Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 160(1-2): 269-84, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594183

RESUMEN

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).


Asunto(s)
Huesos/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Intestino Delgado/citología , Células Madre Mesenquimatosas/citología , Animales , Cartílago/metabolismo , Intestino Delgado/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Mol Cell ; 82(16): 3045-3060.e11, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35752173

RESUMEN

Cancer mortality is primarily a consequence of its metastatic spread. Here, we report that methionine sulfoxide reductase A (MSRA), which can reduce oxidized methionine residues, acts as a suppressor of pancreatic ductal adenocarcinoma (PDA) metastasis. MSRA expression is decreased in the metastatic tumors of PDA patients, whereas MSRA loss in primary PDA cells promotes migration and invasion. Chemoproteomic profiling of pancreatic organoids revealed that MSRA loss results in the selective oxidation of a methionine residue (M239) in pyruvate kinase M2 (PKM2). Moreover, M239 oxidation sustains PKM2 in an active tetrameric state to promote respiration, migration, and metastasis, whereas pharmacological activation of PKM2 increases cell migration and metastasis in vivo. These results demonstrate that methionine residues can act as reversible redox switches governing distinct signaling outcomes and that the MSRA-PKM2 axis serves as a regulatory nexus between redox biology and cancer metabolism to control tumor metastasis.


Asunto(s)
Carcinoma Ductal Pancreático , Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias Pancreáticas , Hormonas Tiroideas/metabolismo , Carcinoma Ductal Pancreático/genética , Humanos , Metionina , Metionina Sulfóxido Reductasas/química , Metionina Sulfóxido Reductasas/metabolismo , Oxidación-Reducción , Neoplasias Pancreáticas/genética , Piruvato Quinasa/metabolismo , Proteínas de Unión a Hormona Tiroide , Neoplasias Pancreáticas
3.
Nature ; 610(7931): 356-365, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36198802

RESUMEN

Hepatocellular carcinoma (HCC), the fourth leading cause of cancer mortality worldwide, develops almost exclusively in patients with chronic liver disease and advanced fibrosis1,2. Here we interrogated functions of hepatic stellate cells (HSCs), the main source of liver fibroblasts3, during hepatocarcinogenesis. Genetic depletion, activation or inhibition of HSCs in mouse models of HCC revealed their overall tumour-promoting role. HSCs were enriched in the preneoplastic environment, where they closely interacted with hepatocytes and modulated hepatocarcinogenesis by regulating hepatocyte proliferation and death. Analyses of mouse and human HSC subpopulations by single-cell RNA sequencing together with genetic ablation of subpopulation-enriched mediators revealed dual functions of HSCs in hepatocarcinogenesis. Hepatocyte growth factor, enriched in quiescent and cytokine-producing HSCs, protected against hepatocyte death and HCC development. By contrast, type I collagen, enriched in activated myofibroblastic HSCs, promoted proliferation and tumour development through increased stiffness and TAZ activation in pretumoural hepatocytes and through activation of discoidin domain receptor 1 in established tumours. An increased HSC imbalance between cytokine-producing HSCs and myofibroblastic HSCs during liver disease progression was associated with increased HCC risk in patients. In summary, the dynamic shift in HSC subpopulations and their mediators during chronic liver disease is associated with a switch from HCC protection to HCC promotion.


Asunto(s)
Carcinogénesis , Carcinoma Hepatocelular , Células Estrelladas Hepáticas , Neoplasias Hepáticas , Animales , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Proliferación Celular , Colágeno Tipo I/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Progresión de la Enfermedad , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Factor de Crecimiento de Hepatocito/metabolismo , Hepatocitos , Humanos , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/patología , Ratones , Miofibroblastos/patología
4.
J Card Fail ; 30(4): 580-591, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37625581

