cytometry in freshly isolated blood from healthy controls (n = 21) and patients with CD treated with (IFX, n = 24) and without (non-IFX, n = 20) IFX. For the IFX-CD group, blood was sampled immediately before (tough-IFX) and after (peak-IFX) infusion. Monocyte responses to lipopolysaccharide were analyzed by whole-blood intracellular cytokine staining. RESULTS: Non-IFX and IFX-CD patients had increased numbers of intermediate (CD14CD16) monocytes compared with healthy controls, whereas classical (CD14CD16) and nonclassical (CD14CD16) monocytes were numerically reduced in the IFX-CD group alone. In all groups, monocyte subsets expressed high surface levels of transmembrane (tm)TNFα. After IFX infusion, a significant reduction in monocyte numbers occurred. Post-IFX monocytopenia was proportionately greatest for classical and intermediate subsets, correlated with postinfusion IFX levels and was not associated with monocyte apoptosis. In contrast, lipopolysaccharide-induced production of TNFα and IL-12 by monocytes was significantly reduced in peak-IFX compared with trough-IFX blood samples. CONCLUSIONS: Actively managed CD is associated with monocyte repertoire skewing suggestive of chronic inflammatory stimulation. Infused IFX acutely targets monocytes, likely by binding to tmTNFα, resulting in a non-apoptosis-related decline in circulating monocyte numbers and blunting of the inflammatory response of monocytes remaining in the blood.


Asunto(s)
Enfermedad de Crohn/tratamiento farmacológico , Fármacos Gastrointestinales/farmacología , Infliximab/farmacología , Monocitos/efectos de los fármacos , Adulto , Anciano , Estudios de Casos y Controles , Enfermedad de Crohn/sangre , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Monocitos/citología , Adulto Joven
11.
Oncotarget ; 7(37): 60752-60774, 2016 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-27542276

RESUMEN

The tumour microenvironment (TME) is an important factor in determining the growth and metastasis of colorectal cancer, and can aid tumours by both establishing an immunosuppressive milieu, allowing the tumour avoid immune clearance, and by hampering the efficacy of various therapeutic regimens. The tumour microenvironment is composed of many cell types including tumour, stromal, endothelial and immune cell populations. It is widely accepted that cells present in the TME acquire distinct functional phenotypes that promote tumorigenesis. One such cell type is the mesenchymal stromal cell (MSC). Evidence suggests that MSCs exert effects in the colorectal tumour microenvironment including the promotion of angiogenesis, invasion and metastasis. MSCs immunomodulatory capacity may represent another largely unexplored central feature of MSCs tumour promoting capacity. There is considerable evidence to suggest that MSCs and their secreted factors can influence the innate and adaptive immune responses. MSC-immune cell interactions can skew the proliferation and functional activity of T-cells, dendritic cells, natural killer cells and macrophages, which could favour tumour growth and enable tumours to evade immune cell clearance. A better understanding of the interactions between the malignant cancer cell and stromal components of the TME is key to the development of more specific and efficacious therapies for colorectal cancer. Here, we review and explore MSC- mediated mechanisms of suppressing anti-tumour immune responses in the colon tumour microenvironment. Elucidation of the precise mechanism of immunomodulation exerted by tumour-educated MSCs is critical to inhibiting immunosuppression and immune evasion established by the TME, thus providing an opportunity for targeted and efficacious immunotherapy for colorectal cancer growth and metastasis.


Asunto(s)
Colon/inmunología , Neoplasias Colorrectales/inmunología , Células Madre Mesenquimatosas/inmunología , Animales , Antígenos de Neoplasias/inmunología , Humanos , Inmunidad , Terapia de Inmunosupresión , Escape del Tumor , Microambiente Tumoral
12.
Physiol Genomics ; 48(4): 257-73, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26812987

