Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Sci Rep ; 11(1): 13699, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34211054

RESUMEN

Oral iron promotes intestinal tumourigenesis in animal models. In humans, expression of iron transport proteins are altered in colorectal cancer. This study examined whether the route of iron therapy alters iron transport and tumour growth. Colorectal adenocarcinoma patients with pre-operative iron deficiency anaemia received oral ferrous sulphate (n = 15), or intravenous ferric carboxymaltose (n = 15). Paired (normal and tumour tissues) samples were compared for expression of iron loading, iron transporters, proliferation, apoptosis and Wnt signalling using immunohistochemistry and RT-PCR. Iron loading was increased in tumour and distributed to the stroma in intravenous treatment and to the epithelium in oral treatment. Protein and mRNA expression of proliferation and iron transporters were increased in tumours compared to normal tissues but there were no significant differences between the treatment groups. However, intravenous iron treatment reduced ferritin mRNA levels in tumours and replenished body iron stores. Iron distribution to non-epithelial cells in intravenous iron suggests that iron is less bioavailable to tumour cells. Therefore, intravenous iron may be a better option in the treatment of colorectal cancer patients with iron deficiency anaemia due to its efficiency in replenishing iron levels while its effect on proliferation and iron metabolism is similar to that of oral iron treatment.


Asunto(s)
Anemia Ferropénica/complicaciones , Neoplasias Colorrectales/complicaciones , Compuestos Férricos/uso terapéutico , Compuestos Ferrosos/uso terapéutico , Maltosa/análogos & derivados , Administración Intravenosa , Administración Oral , Anciano , Anciano de 80 o más Años , Anemia Ferropénica/metabolismo , Anemia Ferropénica/terapia , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/terapia , Femenino , Compuestos Férricos/administración & dosificación , Compuestos Ferrosos/administración & dosificación , Humanos , Hierro/metabolismo , Masculino , Maltosa/administración & dosificación , Maltosa/uso terapéutico , Persona de Mediana Edad
2.
Gastroenterology ; 161(4): 1245-1256.e20, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34146566

RESUMEN

BACKGROUND & AIMS: Irritable bowel syndrome (IBS) and inflammatory bowel diseases result in a substantial reduction in quality of life and a considerable socioeconomic impact. In IBS, diagnosis and treatment options are limited, but evidence for involvement of the gut microbiome in disease pathophysiology is emerging. Here we analyzed the prevalence of endoscopically visible mucosal biofilms in gastrointestinal disease and associated changes in microbiome composition and metabolism. METHODS: The presence of mucosal biofilms was assessed in 1426 patients at 2 European university-based endoscopy centers. One-hundred and seventeen patients were selected for in-depth molecular and microscopic analysis using 16S ribosomal RNA gene amplicon-sequencing of colonic biopsies and fecal samples, confocal microscopy with deep learning-based image analysis, scanning electron microscopy, metabolomics, and in vitro biofilm formation assays. RESULTS: Biofilms were present in 57% of patients with IBS and 34% of patients with ulcerative colitis compared with 6% of controls (P < .001). These yellow-green adherent layers of the ileum and right-sided colon were microscopically confirmed to be dense bacterial biofilms. 16S-sequencing links the presence of biofilms to a dysbiotic gut microbiome, including overgrowth of Escherichia coli and Ruminococcus gnavus. R. gnavus isolates cultivated from patient biofilms also formed biofilms in vitro. Metabolomic analysis found an accumulation of bile acids within biofilms that correlated with fecal bile acid excretion, linking this phenotype with a mechanism of diarrhea. CONCLUSIONS: The presence of mucosal biofilms is an endoscopic feature in a subgroup of IBS and ulcerative colitis with disrupted bile acid metabolism and bacterial dysbiosis. They provide novel insight into the pathophysiology of IBS and ulcerative colitis, illustrating that biofilm can be seen as a tipping point in the development of dysbiosis and disease.


Asunto(s)
Bacterias/crecimiento & desarrollo , Biopelículas/crecimiento & desarrollo , Colitis Ulcerosa/microbiología , Colon/microbiología , Colonoscopía , Microbioma Gastrointestinal , Mucosa Intestinal/microbiología , Síndrome del Colon Irritable/microbiología , Austria , Bacterias/metabolismo , Bacterias/ultraestructura , Estudios de Casos y Controles , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Colon/metabolismo , Colon/patología , Aprendizaje Profundo , Alemania , Humanos , Interpretación de Imagen Asistida por Computador , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Síndrome del Colon Irritable/metabolismo , Síndrome del Colon Irritable/patología , Metabolómica , Microscopía Confocal , Microscopía Electrónica de Rastreo , Valor Predictivo de las Pruebas , Ribotipificación
3.
Sci Rep ; 11(1): 5188, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33664327