RESUMEN

BACKGROUND: Venous congestion (VC) is a hallmark of symptomatic heart failure (HF) requiring hospitalization; however, its role in the pathogenesis of HF progression remains unclear. We investigated whether peripheral VC exacerbates inflammation, oxidative stress and neurohormonal and endothelial cell (EC) activation in patients with HF with reduced ejection fraction (HFrEF). METHODS AND RESULTS: Two matched groups of patients with HFrEF and with no peripheral VC vs without recent HF hospitalization were studied. We modeled peripheral VC by inflating a cuff around the dominant arm, targeting ∼ 30 mmHg increase in venous pressure (venous stress test [VST]). Blood and ECs were sampled before and after 90 minutes of VST. We studied 44 patients (age 53 ± 12 years, 32% female). Circulating endothelin-1, tumor necrosis factor-α, interleukin-6, isoprostane, angiotensin II (ang-2), angiopoietin-2, vascular cell adhesion molecule-1, and CD146 significantly increased after the VST. Enhanced endothelin-1 and angiopoietin-2 responses to the VST were present in patients with vs without recent hospitalization and were prospectively associated with incident HF-related events; 6698 messenger ribonucleic acid (mRNA probe sets were differentially expressed in ECs after VST. CONCLUSIONS: Experimental VC exacerbates inflammation, oxidative stress, neurohormonal and EC activation and promotes unfavorable transcriptome remodeling in ECs of patients with HFrEF. A distinct biological sensitivity to VC appears to be associated with high risk for HF progression.


Asunto(s)
Insuficiencia Cardíaca Sistólica , Insuficiencia Cardíaca , Hiperemia , Humanos , Femenino , Adulto , Persona de Mediana Edad , Anciano , Masculino , Angiopoyetina 2/metabolismo , Endotelina-1 , Volumen Sistólico , Inflamación , Células Endoteliales , Estrés Oxidativo
6.
Nature ; 543(7645): 385-390, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28273060

RESUMEN

Bone has recently emerged as a pleiotropic endocrine organ that secretes at least two hormones, FGF23 and osteocalcin, which regulate kidney function and glucose homeostasis, respectively. These findings have raised the question of whether other bone-derived hormones exist and what their potential functions are. Here we identify, through molecular and genetic analyses in mice, lipocalin 2 (LCN2) as an osteoblast-enriched, secreted protein. Loss- and gain-of-function experiments in mice demonstrate that osteoblast-derived LCN2 maintains glucose homeostasis by inducing insulin secretion and improves glucose tolerance and insulin sensitivity. In addition, osteoblast-derived LCN2 inhibits food intake. LCN2 crosses the blood-brain barrier, binds to the melanocortin 4 receptor (MC4R) in the paraventricular and ventromedial neurons of the hypothalamus and activates an MC4R-dependent anorexigenic (appetite-suppressing) pathway. These results identify LCN2 as a bone-derived hormone with metabolic regulatory effects, which suppresses appetite in a MC4R-dependent manner, and show that the control of appetite is an endocrine function of bone.


Asunto(s)
Regulación del Apetito/fisiología , Huesos/metabolismo , Lipocalina 2/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Huesos/citología , AMP Cíclico/metabolismo , Ingestión de Alimentos/fisiología , Femenino , Factor-23 de Crecimiento de Fibroblastos , Glucosa/metabolismo , Homeostasis , Hipotálamo/citología , Hipotálamo/metabolismo , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Masculino , Ratones , Neuronas/metabolismo , Obesidad/metabolismo , Osteoblastos/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Delgadez/metabolismo
8.
Carcinogenesis ; 42(2): 220-231, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32780107

RESUMEN

Prostate cancer is the second most common type of cancer and the second leading cause of cancer death in American men. RAD9 stabilizes the genome, but prostate cancer cells and tumors often have high quantities of the protein. Reduction of RAD9 level within prostate cancer cells decreases tumorigenicity of nude mouse xenographs and metastasis phenotypes in culture, indicating that RAD9 overproduction is essential for the disease. In prostate cancer DU145 cells, CpG hypermethylation in a transcription suppressor site of RAD9 intron 2 causes high-level gene expression. Herein, we demonstrate that DNA methyltransferases DNMT1 and DNMT3B are highly abundant in prostate cancer cells DU145, CWR22, LNCaP and PC-3; yet, these DNMTs bind primarily to the transcription suppressor in DU145, the only cells where methylation is critical for RAD9 regulation. For DU145 cells, DNMT1 or DNMT3B shRNA reduced RAD9 level and tumorigenicity, and RAD9 ectopic expression restored this latter activity in the DNMT knockdown cells. High levels of RAD9, DNMT1, DNMT3B and RAD9 transcription suppressor hypermethylation were significantly correlated in prostate tumors, and not in normal prostate tissues. Based on these results, we propose a novel model where RAD9 is regulated epigenetically by DNMT1 and DNMT3B, via targeted hypermethylation, and that consequent RAD9 overproduction promotes prostate tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Neoplasias de la Próstata/genética , Animales , Línea Celular Tumoral , Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Próstata/patología , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto , ADN Metiltransferasa 3B
9.
Thorax ; 76(4): 350-359, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33298583