RESUMEN

Region and cell-type specific differences in the molecular make up of colon epithelial cells have been reported. Those differences may underlie the region-specific characteristics of common colon epithelial diseases such as colorectal cancer and inflammatory bowel disease. DNA methylation is a cell-type specific epigenetic mark, essential for transcriptional regulation, silencing of repetitive DNA and genomic imprinting. Little is known about any region-specific variations in methylation patterns in human colon epithelial cells. Using purified epithelial cells and whole biopsies (n= 19) from human subjects, we generated epigenome-wide DNA methylation data (using the HELP-tagging assay), comparing the methylation signatures of the proximal and distal colon. We identified a total of 125 differentially methylated sites (DMS) mapping to transcription start sites of protein-coding genes, most notably several members of the homeobox (HOX) family of genes. Patterns of differential methylation were validated with MassArray EpiTYPER. We also examined DNA methylation in whole biopsies, applying a computational technique to deconvolve variation in methylation within cell types and variation in cell-type composition across biopsies. Including inferred epithelial proportions as a covariate in differential methylation analysis applied to the whole biopsies resulted in greater overlap with the results obtained from purified epithelial cells compared with when the covariate was not included. Results obtained from both approaches highlight region-specific methylation patterns of HOX genes in colonic epithelium. Regional variation in methylation patterns has implications for the study of diseases that exhibit regional expression patterns in the human colon, such as inflammatory bowel disease and colorectal cancer.


Asunto(s)
Colon/citología , Metilación de ADN , Genes Homeobox , Biopsia , Colon/fisiología , Epigénesis Genética , Células Epiteliales/fisiología , Humanos , Reproducibilidad de los Resultados
13.
Inflamm Bowel Dis ; 21(12): 2909-20, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26348448

RESUMEN

Tumor necrosis factor alpha (TNF-α) has been widely accepted as a therapeutic target for inflammatory disorders including inflammatory bowel disease. Anti-TNF-α monoclonal antibodies (mAbs) including infliximab, adalimumab, golimumab, and certolizumab pegol have revolutionized therapy for these chronic inflammatory disorders. These agents are potent inhibitors of TNF-α, but significant evidence points to the fact that their actions extend beyond simple neutralization of the cytokine. Recent advances in understanding the mechanism of action of anti-TNF-α mAbs has discovered a number of previously unrecognized actions that are likely to be relevant in mediating their anti-inflammatory effects. Many of those actions are mediated by the binding of the antibodies to transmembrane TNF-α (tmTNF-α) and involve complex interactions with other molecular factors and cells. In this review, we have highlighted new information on the mechanism of actions of anti-TNF-α mAbs, from in vitro and in vivo studies. Despite obvious benefits in many patients, the clinical use of these antibodies are hampered by the fact that some patients do not respond to them, and among patients who do respond, many will develop recurrent disease despite continued dosing. Although pharmacokinetic factors explain some of the observed cases of partial or complete resistance to the effects of anti-TNF-α mAbs, other nonresponder patients may be resistant to those agents mechanism of action. A more thorough understanding of the mechanism of action of anti-TNF-α mAbs may allow the development of strategies to individualize therapy and to overcome resistance.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adalimumab/farmacología , Anticuerpos Monoclonales/farmacología , Certolizumab Pegol/farmacología , Resistencia a Medicamentos/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Infliximab/farmacología
14.
Curr Opin Gastroenterol ; 31(2): 124-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25603403

RESUMEN

PURPOSE OF REVIEW: To discuss the recent landmark findings that have increased our understanding not only of the role of the epithelial cell cycle in the homeostasis of the small intestine, but also its relevance to inflammation and cancer. RECENT FINDINGS: Recent data have unveiled novel information on protein interactions directly involved in the cell cycle as well as in the pathways that transduce external environmental signals to the cell cycle. A growing body of the recent evidence confirms the importance of food as well as hormonal regulation in the gut on cell cycle. Information on the contribution of the epithelial microenvironment, including the microbiota, has grown substantially in the recent years as well as on the gene-environment interactions and the multiple epigenetic mechanisms involved in regulating cell-cycle proteins and signalling. Finally, further studies investigating the dysregulation of the cell cycle during inflammation and proliferation have increased our understanding of the pathophysiology of chronic inflammatory diseases and cancer. SUMMARY: This review highlights some of the most recent advances that further emphasize the importance of the cell cycle in the small intestine during homeostasis as well as in inflammation and cancer.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Células Epiteliales/metabolismo , Homeostasis/inmunología , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Ciclo Celular , Metilación de ADN , Humanos , Inflamación/patología , Enfermedades Inflamatorias del Intestino/patología , ARN Mensajero
15.
Gut ; 63(2): e1, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24381262
16.
World J Gastroenterol ; 19(2): 185-98, 2013 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-23345941