RESUMEN

Inflammatory bowel disease is a group of conditions with rising incidence caused by genetic and environmental factors including diet. The chelator ethylenediaminetetraacetate (EDTA) is widely used by the food and pharmaceutical industry among numerous other applications, leading to a considerable environmental exposure. Numerous safety studies in healthy animals have revealed no relevant toxicity by EDTA. Here we show that, in the presence of intestinal inflammation, EDTA is surprisingly capable of massively exacerbating inflammation and even inducing colorectal carcinogenesis at doses that are presumed to be safe. This toxicity is evident in two biologically different mouse models of inflammatory bowel disease, the AOM/DSS and the IL10-/- model. The mechanism of this effect may be attributed to disruption of intercellular contacts as demonstrated by in vivo confocal endomicroscopy, electron microscopy and cell culture studies. Our findings add EDTA to the list of food additives that might be detrimental in the presence of intestinal inflammation, but the toxicity of which may have been missed by regulatory safety testing procedures that utilize only healthy models. We conclude that the current use of EDTA especially in food and pharmaceuticals should be reconsidered. Moreover, we suggest that intestinal inflammatory models should be implemented in the testing of food additives to account for the exposure of this primary organ to environmental and dietary stress.


Asunto(s)
Carcinogénesis/genética , Colitis/patología , Neoplasias del Colon/patología , Ácido Edético/efectos adversos , Animales , Carcinogénesis/efectos de los fármacos , Colitis/inducido químicamente , Colitis/genética , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Modelos Animales de Enfermedad , Aditivos Alimentarios/efectos adversos , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Interleucina-10/genética , Ratones , Ratones Noqueados
4.
Haematologica ; 106(3): 782-794, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32079699

RESUMEN

Iron deficiency (ID) is globally prevalent, and apart from anemia is associated with thrombocytosis. While considered benign, studies linking thrombotic events with prior ID anemia suggest otherwise. Herein we used animal models to assess the influence of ID on thrombotic tendency. Sprague-Dawley rats were fed control or iron deficient diets and ferric carboxymaltose was used to reverse ID. Thrombosis was induced via stenosis of the inferior vena cava or damage to the right carotid artery using ferric chloride. Thrombi were evaluated histologically and via high frequency ultrasound in the venous model. ID consistently induced thrombocytosis alongside anemia. Venous thrombus growth and final dimensions in both arterial and venous thrombi were largest in ID. In both models, platelet numbers correlated with the final thrombus size, with ID thrombi having the largest platelet areas. Platelet function was also evaluated in surgically naive rats. Coagulability on thromboelastography and hemostasis on tail transection were augmented in ID. Platelet and plasma P-selectin expression were both higher in ID. Platelet adhesion and aggregation in ID was impaired under shear flow but was intact on static assays. Iron replacement therapy reversed all ID-related changes in hematological parameters, thrombus dimensions, and platelet assays. In summary, ID alone increases thrombotic tendency. Iron replacement therapy reverses these changes, making it a viable strategy for prevention of ID-related thrombotic disease. This may be of importance in patients with chronic illnesses which may already be at increased risk for thrombosis such as inflammatory bowel disease, chronic kidney disease, or cancer.


Asunto(s)
Anemia Ferropénica , Trombocitosis , Trombosis , Anemia Ferropénica/etiología , Animales , Plaquetas , Humanos , Ratas , Ratas Sprague-Dawley , Trombocitosis/etiología , Trombosis/etiología
5.
Sci Rep ; 10(1): 2869, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32071351

RESUMEN

Bioimpedance spectroscopy (BIS) is routinely used in peritoneal dialysis patients and might aid fluid status assessment in patients with liver cirrhosis, but the effect of ascites volume removal on BIS-readings is unknown. Here we determined changes in BIS-derived parameters and clinical signs of fluid overload from before to after abdominal paracentesis. Per our pre-specified sample size calculation, we studied 31 cirrhotic patients, analyzing demographics, labs and clinical parameters along with BIS results. Mean volume of the abdominal paracentesis was 7.8 ± 2.6 L. From pre-to post-paracentesis, extracellular volume (ECV) decreased (20.2 ± 5.2 L to 19.0 ± 4.8 L), total body volume decreased (39.8 ± 9.8 L to 37.8 ± 8.5 L) and adipose tissue mass decreased (38.4 ± 16.0 kg to 29.9 ± 12.9 kg; all p < 0.002). Correlation of BIS-derived parameters from pre to post-paracentesis ranged from R² = 0.26 for body cell mass to R² = 0.99 for ECV. Edema did not correlate with BIS-derived fluid overload (FO ≥ 15% ECV), which occurred in 16 patients (51.6%). In conclusion, BIS-derived information on fluid status did not coincide with clinical judgement. The changes in adipose tissue mass support the BIS-model assumption that fluid in the peritoneal cavity is not detectable, suggesting that ascites (or peritoneal dialysis fluid) mass should be subtracted from adipose tissue if BIS is used in patients with a full peritoneal cavity.