RESUMEN

BACKGROUND: Identifying subtypes of acute respiratory failure survivors may facilitate patient selection for post-intensive care unit (ICU) follow-up clinics and trials. METHODS: We conducted a single-centre prospective cohort study of 185 acute respiratory failure survivors, aged ≥ 65 years. We applied latent class modelling to identify frailty subtypes using frailty phenotype and cognitive impairment measurements made during the week before hospital discharge. We used Fine-Gray competing risks survival regression to test associations between frailty subtypes and recovery, defined as returning to a basic Activities of Daily Living disability count less than or equal to the pre-hospitalisation count within 6 months. We characterised subtypes by pre-ICU frailty (Clinical Frailty Scale score ≥ 5), the post-ICU frailty phenotype, and serum inflammatory cytokines, hormones and exosome proteomics during the week before hospital discharge. RESULTS: We identified five frailty subtypes. The recovery rate decreased 49% across each subtype independent of age, sex, pre-existing disability, comorbidity and Acute Physiology and Chronic Health Evaluation II score (recovery rate ratio: 0.51, 95% CI 0.41 to 0.63). Post-ICU frailty phenotype prevalence increased across subtypes, but pre-ICU frailty prevalence did not. In the subtype with the slowest recovery, all had cognitive impairment. The three subtypes with the slowest recovery had higher interleukin-6 levels (p=0.03) and a higher prevalence of ≥ 2 deficiencies in insulin growth factor-1, dehydroepiandrostersone-sulfate, or free-testosterone (p=0.02). Exosome proteomics revealed impaired innate immunity in subtypes with slower recovery. CONCLUSIONS: Frailty subtypes varied by prehospitalisation frailty and cognitive impairment at hospital discharge. Subtypes with the slowest recovery were similarly characterised by greater systemic inflammation and more anabolic hormone deficiencies at hospital discharge.


Asunto(s)
Trastornos del Conocimiento/diagnóstico , Fragilidad/clasificación , Insuficiencia Respiratoria/fisiopatología , Actividades Cotidianas , Anciano , Anciano de 80 o más Años , Citocinas/sangre , Femenino , Hormonas/sangre , Hospitalización , Humanos , Unidades de Cuidados Intensivos , Análisis de Clases Latentes , Masculino , Alta del Paciente , Fenotipo , Proyectos Piloto , Estudios Prospectivos , Proteómica , Sobrevivientes
10.
Gastroenterology ; 159(2): 575-590, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32325086

RESUMEN

BACKGROUND & AIMS: Studies are needed to determine the mechanism by which Barrett's esophagus (BE) progresses to esophageal adenocarcinoma (EAC). Notch signaling maintains stem cells in the gastrointestinal tract and is dysregulated during carcinogenesis. We explored the relationship between Notch signaling and goblet cell maturation, a feature of BE, during EAC pathogenesis. METHODS: We measured goblet cell density and levels of Notch messenger RNAs in BE tissues from 164 patients, with and without dysplasia or EAC, enrolled in a multicenter study. We analyzed the effects of conditional expression of an activated form of NOTCH2 (pL2.Lgr5.N2IC), conditional deletion of NOTCH2 (pL2.Lgr5.N2fl/fl), or loss of nuclear factor κB (NF-κB) (pL2.Lgr5.p65fl/fl), in Lgr5+ (progenitor) cells in L2-IL1B mice (which overexpress interleukin 1 beta in esophagus and squamous forestomach and are used as a model of BE). We collected esophageal and stomach tissues and performed histology, immunohistochemistry, flow cytometry, transcriptome, and real-time polymerase chain reaction analyses. Cardia and forestomach tissues from mice were cultured as organoids and incubated with inhibitors of Notch or NF-kB. RESULTS: Progression of BE to EAC was associated with a significant reduction in goblet cell density comparing nondysplastic regions of tissues from patients; there was an inverse correlation between goblet cell density and levels of NOTCH3 and JAG2 messenger RNA. In mice, expression of the activated intracellular form of NOTCH2 in Lgr5+ cells reduced goblet-like cell maturation, increased crypt fission, and accelerated the development of tumors in the squamocolumnar junction. Mice with deletion of NOTCH2 from Lgr5+ cells had increased maturation of goblet-like cells, reduced crypt fission, and developed fewer tumors. Esophageal tissues from in pL2.Lgr5.N2IC mice had increased levels of RelA (which encodes the p65 unit of NF-κB) compared to tissues from L2-IL1B mice, and we found evidence of increased NF-κB activity in Lgr5+ cells. Esophageal tissues from pL2.Lgr5.p65fl/fl mice had lower inflammation and metaplasia scores than pL2.Lgr5.N2IC mice. In organoids derived from pL2-IL1B mice, the NF-κB inhibitor JSH-23 reduced cell survival and proliferation. CONCLUSIONS: Notch signaling contributes to activation of NF-κB and regulates differentiation of gastric cardia progenitor cells in a mouse model of BE. In human esophageal tissues, progression of BE to EAC was associated with reduced goblet cell density and increased levels of Notch expression. Strategies to block this pathway might be developed to prevent EAC in patients with BE.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Carcinogénesis/patología , Neoplasias Esofágicas/patología , Células Caliciformes/patología , Receptores Notch/metabolismo , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Anciano , Animales , Esófago de Barrett/diagnóstico , Esófago de Barrett/genética , Biopsia , Carcinogénesis/genética , Diferenciación Celular/genética , Estudios Transversales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Mucosa Esofágica/citología , Mucosa Esofágica/diagnóstico por imagen , Mucosa Esofágica/patología , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Esofagoscopía , Femenino , Mucosa Gástrica/citología , Mucosa Gástrica/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , FN-kappa B/metabolismo , Estudios Prospectivos , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores Notch/genética , Transducción de Señal
11.
Int J Cancer ; 145(10): 2754-2766, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31001805