RESUMEN

Ionising radiation therapy is a common treatment modality for different types of cancer and its use is expected to increase with advances in screening and early detection of cancer. Radiation injury to the gastrointestinal tract is important factor working against better utility of this important therapeutic modality. Cancer survivors can suffer a wide variety of acute and chronic symptoms following radiotherapy, which significantly reduces their quality of life as well as adding an extra burden to the cost of health care. The accurate diagnosis and treatment of intestinal radiation injury often represents a clinical challenge to practicing physicians in both gastroenterology and oncology. Despite the growing recognition of the problem and some advances in understanding the cellular and molecular mechanisms of radiation injury, relatively little is known about the pathophysiology of gastrointestinal radiation injury or any possible susceptibility factors that could aggravate its severity. The aims of this review are to examine the various clinical manifestations of post-radiation gastrointestinal symptoms, to discuss possible patient and treatment factors implicated in normal gastrointestinal tissue radiosensitivity and to outline different mechanisms of intestinal tissue injury.


Asunto(s)
Tracto Gastrointestinal/lesiones , Traumatismos por Radiación/etiología , Radioterapia/efectos adversos , Tracto Gastrointestinal/efectos de la radiación , Humanos , Neoplasias/radioterapia , Traumatismos por Radiación/epidemiología , Dosificación Radioterapéutica , Factores de Riesgo
17.
World J Gastroenterol ; 19(2): 199-208, 2013 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-23345942

RESUMEN

With the recent advances in detection and treatment of cancer, there is an increasing emphasis on the efficacy and safety aspects of cancer therapy. Radiation therapy is a common treatment for a wide variety of cancers, either alone or in combination with other treatments. Ionising radiation injury to the gastrointestinal tract is a frequent side effect of radiation therapy and a considerable proportion of patients suffer acute or chronic gastrointestinal symptoms as a result. These side effects often cause morbidity and may in some cases lower the efficacy of radiotherapy treatment. Radiation injury to the gastrointestinal tract can be minimised by either of two strategies: technical strategies which aim to physically shift radiation dose away from the normal intestinal tissues, and biological strategies which aim to modulate the normal tissue response to ionising radiation or to increase its resistance to it. Although considerable improvement in the safety of radiotherapy treatment has been achieved through the use of modern optimised planning and delivery techniques, biological techniques may offer additional further promise. Different agents have been used to prevent or minimize the severity of gastrointestinal injury induced by ionising radiation exposure, including biological, chemical and pharmacological agents. In this review we aim to discuss various technical strategies to prevent gastrointestinal injury during cancer radiotherapy, examine the different therapeutic options for acute and chronic gastrointestinal radiation injury and outline some examples of research directions and considerations for prevention at a pre-clinical level.


Asunto(s)
Tracto Gastrointestinal/lesiones , Traumatismos por Radiación/prevención & control , Traumatismos por Radiación/terapia , Radioterapia/efectos adversos , Antioxidantes/uso terapéutico , Endoscopía Gastrointestinal/métodos , Tracto Gastrointestinal/efectos de la radiación , Humanos , Neoplasias/radioterapia , Apoyo Nutricional , Traumatismos por Radiación/etiología , Dosificación Radioterapéutica
18.
Carcinogenesis ; 33(4): 723-31, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22235026