Asunto(s)
Ascitis/metabolismo , Espectroscopía Dieléctrica , Líquido Extracelular/metabolismo , Cirrosis Hepática/metabolismo , Anciano , Ascitis/patología , Composición Corporal , Soluciones para Diálisis/metabolismo , Femenino , Humanos , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/patología , Masculino , Persona de Mediana Edad , Diálisis Peritoneal/efectos adversos , Desequilibrio Hidroelectrolítico/metabolismo , Desequilibrio Hidroelectrolítico/patología
6.
Sci Rep ; 9(1): 2842, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30809073

RESUMEN

Disruption of mucosal structure and barrier function contribute to the pathogenesis of inflammatory bowel disease (IBD). Efficacy of therapy in IBD is based on endoscopic mucosal healing, which occurs by a dynamic interplay of epithelial cell regeneration, migration and differentiation. Both mesalamine (5-ASA) and azathioprine (AZTP) promote this process through mechanisms not clearly understood. We examined molecular pathways implicated in epithelial barrier function that were altered by 5-ASA and AZTP. Paracellular permeability induced by inflammatory mediators was mitigated by both compounds through restoration of cellular anchoring complexes. 5-ASA and AZTP induced rearrangement and membranous localization of junctional proteins and modulated genes involved in tight junctions. Intestinal organoids from wildtype-mice treated with TNF-α and IL-10- deficient-mice displayed impaired epithelial barrier with loss of membranous E-cadherin and reduced Desmoglein-2 expression. These effects were counteracted by 5-ASA and AZTP. Unlike AZTP that exhibited antiproliferative effects, 5-ASA promoted wound healing in colon epithelial cells. Both affected cellular senescence, cell cycle distribution and restricted cells in G1 or S phase without inducing apoptosis. This study provides mechanistic evidence that molecular actions of 5-ASA and AZTP on intestinal epithelia are fundamental in the resolution of barrier dysfunction.


Asunto(s)
Azatioprina/farmacología , Células Epiteliales/efectos de los fármacos , Inflamación , Enfermedades Inflamatorias del Intestino/fisiopatología , Intestinos/efectos de los fármacos , Mesalamina/farmacología , Animales , Antiinflamatorios no Esteroideos , Azatioprina/uso terapéutico , Colitis , Células Epiteliales/fisiología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Intestinos/fisiopatología , Mesalamina/uso terapéutico , Ratones , Cicatrización de Heridas
7.
Cancer Res ; 77(9): 2424-2438, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28428272

RESUMEN

HuR is an RNA-binding protein implicated in immune homeostasis and various cancers, including colorectal cancer. HuR binding to AU-rich elements within the 3' untranslated region of mRNAs encoding oncogenes, growth factors, and various cytokines leads message stability and translation. In this study, we evaluated HuR as a small-molecule target for preventing colorectal cancer in high-risk groups such as those with familial adenomatosis polyposis (FAP) or inflammatory bowel disease (IBD). In human specimens, levels of cytoplasmic HuR were increased in colonic epithelial cells from patients with IBD, IBD-cancer, FAP-adenoma, and colorectal cancer, but not in patients with IBD-dysplasia. Intraperitoneal injection of the HuR small-molecule inhibitor MS-444 in AOM/DSS mice, a model of IBD and inflammatory colon cancer, augmented DSS-induced weight loss and increased tumor multiplicity, size, and invasiveness. MS-444 treatment also abrogated tumor cell apoptosis and depleted tumor-associated eosinophils, accompanied by a decrease in IL18 and eotaxin-1. In contrast, HuR inhibition in APCMin mice, a model of FAP and colon cancer, diminished the number of small intestinal tumors generated. In this setting, fecal microbiota, evaluated by 16S rRNA gene amplicon sequencing, shifted to a state of reduced bacterial diversity, with an increased representation of Prevotella, Akkermansia, and Lachnospiraceae Taken together, our results indicate that HuR activation is an early event in FAP-adenoma but is not present in IBD-dysplasia. Furthermore, our results offer a preclinical proof of concept for HuR inhibition as an effective means of FAP chemoprevention, with caution advised in the setting of IBD. Cancer Res; 77(9); 2424-38. ©2017 AACR.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Neoplasias Colorrectales/genética , Proteína 1 Similar a ELAV/genética , Enfermedades Inflamatorias del Intestino/genética , Poliposis Adenomatosa del Colon/microbiología , Poliposis Adenomatosa del Colon/patología , Animales , Apoptosis/efectos de los fármacos , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Quimiocina CCL11/genética , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Proteína 1 Similar a ELAV/antagonistas & inhibidores , Heces/microbiología , Furanos/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Células HCT116 , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Interleucina-18/genética , Ratones , Naftoles/administración & dosificación , Células RAW 264.7
8.
Wien Med Wochenschr ; 166(13-14): 437-446, 2016 Oct.
Artículo en Alemán | MEDLINE | ID: mdl-27682430