RESUMEN

The main risk factor for esophageal dysplasia and adenocarcinoma (DAC) is Barrett's esophagus (BE), characterized by intestinal metaplasia. The critical genomic mechanisms that lead to progression of nondysplastic BE to DAC remain poorly understood and require analyses of longitudinal patient cohorts and high-resolution assays. We tested BE tissues from 74 patients, including 42 nonprogressors from two separate groups of 21 patients each and 32 progressors (16 in a longitudinal cohort before DAC/preprogression-BE and 16 with temporally concurrent but spatially separate DAC/concurrent-BE). We interrogated genome-wide somatic copy number alterations (SCNAs) at the exon level with high-resolution SNP arrays in DNA from formalin-fixed samples histologically confirmed as nondysplastic BE. The most frequent abnormalities were SCNAs involving FHIT exon 5, CDKN2A/B or both in 88% longitudinal BE progressors to DAC vs. 24% in both nonprogressor groups (p = 0.0004). Deletions in other genomic regions were found in 56% of preprogression-BE but only in one nonprogressor-BE (p = 0.0004). SCNAs involving FHIT exon 5 and CDKN2A/B were also frequently detected in BE temporally concurrent with DAC. TP53 losses were detected in concurrent-BE but not earlier in preprogression-BE tissues of patients who developed DAC. CDKN2A/p16 immunohistochemistry showed significant loss of expression in BE of progressors vs. nonprogressors, supporting the genomic data. Our data suggest a role for CDKN2A/B and FHIT in early progression of BE to dysplasia and adenocarcinoma that warrants future mechanistic research. Alterations in CDKN2A/B and FHIT by high-resolution assays may serve as biomarkers of increased risk of progression to DAC when detected in BE tissues.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/genética , Biomarcadores de Tumor/genética , Mucosa Esofágica/patología , Neoplasias Esofágicas/patología , Lesiones Precancerosas/genética , Ácido Anhídrido Hidrolasas/genética , Adulto , Anciano , Esófago de Barrett/patología , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Exones/genética , Femenino , Humanos , Hibridación Fluorescente in Situ , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Lesiones Precancerosas/patología , Proteína p53 Supresora de Tumor/genética
12.
Hepatology ; 67(6): 2414-2429, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29091291