RESUMEN

Inflammation can result from a range of sources including microbial infections, exposure to allergens and toxic chemicals, autoimmune disease and obesity. A well-balanced immune response can be anti-tumorigenic; however, a sustained or chronic inflammatory response is generally harmful as the immune response becomes distorted. A causal link between chronic inflammation and cancer is now well accepted and many chronically inflamed organs of the gastrointestinal tract show this association. For example, patients with inflammatory bowel disease (IBD), including both ulcerative colitis and Crohn's disease, have a 2- to 3-fold greater lifetime risk of developing colorectal cancer compared with the general population. The development of colitis-associated cancer (CAC) is thought to be multifaceted and is probably due to a combination of genetic factors, epigenetic factors and the duration, extent and severity of disease. Recently, epigenetic alterations, in particular alterations in DNA methylation, have been observed during inflammation and inflammation-associated carcinogenesis. The mediators of this, the significance of these changes in DNA methylation and the effect this has on gene expression and the malignant transformation of the epithelial cells during IBD and CAC are discussed in this review. The recent advances in technologies to study genome-wide DNA methylation and the therapeutic potential of understanding these molecular mechanisms are also highlighted.


Asunto(s)
Colitis/fisiopatología , Neoplasias Colorrectales/fisiopatología , Metilación de ADN , Inflamación/fisiopatología , Colitis/complicaciones , Colitis/genética , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/genética , Humanos , Inflamación/complicaciones , Inflamación/genética , Factores de Riesgo
19.
Dig Dis ; 29(2): 172-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21734381

RESUMEN

Classically, adverse drug reactions had been considered as type A reactions which are related to the main pharmacological action of the drug and therefore are predictable. Such reactions are predictable, reversible, and usually can be managed by lowering the dose of the offending drug. However, other adverse effects of drugs can occur which are unrelated to the main pharmacological action of the drug - type B reactions. Such adverse effects are termed idiosyncratic and are often initiated by metabolites of the parent drug or by other indirect mechanisms. The detailed understanding of adverse drug events has become a major focus of the regulatory agencies throughout the world. The pharmacotherapy of gastrointestinal and liver disorders is becoming increasingly complex. In recent years, with the advent of novel therapeutic agents to treat a host of disorders, including viral hepatitis, gastrointestinal motility disorders, inflammatory bowel disease and others, the potential for serious clinically relevant drug reactions has increased. In the pharmacotherapy of gastrointestinal and liver diseases, a significant number of adverse events that occur can be explained by drug interactions. Some pharmacokinetic drug interactions are based on the competitive inhibition of the rate of drug metabolism of one of the drugs, leading to an increased concentration of the drug which was not intended. In other examples, the interaction can be mechanistic in which one or more drugs when co-administered potentiate each other's actions without any change in drug levels, termed pharmacodynamic interactions.


Asunto(s)
Tolerancia a Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Interacciones Farmacológicas , Humanos
20.
J Biol Chem ; 286(26): 22886-93, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21454695

RESUMEN

Transcription factor NF-κB is persistently activated in many chronic inflammatory diseases and cancers. The short term regulation of NF-κB is well understood, but little is known about the mechanisms of its long term activation. We studied the effect of a single application of TNF-α on NF-κB activity for up to 48 h in intestinal epithelial cells. Results show that NF-κB remained persistently activated up to 48 h after TNF-α and that the long term activation of NF-κB was accompanied by a biphasic degradation of IκBα. The first phase of IκBα degradation was proteasome-dependent, but the second was not. Further investigation showed that TNF-α stimulated formation of autophagosomes in intestinal epithelial cells and that IκBα co-localized with autophagosomal vesicles. Pharmacological or genetic blockade of autophagosome formation or the inhibition of lysosomal proteases decreased TNF-α-induced degradation of IκBα and lowered NF-κB target gene expression. Together, these findings indicate a role of autophagy in the control of long term NF-κB activity. Because abnormalities in autophagy have been linked to ineffective innate immunity, we propose that alterations in NF-κB may mediate this effect.


Asunto(s)
Autofagia/fisiología , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Fagosomas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/inmunología , Inmunidad Innata/fisiología , Ratones , Ratones Noqueados , Inhibidor NF-kappaB alfa , FN-kappa B/genética , FN-kappa B/inmunología , Células 3T3 NIH , Fagosomas/genética , Fagosomas/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...