RESUMEN

Iron deficiency, the most common nutritional deficiency worldwide, is often associated with reactive thrombocytosis. Although secondary thrombocytosis is commonly considered to be harmless, there is accumulating evidence that elevated platelet counts, especially in the setting of iron deficiency, can lead to an increased thromboembolic risk in both arterial and venous systems. Here we present the mechanisms of iron deficiency-induced thrombocytosis and summarize its clinical consequences especially in patients with inflammatory bowel diseases, chronic kidney disease or cancer. We hypothesize that iron deficiency is an underestimated thromboembolic risk factor, and that iron replacement therapy can become an effective preventive strategy in a variety of clinical settings.


Asunto(s)
Anemia Ferropénica/fisiopatología , Trombocitosis/fisiopatología , Tromboembolia/fisiopatología , Trombosis/fisiopatología , Adolescente , Adulto , Anemia Ferropénica/complicaciones , Niño , Preescolar , Femenino , Hemoglobinometría , Humanos , Lactante , Masculino , Persona de Mediana Edad , Factores de Riesgo , Adulto Joven
10.
Wien Klin Wochenschr ; 127(23-24): 907-19, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26373748

RESUMEN

Iron deficiency and iron deficiency-associated anemia are common complications in cancer patients. Most iron deficient cancer patients present with functional iron deficiency (FID), a status with adequate storage iron, but insufficient iron supply for erythroblasts and other iron dependent tissues. FID is the consequence of the cancer-associated cytokine release, while in absolute iron deficiency iron stores are depleted resulting in similar but often more severe symptoms of insufficient iron supply. Here we present a short review on the epidemiology, pathophysiology, diagnosis, clinical symptoms, and treatment of iron deficiency in cancer patients. Special emphasis is given to intravenous iron supplementation and on the benefits and limitations of different formulations. Based on these considerations and recommendations from current international guidelines we developed recommendations for clinical practice and classified the level of evidence and grade of recommendation according to the principles of evidence-based medicine.


Asunto(s)
Anemia Ferropénica/metabolismo , Anemia Ferropénica/prevención & control , Suplementos Dietéticos/normas , Hierro/metabolismo , Hierro/uso terapéutico , Neoplasias/metabolismo , Anemia Ferropénica/etiología , Austria , Medicina Basada en la Evidencia , Humanos , Oncología Médica/normas , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Guías de Práctica Clínica como Asunto , Resultado del Tratamiento
11.
Cancer Prev Res (Phila) ; 8(11): 1093-101, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26304465

RESUMEN

p21-activated kinase 1 (PAK1) is a serine/threonine kinase that is overexpressed in colorectal cancer. PAK1 is a target of mesalamine [5-aminosylicylic acid (5-ASA)], a common drug for the treatment of ulcerative colitis with prospective chemopreventive properties. Here, we investigated whether PAK1 deletion impedes tumorigenesis in murine intestinal cancer models. Ten-week-old APC(min) or APC(min)/PAK1(-/-) mice were monitored for 8 weeks, euthanized, and assessed for tumor number and size. Six- to 8-week-old PAK1(-/-) and wild-type (WT) mice received one 10 mg/kg intraperitoneal injection of azoxymethane (AOM) and four cycles of 1.7% dextran sodium sulfate (DSS) for 4 days followed by 14 days of regular water. Mice also received 5-ASA via diet. Tumor incidence and size was assessed via colonoscopy and pathology. Molecular targets of PAK1 and 5-ASA were evaluated via immunohistochemistry (IHC) in both models. PAK1 deletion reduced tumor multiplicity and tumor burden but did not alter average tumor size in APC(min) mice. IHC revealed that PAK1 deletion reduced p-AKT, ß-catenin, and c-Myc expression in APC(min) adenomas. Colonoscopy and pathologic analysis revealed that PAK1 deletion reduced tumor multiplicity without affecting tumor size in AOM/DSS-treated mice. 5-ASA treatment and PAK1 deletion impeded tumor multiplicity and dysplastic lesions in AOM/DSS mice. IHC further revealed that 5-ASA blocked ß-catenin signaling via inhibition of PAK1/p-AKT. These data indicate that PAK1 contributes to initiation of intestinal carcinogenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Intestinales/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Azoximetano/química , Carcinogénesis , Colonoscopía , Dextranos/química , Femenino , Eliminación de Gen , Genotipo , Heterocigoto , Inmunohistoquímica , Inflamación , Masculino , Mesalamina/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , beta Catenina/metabolismo
12.
Biochim Biophys Acta ; 1853(10 Pt A): 2349-60, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26036343