RESUMEN

Fibrosis and cancer represent two major complications of chronic liver disease. MicroRNAs have been implicated in the development of fibrosis and cancer, thus constituting potential therapeutic targets. Here, we investigated the role of microRNA-21 (miR-21), a microRNA that has been implicated in the development of fibrosis in multiple organs and has also been suggested to act as an "oncomir." Accordingly, miR-21 was the microRNA that showed the strongest up-regulation in activated hepatic stellate cells (HSCs) in multiple models of fibrogenesis, with an 8-fold to 24-fold induction compared to quiescent HSCs. However, miR-21 antisense inhibition did not suppress the activation of murine or human HSCs in culture or in liver slices. Moreover, genetic deletion of miR-21 in two independently generated knockout mice or miR-21 antisense inhibition did not alter HSC activation or liver fibrosis in models of toxic and biliary liver injury. Despite a strong up-regulation of miR-21 in injury-associated hepatocellular carcinoma and in cholangiocarcinoma, miR-21 deletion or antisense inhibition did not reduce the development of liver tumors. As inhibition of the most up-regulated microRNA did not affect HSC activation, liver fibrosis, or fibrosis-associated liver cancer, we additionally tested the role of microRNAs in HSCs by HSC-specific Dicer deletion. Although Dicer deletion decreased microRNA expression in HSCs and altered the expression of select genes, it only exerted negligible effects on HSC activation and liver fibrosis. CONCLUSION: Genetic and pharmacologic manipulation of miR-21 does not inhibit the development of liver fibrosis and liver cancer. Moreover, suppression of microRNA synthesis does not significantly affect HSC phenotype and activation. (Hepatology 2018;67:2414-2429).


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/etiología , MicroARNs/fisiología , Ribonucleasa III/fisiología , Animales , Femenino , Humanos , Masculino , Ratones Noqueados
13.
Am J Physiol Renal Physiol ; 315(6): F1601-F1612, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30132346

RESUMEN

Diaphanous 1 (DIAPH1), a member of the formin family, binds to the cytoplasmic domain of the receptor for advanced glycation end products (RAGE) and is required for RAGE signal transduction. Experiments employing genetic overexpression or deletion of Ager (the gene encoding RAGE) or its pharmacological antagonism implicate RAGE in the pathogenesis of diabetes-associated nephropathy. We hypothesized that DIAPH1 contributes to pathological and functional derangements in the kidneys of diabetic mice. We show that DIAPH1 is expressed in the human and murine diabetic kidney, at least in part in the tubulointerstitium and glomerular epithelial cells or podocytes. To test the premise that DIAPH1 is linked to diabetes-associated derangements in the kidney, we rendered male mice globally devoid of Diaph1 ( Diaph1-/-) or wild-type controls (C57BL/6 background) diabetic with streptozotocin. Control mice received equal volumes of citrate buffer. After 6 mo of hyperglycemia, diabetic Diaph1-/- mice displayed significantly reduced mesangial sclerosis, podocyte effacement, glomerular basement thickening, and urinary albumin-to-creatinine ratio compared with diabetic mice expressing Diaph1. Analysis of whole kidney cortex revealed that deletion of Diaph1 in diabetic mice significantly reduced expression of genes linked to fibrosis and inflammation. In glomerular isolates, expression of two genes linked to podocyte stress, growth arrest-specific 1 ( Gas1) and cluster of differentiation 36 ( Cd36), was significantly attenuated in diabetic Diaph1-/- mice compared with controls, in parallel with significantly higher levels of nestin (Nes) mRNA, a podocyte marker. Collectively, these data implicate DIAPH1 in the pathogenesis of diabetes-associated nephropathy and suggest that the RAGE-DIAPH1 axis is a logical target for therapeutic intervention in this disorder.


Asunto(s)
Proteínas Portadoras/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/prevención & control , Riñón/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Albuminuria/patología , Albuminuria/prevención & control , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas Portadoras/genética , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Forminas , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Eliminación de Gen , Humanos , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nestina/genética , Nestina/metabolismo , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal , Estreptozocina
15.
J Cell Sci ; 126(Pt 17): 3927-38, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23788429