RESUMEN

P21-activated kinases (PAKs) are multifunctional effectors of Rho GTPases with both kinase and scaffolding activity. Here, we investigated the effects of inflammation on PAK1 signaling and its role in colitis-driven carcinogenesis. PAK1 and p-PAK1 (Thr423) were assessed by immunohistochemistry, immunofluorescence, and Western blot. C57BL6/J wildtype mice were treated with a single intraperitoneal TNFα injection. Small intestinal organoids from these mice and from PAK1-KO mice were cultured with TNFα. NF-κB and PPARγ were analyzed upon PAK1 overexpression and silencing for transcriptional/translational regulation. PAK1 expression and activation was increased on the luminal intestinal epithelial surface in inflammatory bowel disease and colitis-associated cancer. PAK1 was phosphorylated upon treatment with IFNγ, IL-1ß, and TNFα. In vivo, mice administered with TNFα showed increased p-PAK1 in intestinal villi, which was associated with nuclear p65 and NF-κB activation. p65 nuclear translocation downstream of TNFα was strongly inhibited in PAK1-KO small intestinal organoids. PAK1 overexpression induced a PAK1-p65 interaction as visualized by co-immunoprecipitation, nuclear translocation, and increased NF-κB transactivation, all of which were impeded by kinase-dead PAK1. Moreover, PAK1 overexpression downregulated PPARγ and mesalamine recovered PPARγ through PAK1 inhibition. On the other hand PAK1 silencing inhibited NF-κB, which was recovered using BADGE, a PPARγ antagonist. Altogether these data demonstrate that PAK1 overexpression and activation in inflammation and colitis-associated cancer promote NF-κB activity via suppression of PPARγ in intestinal epithelial cells.


Asunto(s)
Colitis/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Animales , Línea Celular , Colitis/genética , Colitis/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Intestinos/patología , Ratones , Ratones Noqueados , FN-kappa B/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , PPAR gamma/genética , Quinasas p21 Activadas/genética
13.
Gut ; 64(12): 1905-12, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25429050

RESUMEN

OBJECTIVE: Lynch syndrome is caused by germline mutations in DNA mismatch repair genes leading to microsatellite instability (MSI) and colorectal cancer. Mesalazine, commonly used for the treatment of UC, reduces MSI in vitro. Here, we tested natural compounds for such activity and applied mesalazine and thymoquinone in a Msh2(loxP/loxP) Villin-Cre mouse model for Lynch syndrome. DESIGN: Flow cytometry was used for quantitation of mutation rates at a CA13 microsatellite in human colon cancer (HCT116) cells that had been stably transfected with pIREShyg2-enhanced green fluorescent protein/CA13, a reporter for frameshift mutations. Mice were treated for 43 weeks with mesalazine, thymoquinone or control chow. Intestines were analysed for tumour incidence, tumour multiplicity and size. MSI testing was performed from microdissected normal intestinal or tumour tissue, compared with mouse tails and quantified by the number of mutations per marker (NMPM). RESULTS: Besides mesalazine, thymoquinone significantly improved replication fidelity at 1.25 and 2.5 µM in HCT116 cells. In Msh2(loxP/loxP) Villin-Cre mice, tumour incidence was reduced by mesalazine from 94% to 69% (p=0.04) and to 56% (p=0.003) by thymoquinone. The mean number of tumours was reduced from 3.1 to 1.4 by mesalazine (p=0.004) and to 1.1 by thymoquinone (p<0.001). Interestingly, MSI was reduced in normal intestinal tissue from 1.5 to 1.2 NMPM (p=0.006) and to 1.1 NMPM (p=0.01) by mesalazine and thymoquinone, respectively. Thymoquinone, but not mesalazine, reduced MSI in tumours. CONCLUSIONS: Mesalazine and thymoquinone reduce tumour incidence and multiplicity in Msh2(loxP/loxP) Villin-Cre mice by reduction of MSI independent of a functional mismatch repair system. Both substances are candidate compounds for chemoprevention in Lynch syndrome mutation carriers.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Benzoquinonas/uso terapéutico , Neoplasias Colorrectales Hereditarias sin Poliposis/prevención & control , Mesalamina/uso terapéutico , Proteína 2 Homóloga a MutS/genética , Animales , Antiinflamatorios no Esteroideos/farmacología , Benzoquinonas/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Modelos Animales de Enfermedad , Femenino , Mutación del Sistema de Lectura , Células HCT116 , Humanos , Mucosa Intestinal/metabolismo , Masculino , Mesalamina/farmacología , Ratones , Inestabilidad de Microsatélites/efectos de los fármacos , Proteína 2 Homóloga a MutS/metabolismo , Tasa de Mutación , Carga Tumoral/efectos de los fármacos
14.
Am J Hematol ; 89(5): 524-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24464533

RESUMEN

Iron deficiency is a common cause of reactive thrombocytosis, however, the exact pathways have not been revealed. Here we aimed to study the mechanisms behind iron deficiency-induced thrombocytosis. Within few weeks, iron-depleted diet caused iron deficiency in young Sprague-Dawley rats, as reflected by a drop in hemoglobin, mean corpuscular volume, hepatic iron content and hepcidin mRNA in the liver. Thrombocytosis established in parallel. Moreover, platelets produced in iron deficient animals displayed a higher mean platelet volume and increased aggregation. Bone marrow studies revealed subtle alterations that are suggestive of expansion of megakaryocyte progenitors, an increase in megakaryocyte ploidy and accelerated megakaryocyte differentiation. Iron deficiency did not alter the production of hematopoietic growth factors such as thrombopoietin, interleukin 6 or interleukin 11. Megakaryocytic cell lines grown in iron-depleted conditions exhibited reduced proliferation but increased ploidy and cell size. Our data suggest that iron deficiency increases megakaryopoietic differentiation and alters platelet phenotype without changes in megakaryocyte growth factors, specifically TPO. Iron deficiency-induced thrombocytosis may have evolved to maintain or increase the coagulation capacity in conditions with chronic bleeding.


Asunto(s)
Plaquetas/metabolismo , Deficiencias de Hierro , Hierro/sangre , Megacariocitos/metabolismo , Mielopoyesis/fisiología , Trombopoyetina/metabolismo , Animales , Plaquetas/citología , Hierro/metabolismo , Masculino , Megacariocitos/citología , Fenotipo , Agregación Plaquetaria/fisiología , Recuento de Plaquetas/métodos , Ratas , Ratas Sprague-Dawley
15.
Mol Cancer ; 12(1): 41, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23668310

RESUMEN

BACKGROUND: Patients with familial adenomatous polyposis (FAP) are at increased risk for the development of colorectal cancer. Surgery and chemoprevention are the most effective means to prevent cancer development. Thymoquinone (TQ) is considered the main compound of the volatile Nigella sativa seed oil and has been reported to possess anticarcinogenic properties. In this study we evaluated the chemopreventive properties of TQ in a mouse model of FAP. METHODS: APCMin mice were fed with chow containing 37.5 mg/kg or 375 mg/kg TQ for 12 weeks. H&E stained intestine tissue sections were assessed for tumor number, localization, size, and grade. Immunohistochemistry for ß-catenin, c-myc, Ki-67 and TUNEL-staining was performed to investigate TQ's effect on major colorectal cancer pathways. TQ's impact on GSK-3ß and ß-catenin were studied in RKO cells. RESULTS: 375 mg/kg but not 37.5 mg/kg TQ decreased the number of large polyps in the small intestine of APCMin mice. TQ induced apoptosis in the neoplastic tissue but not in the normal mucosa. Furthermore, upon TQ treatment, ß-catenin was retained at the membrane and c-myc decreased in the nucleus, which was associated with a reduced cell proliferation in the villi. In vitro, TQ activated GSK-3ß, which induced membranous localization of ß-catenin and reduced nuclear c-myc expression. CONCLUSIONS: In summary, TQ interferes with polyp progression in ApcMin mice through induction of tumor-cell specific apoptosis and by modulating Wnt signaling through activation of GSK-3ß. Nigella sativa oil (or TQ) might be useful as nutritional supplement to complement surgery and chemoprevention in FAP.


Asunto(s)
Poliposis Adenomatosa del Colon/tratamiento farmacológico , Anticarcinógenos/farmacología , Benzoquinonas/farmacología , Neoplasias Colorrectales/prevención & control , Vía de Señalización Wnt , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Animales , Anticarcinógenos/uso terapéutico , Apoptosis , Benzoquinonas/uso terapéutico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Mutantes , Aceites de Plantas/química , beta Catenina/metabolismo
16.
Inflamm Bowel Dis ; 19(8): 1609-16, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23644823

RESUMEN

BACKGROUND: Secondary thrombocytosis is a common clinical feature. In patients with cancer, it is a risk factor for venous thromboembolic events. In inflammatory bowel disease (IBD), thrombocytosis is so far considered a marker of active disease and may contribute to the increased thromboembolic risk in this population. Observed effects of iron therapy on normalization of platelet counts led us to hypothesize that iron itself may regulate megakaryopoiesis. Here, we want to test the effect of iron replacement on platelet count and activity in IBD-associated thrombocytosis. METHODS: We performed a randomized, single-blinded placebo-controlled trial testing the effect of ferric carboxymaltose (FCM) in patients with IBD with secondary thrombocytosis (platelets > 450 G/L). Changes in platelet counts, hemoglobin, iron parameters, disease activity, megakaryopoietic growth factors, erythropoietin, and platelet activity were assessed. Patients received placebo or up to 1500 mg iron as FCM. Endpoints were evaluated at week 6. RESULTS: A total of 26 patients were included in the study, 15 patients were available for the per protocol analysis. A drop in platelets >25% (primary endpoint) was observed in 4 of 8 (50%, iron group) and 1 of 7 patients (14%, placebo group, P = 0.143). Mean platelet counts dropped on FCM but not on placebo (536 G/L to 411 G/L versus 580 G/L to 559 G/L; P = 0.002). Disease activity and megakaryopoietic growth factors remained unchanged and hemoglobin and iron parameters increased on FCM. The normalization of platelet counts was associated with a decrease in platelet aggregation and P-selectin expression. CONCLUSION: FCM lowers platelet counts and platelet activation in patients with IBD-associated secondary thrombocytosis.