RESUMEN

In mitotic cells, RAD9A functions in repairing DNA double-strand breaks (DSBs) by homologous recombination and facilitates the process by cell cycle checkpoint control in response to DNA damage. DSBs occur naturally in the germline during meiosis but whether RAD9A participates in repairing such breaks is not known. In this study, we determined that RAD9A is indeed expressed in the male germ line with a peak of expression in late pachytene and diplotene stages, and the protein was found associated with the XY body. As complete loss of RAD9A is embryonic lethal, we constructed and characterized a mouse strain with Stra8-Cre driven germ cell-specific ablation of Rad9a beginning in undifferentiated spermatogonia in order to assess its role in spermatogenesis. Adult mutant male mice were infertile or sub-fertile due to massive loss of spermatogenic cells. The onset of this loss occurs during meiotic prophase, and there was an increase in the numbers of apoptotic spermatocytes as determined by TUNEL. Spermatocytes lacking RAD9A usually arrested in meiotic prophase, specifically in pachytene. The incidence of unrepaired DNA breaks increased, as detected by accumulation of γH2AX and DMC1 foci on the axes of autosomal chromosomes in pachytene spermatocytes. The DNA topoisomerase IIß-binding protein 1 (TOPBP1) was still localized to the sex body, albeit with lower intensity, suggesting that RAD9A may be dispensable for sex body formation. We therefore show for the first time that RAD9A is essential for male fertility and for repair of DNA DSBs during meiotic prophase I.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena , Fertilidad/genética , Profase Meiótica I/fisiología , Reparación del ADN por Recombinación/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis/genética , Proteínas Portadoras/metabolismo , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/biosíntesis , Histonas/biosíntesis , Histonas/metabolismo , Masculino , Profase Meiótica I/genética , Ratones , Ratones Transgénicos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfato , Eliminación de Secuencia/genética , Espermatocitos/citología , Espermatogénesis/genética , Testículo
17.
Gut ; 63(3): 385-94, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23729675

RESUMEN

OBJECTIVE: To investigate the role of obesity-associated inflammation and immune modulation in gastric carcinogenesis during Helicobacter-induced chronic gastric inflammation. DESIGN: C57BL/6 male mice were infected with H felis and placed on a high-fat diet (45% calories from fat). Study animals were analysed for gastric and adipose pathology, inflammatory markers in serum, stomach and adipose tissue, and immune responses in blood, spleen, stomach and adipose tissue. RESULTS: H felis-induced gastric carcinogenesis was accelerated in diet-induced obese mice compared with lean controls. Obesity increased bone marrow-derived immature myeloid cells in blood and gastric tissue of H felis-infected mice. Obesity also led to elevations in CD4 T cells, IL-17A, granulocyte macrophage colony-stimulating factor, phosphorylated STAT3 and prosurvival gene expression in gastric tissue of H felis-infected mice. Conversely, in adipose tissue of obese mice, H felis infection increased macrophage accumulation and expression of IL-6, C-C motif ligand 7 (CCL7) and leptin. Finally, the combination of obesity and gastric inflammation synergistically increased serum proinflammatory cytokines, including IL-6. CONCLUSIONS: Here, we have established a model to study the molecular mechanism by which obesity predisposes individuals to gastric cancer. In H felis-infected mice, obesity increased proinflammatory immune responses and accelerated gastric carcinogenesis. Interestingly, gastric inflammation augmented obesity-induced adipose inflammation and production of adipose-derived factors in obese, but not lean, mice. Our findings suggest that obesity accelerates Helicobacter-associated gastric cancer through cytokine-mediated cross-talk between inflamed gastric and adipose tissues, augmenting immune responses at both tissue sites, and thereby contributing to a protumorigenic gastric microenvironment.


Asunto(s)
Gastritis/complicaciones , Infecciones por Helicobacter/complicaciones , Helicobacter felis , Células Progenitoras Mieloides/fisiología , Obesidad/complicaciones , Neoplasias Gástricas/etiología , Células Th17/fisiología , Animales , Biomarcadores/metabolismo , Movimiento Celular , Citocinas/metabolismo , Dieta Alta en Grasa , Citometría de Flujo , Gastritis/diagnóstico , Gastritis/metabolismo , Gastritis/microbiología , Infecciones por Helicobacter/inmunología , Inflamación/complicaciones , Inflamación/metabolismo , Inflamación/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/inmunología , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/microbiología
18.
Gastroenterology ; 145(4): 820-30.e10, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23891976