Asunto(s)
Anemia Ferropénica/fisiopatología , Compuestos Férricos/uso terapéutico , Enfermedades Inflamatorias del Intestino/complicaciones , Maltosa/análogos & derivados , Activación Plaquetaria , Trombocitosis/etiología , Adolescente , Adulto , Eritropoyetina/metabolismo , Femenino , Hepcidinas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Maltosa/uso terapéutico , Persona de Mediana Edad , Selectina-P/metabolismo , Agregación Plaquetaria , Recuento de Plaquetas , Pronóstico , Estudios Prospectivos , Trombocitosis/tratamiento farmacológico , Adulto Joven
17.
Clin Gastroenterol Hepatol ; 11(3): 269-77, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23078888

RESUMEN

BACKGROUND & AIMS: Iron-deficiency anemia is the most common systemic complication of inflammatory bowel diseases (IBD). Iron-deficiency anemia recurs frequently and rapidly after iron-replacement therapy in patients with IBD. We performed a randomized, placebo-controlled trial to determine if administration of ferric carboxymaltose (FCM) prevents anemia in patients with IBD and low levels of serum ferritin. METHODS: We performed a single-blind, multicenter study of nonanemic patients who had completed the FERGIcor study. Serum levels of ferritin were assessed every second month, and patients were given FCM (total iron dose, 1181 ± 662 mg; n = 105) or placebo (n = 99) when levels decreased to less than 100 µg/L. The primary end point was time to recurrence of anemia within 8 months. Secondary end points included changes of quality of life, disease activity, results from laboratory tests, and adverse events. RESULTS: Anemia recurred in 26.7% of subjects given FCM and in 39.4% given placebo. The time to anemia recurrence was longer in the FCM group (hazard ratio, 0.62; 95% confidence interval, 0.38-1.00; P = .049). Markers of body levels of iron increased or remained at normal levels in subjects given FCM (ferritin increased by 30.3 µg/L, transferrin saturation increased by 0.6%) but decreased in the group given placebo (ferritin decreased by 36.1 µg/L, transferrin saturation decreased by 4.0%). Changes in quality of life and disease activity were comparable between groups. Adverse events were reported in 59.0% of the FCM group and 50.5% of the placebo group, and serious adverse events were reported in 6.7% and 8.1%, respectively. CONCLUSIONS: FCM prevents recurrence of anemia in patients with IBD, compared with placebo. Nevertheless, the high rate of anemia recurrence warrants optimization of the frequency and requirements for FCM treatment.


Asunto(s)
Anemia Ferropénica/tratamiento farmacológico , Suplementos Dietéticos , Compuestos Férricos/uso terapéutico , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Maltosa/análogos & derivados , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Maltosa/uso terapéutico , Persona de Mediana Edad , Placebos/administración & dosificación , Prevención Secundaria , Método Simple Ciego , Resultado del Tratamiento , Adulto Joven
18.
Biochem Pharmacol ; 85(2): 234-44, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23146664

RESUMEN

Mesalamine (5-ASA) is widely used for the treatment of ulcerative colitis, a remitting condition characterized by chronic inflammation of the colon. Knowledge about the molecular and cellular targets of 5-ASA is limited and a clear understanding of its activity in intestinal homeostasis and interference with neoplastic progression is lacking. We sought to identify molecular pathways interfered by 5-ASA, using CRC cell lines with different genetic background. Microarray was performed for gene expression profile of 5-ASA-treated and untreated cells (HCT116 and HT29). Filtering and analysis of data identified three oncogenic pathways interfered by 5-ASA: MAPK/ERK pathway, cell adhesion and ß-catenin/Wnt signaling. PAK1 emerged as a consensus target of 5-ASA, orchestrating these pathways. We further investigated the effect of 5-ASA on cell adhesion. 5-ASA increased cell adhesion which was measured by cell adhesion assay and transcellular-resistance measurement. Moreover, 5-ASA treatment restored membranous expression of adhesion molecules E-cadherin and ß-catenin. Role of PAK1 as a mediator of mesalamine activity was validated in vitro and in vivo. Inhibition of PAK1 by RNA interference also increased cell adhesion. PAK1 expression was elevated in APC(min) polyps and 5-ASA treatment reduced its expression. Our data demonstrates novel pharmacological mechanism of mesalamine in modulation of cell adhesion and role of PAK1 in APC(min) polyposis. We propose that inhibition of PAK1 expression by 5-ASA can impede with neoplastic progression in colorectal carcinogenesis. The mechanism of PAK1 inhibition and induction of membranous translocation of adhesion proteins by 5-ASA might be independent of its known anti-inflammatory action.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Anticarcinógenos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Mesalamina/farmacología , Quinasas p21 Activadas/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Anticarcinógenos/uso terapéutico , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Neoplasias del Colon/prevención & control , Femenino , Perfilación de la Expresión Génica , Humanos , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Pólipos Intestinales/prevención & control , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Mesalamina/uso terapéutico , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Interferencia de ARN , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/genética , beta Catenina/metabolismo , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/genética
19.
PLoS One ; 7(4): e34520, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22506024

RESUMEN

BACKGROUND AND AIMS: Secondary thrombocytosis is a clinical feature of unknown significance. In inflammatory bowel disease (IBD), thrombocytosis is considered a marker of active disease; however, iron deficiency itself may trigger platelet generation. In this study we tested the effect of iron therapy on platelet counts in patients with IBD-associated anemia. METHODS: Platelet counts were analyzed before and after iron therapy from four prospective clinical trials. Further, changes in hemoglobin, transferrin saturation, ferritin, C-reactive protein, and leukocyte counts, before and after iron therapy were compared. In a subgroup the effect of erythropoietin treatment was tested. The results were confirmed in a large independent cohort (FERGIcor). RESULTS: A total of 308 patient records were available for the initial analysis. A dose-depended drop in platelet counts (mean 425 G/L to 320 G/L; p<0.001) was found regardless of the type of iron preparation (iron sulphate, iron sucrose, or ferric carboxymaltose). Concomitant erythropoietin therapy as well as parameters of inflammation (leukocyte counts, C-reactive protein) had no effect on the change in platelet counts. This effect of iron therapy on platelets was confirmed in the FERGIcor study cohort (n=448, mean platelet counts before iron therapy: 383 G/L, after: 310 G/L, p<0.001). CONCLUSION: Iron therapy normalizes elevated platelet counts in patients with IBD-associated anemia. Thus, iron deficiency is an important pathogenetic mechanism of secondary thrombocytosis in IBD.


Asunto(s)
Anemia Ferropénica/sangre , Anemia Ferropénica/tratamiento farmacológico , Plaquetas/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Compuestos de Hierro/uso terapéutico , Hierro/uso terapéutico , Anemia Ferropénica/metabolismo , Plaquetas/citología , Plaquetas/metabolismo , Proteína C-Reactiva/metabolismo , Estudios de Cohortes , Eritropoyetina/uso terapéutico , Ferritinas/metabolismo , Hemoglobinas/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/metabolismo , Deficiencias de Hierro , Recuento de Leucocitos , Recuento de Plaquetas/métodos , Estudios Prospectivos , Trombocitosis/sangre , Trombocitosis/tratamiento farmacológico , Trombocitosis/metabolismo , Transferrina/metabolismo
20.
Gut ; 61(6): 933-52, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22016365

RESUMEN

Iron deficiency is a common condition increasingly diagnosed and treated by gastroenterologists. The most common presentation of iron deficiency is anaemia; however, it is a systemic disorder affecting multiple aspects of health in various organs. Iron is an essential element, with iron-containing proteins exerting a variety of vital functions, including oxygen transport, cellular respiration, intermediary metabolism, regulation of transcription and DNA repair. Major pathways of iron utilisation and production of iron-containing proteins include iron sulphur cluster biosynthesis, haem synthesis and storage within ferritin. The main site of iron absorption is the small intestine, but most iron is recycled by the monocyte-macrophage system via phagocytosis of senescent erythrocytes. Hepcidin, the key iron-regulating peptide binds to the iron exporter ferroportin and leads to its degradation, thereby inhibiting intestinal iron absorption and cellular export. Hepcidin levels are regulated on a transcriptional level by various stimuli, including transferrin saturation, erythropoietic activity, hypoxia and inflammation. Iron deficiency evokes adaptive responses resulting in alteration of cellular metabolism, changes in gene expression, activation of signalling pathways, cell cycle regulation, differentiation and cell death. Such responses are mediated by a number of iron-sensitive signalling pathways, including the IRE/IRP system, HIF and haem signalling. This review provides a molecular perspective for the clinician and highlights important biological aspects of iron deficiency.


Asunto(s)
Deficiencias de Hierro , Péptidos Catiónicos Antimicrobianos/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Eritropoyesis/fisiología , Hemo/fisiología , Hemo-Oxigenasa 1/fisiología , Hemocromatosis/metabolismo , Hemocromatosis/fisiopatología , Hepcidinas , Homeostasis/fisiología , Humanos , Factor 1 Inducible por Hipoxia/fisiología , Hierro/metabolismo , Hierro/fisiología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...