RESUMEN

BACKGROUND & AIMS: Progastrin stimulates colonic mucosal proliferation and carcinogenesis through the cholecystokinin 2 receptor (CCK2R)-partly by increasing the number of colonic progenitor cells. However, little is known about the mechanisms by which progastrin stimulates colonic cell proliferation. We investigated the role of bone morphogenetic proteins (BMPs) in progastrin induction of colonic cell proliferation via CCK2R. METHODS: We performed microarray analysis to compare changes in gene expression in the colonic mucosa of mice that express a human progastrin transgene, gastrin knockout mice, and C57BL/6 mice (controls); the effects of progastrin were also determined on in vitro colonic crypt cultures from cholecystokinin 2 receptor knockout and wild-type mice. Human colorectal and gastric cancer cells that expressed CCK2R were incubated with progastrin or Bmp2; levels of ß-arrestin 1 and 2 were knocked down using small interfering RNAs. Cells were analyzed for progastrin binding, proliferation, changes in gene expression, and symmetric cell division. RESULTS: The BMP pathway was down-regulated in the colons of human progastrin mice compared with controls. Progastrin suppressed transcription of Bmp2 through a pathway that required CCK2R and was mediated by ß-arrestin 1 and 2. In mouse colonic epithelial cells, down-regulation of Bmp2 led to decreased phosphorylation of Smads1/5/8 and suppression of inhibitor of DNA binding 4. In human gastric and colorectal cancer cell lines, CCK2R was necessary and sufficient for progastrin binding and induction of proliferation; these effects were blocked when cells were incubated with recombinant Bmp2. Incubation with progastrin increased the number of CD44(+), bromodeoxyuridine+, and NUMB(+) cells, indicating an increase in symmetric divisions of putative cancer stem cells. CONCLUSIONS: Progastrin stimulates proliferation in colons of mice and cultured human cells via CCK2R- and ß-arrestin 1 and 2-dependent suppression of Bmp2 signaling. This process promotes symmetric cell division.


Asunto(s)
Arrestinas/fisiología , Proteína Morfogenética Ósea 2/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Gastrinas/farmacología , Precursores de Proteínas/farmacología , Receptor de Colecistoquinina B/fisiología , Animales , Proteína Morfogenética Ósea 2/fisiología , Colon/citología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Células Madre/efectos de los fármacos , beta-Arrestina 1 , beta-Arrestinas
19.
Gastroenterology ; 144(1): 155-66, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23041326

RESUMEN

BACKGROUND & AIMS: Interleukin (IL)-8 has an important role in initiating inflammation in humans, attracting immune cells such as neutrophils through their receptors CXCR1 and CXCR2. IL-8 has been proposed to contribute to chronic inflammation and cancer. However, mice do not have the IL-8 gene, so human cancer cell lines and xenograft studies have been used to study the role of IL-8 in colon and gastric carcinogenesis. We generated mice that carry a bacterial artificial chromosome that encompasses the entire human IL-8 gene, including its regulatory elements (IL-8Tg mice). METHODS: We studied the effects of IL-8 expression in APCmin(+/-) mice and IL-8Tg mice given azoxymethane and dextran sodium sulfate (DSS). We also examined the effects of IL-8 expression in gastric cancer in INS-GAS mice that overexpress gastrin and IL-8Tg mice infected with Helicobacter felis. RESULTS: In IL-8Tg mice, expression of human IL-8 was controlled by its own regulatory elements, with virtually no messenger RNA or protein detectable under basal conditions. IL-8 was strongly up-regulated on systemic or local inflammatory stimulation, increasing mobilization of immature CD11b(+)Gr-1(+) myeloid cells (IMCs) with thioglycolate-induced peritonitis, DSS-induced colitis, and H. felis-induced gastritis. IL-8 was increased in colorectal tumors from patients and IL-8Tg mice compared with nontumor tissues. IL-8Tg mice developed more tumors than wild-type mice following administration of azoxymethane and DSS. Expression of IL-8 increased tumorigenesis in APCmin(+/-) mice compared with APCmin(+/-) mice that lack IL-8; this was associated with increased numbers of IMCs and angiogenesis in the tumors. CONCLUSIONS: IL-8 contributes to gastrointestinal carcinogenesis by mobilizing IMCs and might be a therapeutic target for gastrointestinal cancers.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Colitis/metabolismo , Neoplasias del Colon/metabolismo , Gastritis/metabolismo , Interleucina-8/metabolismo , Células Mieloides/efectos de los fármacos , ARN Mensajero/metabolismo , Animales , Azoximetano , Línea Celular Tumoral , Colitis/inducido químicamente , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Células Dendríticas/metabolismo , Sulfato de Dextran , Gastritis/microbiología , Infecciones por Helicobacter/complicaciones , Helicobacter felis , Humanos , Interleucina-8/genética , Interleucina-8/farmacología , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Células Mieloides/metabolismo , Cultivo Primario de Células , Carga Tumoral , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA