Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 158(4): 705-721, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126780

RESUMEN

Acquisition of the intestinal microbiota begins at birth, and a stable microbial community develops from a succession of key organisms. Disruption of the microbiota during maturation by low-dose antibiotic exposure can alter host metabolism and adiposity. We now show that low-dose penicillin (LDP), delivered from birth, induces metabolic alterations and affects ileal expression of genes involved in immunity. LDP that is limited to early life transiently perturbs the microbiota, which is sufficient to induce sustained effects on body composition, indicating that microbiota interactions in infancy may be critical determinants of long-term host metabolic effects. In addition, LDP enhances the effect of high-fat diet induced obesity. The growth promotion phenotype is transferrable to germ-free hosts by LDP-selected microbiota, showing that the altered microbiota, not antibiotics per se, play a causal role. These studies characterize important variables in early-life microbe-host metabolic interaction and identify several taxa consistently linked with metabolic alterations. PAPERCLIP:


Asunto(s)
Antibacterianos/administración & dosificación , Modelos Animales de Enfermedad , Intestinos/microbiología , Microbiota , Obesidad/microbiología , Penicilinas/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/metabolismo , Femenino , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Obesidad/metabolismo
2.
Toxicol Pathol ; 51(7-8): 414-431, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38380881

RESUMEN

Biotherapeutic modalities such as cell therapies, gene therapies, nucleic acids, and proteins are increasingly investigated as disease-modifying treatments for severe and life-threatening neurodegenerative disorders. Such diverse bio-derived test articles are fraught with unique and often unpredictable biological consequences, while guidance regarding nonclinical experimental design, neuropathology evaluation, and interpretation is often limited. This paper summarizes key messages offered during a half-day continuing education course on toxicologic neuropathology of neuro-targeted biotherapeutics. Topics included fundamental neurobiology concepts, pharmacology, frequent toxicological findings, and their interpretation including adversity decisions. Covered biotherapeutic classes included cell therapies, gene editing and gene therapy vectors, nucleic acids, and proteins. If agents are administered directly into the central nervous system, initial screening using hematoxylin and eosin (H&E)-stained sections of currently recommended neural organs (brain [7 levels], spinal cord [3 levels], and sciatic nerve) may need to expand to include other components (e.g., more brain levels, ganglia, and/or additional nerves) and/or special neurohistological procedures to characterize possible neural effects (e.g., cell type-specific markers for reactive glial cells). Scientists who evaluate the safety of novel biologics will find this paper to be a practical reference for preclinical safety testing and risk assessment.


Asunto(s)
Neuropatología , Ácidos Nucleicos , Encéfalo , Médula Espinal , Nervio Ciático
3.
Immunity ; 36(5): 742-54, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22503542

RESUMEN

In vitro data suggest that a subgroup of NLR proteins, including NLRP12, inhibits the transcription factor NF-κB, although physiologic and disease-relevant evidence is largely missing. Dysregulated NF-κB activity is associated with colonic inflammation and cancer, and we found Nlrp12(-/-) mice were highly susceptible to colitis and colitis-associated colon cancer. Polyps isolated from Nlrp12(-/-) mice showed elevated noncanonical NF-κB activation and increased expression of target genes that were associated with cancer, including Cxcl13 and Cxcl12. NLRP12 negatively regulated ERK and AKT signaling pathways in affected tumor tissues. Both hematopoietic- and nonhematopoietic-derived NLRP12 contributed to inflammation, but the latter dominantly contributed to tumorigenesis. The noncanonical NF-κB pathway was regulated upon degradation of TRAF3 and activation of NIK. NLRP12 interacted with both NIK and TRAF3, and Nlrp12(-/-) cells have constitutively elevated NIK, p100 processing to p52 and reduced TRAF3. Thus, NLRP12 is a checkpoint of noncanonical NF-κB, inflammation, and tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/patología , Colitis/metabolismo , Colitis/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , FN-kappa B/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Colitis/genética , Neoplasias del Colon/genética , Progresión de la Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Quinasa de Factor Nuclear kappa B
4.
Am J Pathol ; 188(7): 1510-1516, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29684361

RESUMEN

The new paradigm of mutations in chromatin-modifying genes as driver events in the development of cancers has proved challenging to resolve the complex influences over disease phenotypes. In particular, impaired activities of members of the SWI/SNF chromatin remodeling complex have appeared in an increasing variety of tumors. Mutations in SNF5, a member of this ubiquitously expressed complex, arise in almost all cases of malignant rhabdoid tumor in the absence of additional genetic alterations. Therefore, we studied how activation of additional oncogenic pathways might shift the phenotype of disease driven by SNF5 loss. With the use of a genetically engineered mouse model, we examined the effects of a hypomorphic Vhl2B allele on disease phenotype, with a modest up-regulation of the hypoxia response pathway. Snf5+/-;Vhl2B/+ mice did not demonstrate a substantial difference in overall survival or a change in malignant rhabdoid tumor development. However, a high percentage of female mice showed complex hemorrhagic ovarian cysts, a phenotype rarely found in either parental mouse strain. These lesions also showed mosaic expression of SNF5 by immunohistochemistry. Therefore, our studies implicate that modest changes in angiogenic regulation interact with perturbations of SWI/SNF complex activity to modulate disease phenotypes.


Asunto(s)
Hemorragia/patología , Mutación , Quistes Ováricos/patología , Proteína SMARCB1/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Animales , Femenino , Hemorragia/etiología , Hemorragia/metabolismo , Ratones , Ratones Noqueados , Quistes Ováricos/etiología , Quistes Ováricos/metabolismo , Fenotipo
5.
Gene Expr ; 19(1): 61-67, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30092856

RESUMEN

Inbred mice are the most popular animals used for in vivo liver research. These mice are genetically defined, readily available, less expensive to maintain than larger animals, and enjoy a broad array of commercial reagents for scientific characterization. C57BL/6 mice are the most commonly used strain. However, other strains discussed, including BALB/c, C3H, A/J, and FVB/N, may be better suited to a particular disease model or line of investigation. Understanding the phenotypes of different inbred mouse strains facilitates informed decision making during experimental design. Model systems influenced by strain-dependent phenotype include tissue regeneration, drug-induced liver injury (DILI; e.g., acetaminophen), fibrosis (e.g., carbon tetrachloride, CCl4), Fas-induced apoptosis, cholestasis, alcohol-induced liver disease and cirrhosis, nonalcoholic fatty liver disease and steatohepatitis (NAFLD/NASH), and hepatocellular carcinoma (HCC). Thoughtful consideration of the strengths and weaknesses of each inbred strain in a given model system will lead to more robust data and a clearer understanding of translational relevance to human liver disease.


Asunto(s)
Modelos Animales de Enfermedad , Hepatopatías/metabolismo , Ratones Endogámicos/fisiología , Animales , Apoptosis , Carcinoma Hepatocelular/patología , Fibrosis/patología , Genotipo , Humanos , Hígado/patología , Cirrosis Hepática/patología , Hepatopatías/fisiopatología , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos/genética , Ratones Endogámicos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Fenotipo
6.
Proc Natl Acad Sci U S A ; 111(31): 11455-60, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25049387

RESUMEN

Women are more resistant to hepatocellular carcinoma (HCC) than men despite equal exposure to major risk factors, such as hepatitis B or C virus infection. Female resistance is hormone-dependent, as evidenced by the sharp increase in HCC incidence in postmenopausal women who do not take hormone replacement therapy. In rodent models sex-dimorphic HCC phenotypes are pituitary-dependent, suggesting that sex hormones act via the gonadal-hypophyseal axis. We found that the estrogen-responsive pituitary hormone prolactin (PRL), signaling through hepatocyte-predominant short-form prolactin receptors (PRLR-S), constrained TNF receptor-associated factor (TRAF)-dependent innate immune responses invoked by IL-1ß, TNF-α, and LPS/Toll-like receptor 4 (TLR4), but not TRIF-dependent poly(I:C)/TLR3. PRL ubiquitinated and accelerated poststimulatory decay of a "trafasome" comprised of IRAK1, TRAF6, and MAP3K proteins, abrogating downstream activation of c-Myc-interacting pathways, including PI3K/AKT, mTORC1, p38 MAPK, and NF-κB. Consistent with this finding, we documented exaggerated male liver responses to immune stimuli in mice and humans. Tumor promotion through, but regulation above, the level of c-Myc was demonstrated by sex-independent HCC eruption in Alb-Myc transgenic mice. PRL deficiency accelerated liver carcinogenesis in Prl(-/-) mice of both sexes. Conversely, pharmacologic PRL mobilization using the dopamine D2 receptor antagonist domperidone prevented HCC in tumor-prone C3H/HeN males. Viewed together, our results demonstrate that PRL constrains tumor-promoting liver inflammation by inhibiting MAP3K-dependent activation of c-Myc at the level of the trafasome. PRL-targeted therapy may hold promise for reducing the burden of liver cancer in high-risk men and women.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/prevención & control , Inmunidad Innata , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/prevención & control , Prolactina/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/metabolismo , Adulto , Animales , Carcinogénesis/patología , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Domperidona/farmacología , Domperidona/uso terapéutico , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/patología , Interleucina-1beta/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Masculino , Ratones , Modelos Biológicos , FN-kappa B/metabolismo , Prolactina/deficiencia , Prolactina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Prolactina/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Microambiente Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas ras/metabolismo
7.
Infect Immun ; 84(1): 138-48, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26483408

RESUMEN

Infection with enterohemorrhagic Escherichia coli (EHEC) can result in severe disease, including hemorrhagic colitis and the hemolytic uremic syndrome. Shiga toxins (Stx) are the key EHEC virulence determinant contributing to severe disease. Despite inhibiting protein synthesis, Shiga toxins paradoxically induce the expression of proinflammatory cytokines from various cell types in vitro, including intestinal epithelial cells (IECs). This effect is mediated in large part by the ribotoxic stress response (RSR). The Shiga toxin-induced RSR is known to involve the activation of the stress-activated protein kinases (SAPKs) p38 and JNK. In some cell types, Stx also can induce the classical mitogen-activated protein kinases (MAPKs) or ERK1/2, but the mechanism(s) by which this activation occurs is unknown. In this study, we investigated the mechanism by which Stx activates ERK1/2s in IECs and the contribution of ERK1/2 activation to interleukin-8 (IL-8) expression. We demonstrate that Stx1 activates ERK1/2 in a biphasic manner: the first phase occurs in response to StxB1 subunit, while the second phase requires StxA1 subunit activity. We show that the A subunit-dependent ERK1/2 activation is mediated through ZAK-dependent signaling, and inhibition of ERK1/2 activation via the MEK1/2 inhibitors U0126 and PD98059 results in decreased Stx1-mediated IL-8 mRNA. Finally, we demonstrate that ERK1/2 are activated in vivo in the colon of Stx2-intoxicated infant rabbits, a model in which Stx2 induces a primarily neutrophilic inflammatory response. Together, our data support a role for ERK1/2 activation in the development of Stx-mediated intestinal inflammation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Butadienos/farmacología , Línea Celular , Escherichia coli Enterohemorrágica/patogenicidad , Activación Enzimática , Células Epiteliales/inmunología , Infecciones por Escherichia coli/microbiología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Flavonoides/farmacología , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Células HEK293 , Síndrome Hemolítico-Urémico/microbiología , Humanos , Inflamación/inmunología , Interleucina-8/biosíntesis , Interleucina-8/genética , Mucosa Intestinal/inmunología , Quinasas Quinasa Quinasa PAM , Nitrilos/farmacología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Conejos
8.
Mol Ther ; 21(8): 1559-69, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23774791

RESUMEN

There is an urgent need for new therapeutics for the treatment of aggressive and metastatic refractory human non-small-cell lung cancer (NSCLC). Antiangiogenesis therapy and chemotherapy are the two major treatment options. Unfortunately, both types of therapies when used individually have their disadvantages. Integrating antiangiogenesis therapy with chemotherapy is expected to target the tumor's vascular endothelial cells and the tumor cells simultaneously. In this study, we coformulated Vascular endothelial growth factor (VEGF) siRNA targeting VEGFs and gemcitabine monophosphate (GMP) into a single cell-specific, targeted lipid/calcium/phosphate (LCP) nanoparticle formulation. Antitumor effect of the combination therapy using LCP loaded with both VEGF siRNA and GMP was evaluated in both subcutaneous and orthotopic xenograft models of NSCLC with systemic administration. The improved therapeutic response, as compared with either VEGF siRNA or GMP therapy alone, was supported by the observation of 30-40% induction of tumor cell apoptosis, eightfold reduction of tumor cell proliferation and significant decrease of tumor microvessel density (MVD). The combination therapy led to dramatic inhibition of tumor growth, with little in vivo toxicity. In addition, the current studies demonstrated the possibility of incorporating multiple nucleic acid molecules and phosphorylated small-molecule drugs, targeting to different pathways, into a single nanoparticle formulation for profound therapeutic effect.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Desoxicitidina/análogos & derivados , Neoplasias Pulmonares/genética , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Nanopartículas/química , Nanopartículas/toxicidad , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
10.
Int J Cancer ; 132(12): 2767-77, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23197309

RESUMEN

Malignant rhabdoid tumors (MRTs) are rare, aggressive cancers occuring in young children primarily through inactivation of the SNF5(INI1, SMARCB1) tumor suppressor gene. We and others have demonstrated that mice heterozygous for a Snf5 null allele develop MRTs with partial penetrance. We have also shown that Snf5(+/-) mice that lack expression of the pRb family, due to TgT121 transgene expression, develop MRTs with increased penetrance and decreased latency. Here, we report that altering the genetic background has substantial effects upon MRT development in Snf5(+/--) and TgT121 ;Snf5(+/-) mice, with a mixed F1 background resulting in increased latency and the appearance of brain tumors. We also report the establishment of the first mouse MRT cell lines that recapitulate many features of their human counterparts. Our studies provide further insight into the genetic influences on MRT development as well as provide valuable new cell culture and genetically engineered mouse models for the study of CNS-MRT etiology.


Asunto(s)
Línea Celular Tumoral , Tumor Rabdoide/genética , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Neoplasias del Plexo Coroideo/genética , Neoplasias del Plexo Coroideo/patología , Proteínas Cromosómicas no Histona/genética , Modelos Animales de Enfermedad , Genotipo , Humanos , Cariotipo , Ratones , Ratones Transgénicos , Fenotipo , Tumor Rabdoide/patología , Proteína SMARCB1
11.
Blood ; 117(12): 3257-67, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21245485

RESUMEN

Previous authors have suggested that tumor suppressor expression promotes aging while preventing cancer, but direct experimental support for this cancer-aging hypothesis has been elusive. Here, by using somatic, tissue-specific inactivation of the p16(INK4a) tumor suppressor in murine T- or B-lymphoid progenitors, we report that ablation of p16(INK4a) can either rescue aging or promote cancer in a lineage-specific manner. Deletion of p16(INK4a) in the T lineage ameliorated several aging phenotypes, including thymic involution, decreased production of naive T cells, reduction in homeostatic T-cell proliferation, and attenuation of antigen-specific immune responses. Increased T-cell neoplasia was not observed with somatic p16(INK4a) inactivation in T cells. In contrast, B lineage-specific ablation of p16(INK4a) was associated with a markedly increased incidence of systemic, high-grade B-cell neoplasms, which limited studies of the effects of somatic p16(INK4a) ablation on B-cell aging. Together, these data show that expression of p16(INK4a) can promote aging and prevent cancer in related lymphoid progeny of a common stem cell.


Asunto(s)
Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Linfocitos/metabolismo , Linfocitos/fisiología , Neoplasias/genética , Animales , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Proliferación Celular , Transformación Celular Neoplásica/genética , Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Eliminación de Gen , Expresión Génica/fisiología , Integrasas/genética , Integrasas/fisiología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Linfocitos/patología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/prevención & control , Especificidad de Órganos/genética
12.
Am J Pathol ; 179(6): 2855-65, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21967816

RESUMEN

Insulin resistance is a defining feature of metabolic syndrome and type 2 diabetes mellitus but also may occur independently of these conditions. Nonalcoholic fatty liver disease (NAFLD), the hepatic manifestation of these disorders, increases the risk of hepatocellular carcinoma (HCC). However, mechanisms linking hyperinsulinemia to NAFLD and HCC require clarification. We describe a novel model of primary insulin resistance and HCC with strong parent-of-origin effects. Male AB6F1 (A/JCr dam × C57BL/6 sire) but not B6AF1 (B6 dam × A/J sire) mice developed spontaneous insulin resistance, NAFLD, and HCC without obesity or diabetes. A survey of mitochondrial, imprinted, and sex-linked traits revealed modest associations with X-linked genes. However, a diet-induced obesity study, including B6.A chromosome substitution-strain (consomic) mice, showed no segregation by sex chromosome. Thus, parent-of-origin effects were specified within the autosomal genome. Next, we interrogated mechanisms of insulin-associated hepatocarcinogenesis. Steatotic hepatocytes exhibited adipogenic transition characterized by vacuolar metaplasia and up-regulation of vimentin, adipsin, fatty acid translocase (CD36), peroxisome proliferator-activated receptor-γ, and related products. This profile was largely recapitulated in insulin-supplemented primary mouse hepatocyte cultures. Importantly, pyruvate kinase M2, a fetal anabolic enzyme implicated in the Warburg effect, was activated by insulin in vivo and in vitro. Thus, our study reveals parent-of-origin effects in heritable insulin resistance, implicating adipogenic transition with acquired anabolic metabolism in the progression from NAFLD to HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Epistasis Genética/genética , Impresión Genómica/genética , Resistencia a la Insulina/genética , Neoplasias Hepáticas/genética , Adipocitos/patología , Animales , Transformación Celular Neoplásica/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/genética , Femenino , Hepatocitos/patología , Hiperinsulinismo/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Enfermedad del Hígado Graso no Alcohólico
13.
Nat Biotechnol ; 40(10): 1500-1508, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35654979

RESUMEN

Therapeutics based on short interfering RNAs (siRNAs) delivered to hepatocytes have been approved, but new delivery solutions are needed to target additional organs. Here we show that conjugation of 2'-O-hexadecyl (C16) to siRNAs enables safe, potent and durable silencing in the central nervous system (CNS), eye and lung in rodents and non-human primates with broad cell type specificity. We show that intrathecally or intracerebroventricularly delivered C16-siRNAs were active across CNS regions and cell types, with sustained RNA interference (RNAi) activity for at least 3 months. Similarly, intravitreal administration to the eye or intranasal administration to the lung resulted in a potent and durable knockdown. The preclinical efficacy of an siRNA targeting the amyloid precursor protein was evaluated through intracerebroventricular dosing in a mouse model of Alzheimer's disease, resulting in amelioration of physiological and behavioral deficits. Altogether, C16 conjugation of siRNAs has the potential for safe therapeutic silencing of target genes outside the liver with infrequent dosing.


Asunto(s)
Precursor de Proteína beta-Amiloide , Tratamiento con ARN de Interferencia , Animales , Ratones , Primates/genética , Primates/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico
14.
Carcinogenesis ; 32(8): 1244-50, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21565825

RESUMEN

Helicobacter pylori-associated gastric cancer is male predominant and animal studies suggest that sex hormones influence gastric carcinogenesis. We investigated the effects of 17ß-estradiol (E2) or castration on H.pylori-induced gastritis in male INS-GAS/FVB/N (Tg(Ins1-GAS)1Sbr) mice. Comparisons were made to previously evaluated sham (n = 8) and H.pylori-infected (n = 8), intact male INS-GAS mice which had developed severe corpus gastritis accompanied by atrophy, hyperplasia, intestinal metaplasia and dysplasia of the epithelium within 16 weeks postinfection (all P < 0.01). Castration at 8 weeks of age had no sparing effect on lesions in uninfected (n = 5) or H.pylori-infected mice (n = 7) but all lesion subfeatures were attenuated by E2 in H.pylori-infected mice (n = 7) (P < 0.001). Notably, inflammation was not reduced but glandular atrophy, hyperplasia, intestinal metaplasia and dysplasia were also less severe in uninfected, E2-treated mice (n = 7) (P < 0.01). Attenuation of gastric lesions by E2 was associated with lower messenger RNA (mRNA) expression of interferon (IFN)-γ (P < 0.05) and interleukin (IL)-1ß (P < 0.004), and higher IL-10 (P < 0.02) as well as decreased numbers of Foxp3(+) regulatory T cells when compared with infected intact males. Infected E2-treated mice also developed higher Th2-associated anti-H.pylori IgG1 responses (P < 0.05) and significantly lower Ki-67 indices of epithelial proliferation (P < 0.05). E2 elevated expression of mRNA for Foxp3 (P < 0.0001) and IL-10 (P < 0.01), and decreased IL-1ß (P < 0.01) in uninfected, intact male mice compared with controls. Therefore, estrogen supplementation, but not castration, attenuated gastric lesions in H.pylori-infected male INS-GAS mice and to a lesser extent in uninfected mice, potentially by enhancing IL-10 function, which in turn decreased IFN-γ and IL-1ß responses induced by H.pylori.


Asunto(s)
Estradiol/uso terapéutico , Gastritis/prevención & control , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/patogenicidad , Neoplasias Gástricas/patología , Neoplasias Gástricas/prevención & control , Animales , Castración , Ensayo de Inmunoadsorción Enzimática , Estrógenos/uso terapéutico , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Gastritis/etiología , Gastritis/patología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/patología , Técnicas para Inmunoenzimas , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Neoplasias Intestinales/etiología , Neoplasias Intestinales/patología , Neoplasias Intestinales/prevención & control , Masculino , Metaplasia/etiología , Metaplasia/patología , Metaplasia/prevención & control , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estómago/inmunología , Estómago/patología , Neoplasias Gástricas/etiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/microbiología , Linfocitos T Reguladores/patología , Testosterona/sangre
15.
J Clin Invest ; 118(7): 2516-25, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18521188

RESUMEN

Chronic inflammation increases cancer risk. While it is clear that cell signaling elicited by inflammatory cytokines promotes tumor development, the impact of DNA damage production resulting from inflammation-associated reactive oxygen and nitrogen species (RONS) on tumor development has not been directly tested. RONS induce DNA damage that can be recognized by alkyladenine DNA glycosylase (Aag) to initiate base excision repair. Using a mouse model of episodic inflammatory bowel disease by repeated administration of dextran sulfate sodium in the drinking water, we show that Aag-mediated DNA repair prevents colonic epithelial damage and reduces the severity of dextran sulfate sodium-induced colon tumorigenesis. Importantly, DNA base lesions expected to be induced by RONS and recognized by Aag accumulated to higher levels in Aag-deficient animals following stimulation of colonic inflammation. Finally, as a test of the generality of this effect we show that Aag-deficient animals display more severe gastric lesions that are precursors of gastric cancer after chronic infection with Helicobacter pylori. These data demonstrate that the repair of DNA lesions formed by RONS during chronic inflammation is important for protection against colon carcinogenesis.


Asunto(s)
Colon/metabolismo , Neoplasias del Colon/etiología , Daño del ADN , ADN Glicosilasas/genética , Enfermedades Inflamatorias del Intestino/complicaciones , Animales , Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , ADN Glicosilasas/deficiencia , Reparación del ADN , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/crecimiento & desarrollo , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Purinas/análisis , Purinas/metabolismo , Pirimidinas/análisis , Pirimidinas/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/patología , Estómago/microbiología , Estómago/patología , beta Catenina/genética
16.
Gastroenterology ; 138(3): 1022-34.e1-10, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19962981

RESUMEN

BACKGROUND & AIMS: The nuclear factor kappaB (NF-kappaB)/IkappaB-kinase-beta (IKKbeta) pathway has been shown to represent a key link between inflammation and cancer, inducing pro-inflammatory cytokines in myeloid cells and anti-apoptotic pathways in epithelial cells. However, the role of NF-kappaB pathway in gastric carcinogenesis and injury has not been well-defined. We derived mice with a conditional knockout of IKKbeta in gastric epithelial cells (GECs) and myeloid cells, and examined responses to ionizing radiation (IR) and Helicobacter felis infection. METHODS: Ikkbeta(Deltastom) mice were generated by crossing Foxa3-Cre mice to Ikkbeta(F/F) mice. Cellular stress was induced with IR and H felis in Ikkbeta(Deltastom), Ikkbeta(F/F), and cis-NF-kappaB-enhanced green fluorescent protein (GFP) reporter mice. Gastric histopathology, apoptosis, proliferation, necrosis, reactive oxygen species, and expression of cytokines, chemokines, and anti-apoptotic genes were assessed. The role of myeloid IKKbeta in these models was studied by crosses with LysM-Cre mice. RESULTS: NF-kappaB activity was upregulated in myeloid cells with acute H felis infection, but in GECs by IR or long-term H felis infection during progression to dysplasia. Deletion of IKKbeta in GECs led to increased apoptosis, reactive oxygen species, and cellular necrosis, and resulted in up-regulation of interleukin-1alpha and down-regulation of anti-apoptotic genes. Loss of IKKbeta in GECs resulted in worse inflammation and more rapid progression to gastric preneoplasia, while loss of IKKbeta in myeloid cells inhibited development of gastric atrophy. CONCLUSIONS: The loss of IKKbeta/NF-kappaB signaling in GECs results in increased apoptosis and necrosis in response to cellular stress, and accelerated development of dysplasia by Helicobacter infection.


Asunto(s)
Apoptosis , Proliferación Celular , Mucosa Gástrica/enzimología , Infecciones por Helicobacter/enzimología , Helicobacter felis/patogenicidad , Quinasa I-kappa B/deficiencia , Lesiones Precancerosas/enzimología , Neoplasias Gástricas/enzimología , Animales , Apoptosis/efectos de la radiación , Proteínas Reguladoras de la Apoptosis/metabolismo , Atrofia , Proliferación Celular/efectos de la radiación , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Mucosa Gástrica/efectos de la radiación , Genes Reporteros , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Factor Nuclear 3-gamma del Hepatocito/genética , Quinasa I-kappa B/genética , Mediadores de Inflamación/metabolismo , Integrasas/genética , Interleucina-1alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/enzimología , Células Mieloides/microbiología , Células Mieloides/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Necrosis , Estrés Oxidativo , Lesiones Precancerosas/genética , Lesiones Precancerosas/microbiología , Lesiones Precancerosas/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Factores de Tiempo
17.
Am J Pathol ; 175(1): 365-75, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19556515

RESUMEN

We have previously described a synergistic interaction between hypergastrinemia and Helicobacter felis infection on gastric corpus carcinogenesis in FVB/N mice housed under specific-pathogen-free conditions. However, gastrin-deficient (GAS-KO) mice on a mixed C57BL/6/129Sv genetic background maintained in conventional housing were reported to develop spontaneous gastric antral tumors. Therefore, we investigated the role of gastrin in Helicobacter-associated gastric carcinogenesis in H. felis-infected mice on a uniform C57BL/6 background housed in specific-pathogen-free conditions. Hypergastrinemic transgenic (INS-GAS) mice, GAS-KO mice, and C57BL/6 wild-type mice were infected with H. felis for either 12 or 18 months. At 12 months postinfection, INS-GAS mice had mild corpus dysplasia, while B6 wild-type mice had either severe gastritis or metaplasia, and GAS-KO mice had only mild to moderate gastritis. At 18 months postinfection, both INS-GAS and B6 wild-type mice had both severe atrophic gastritis and corpus dysplasia, while GAS-KO mice had severe gastritis with mild gastric atrophy, but no corpus dysplasia. In contrast, both GAS-KO and B6 wild-type mice had mild to moderate antral dysplasia, while INS-GAS mice did not. H. felis antral colonization remained stable over time among the three groups of mice. These results point to a distinct effect of gastrin on carcinogenesis of both the gastric corpus and antrum, suggesting that gastrin is an essential cofactor for gastric corpus carcinogenesis in C57BL/6 mice.


Asunto(s)
Gastrinas/metabolismo , Infecciones por Helicobacter/metabolismo , Lesiones Precancerosas/metabolismo , Neoplasias Gástricas/metabolismo , Animales , Citocinas/biosíntesis , Citocinas/inmunología , Femenino , Gastrinas/genética , Gastritis/genética , Gastritis/inmunología , Gastritis/metabolismo , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/inmunología , Helicobacter felis , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Linfocitos T Colaboradores-Inductores/inmunología
18.
Hepatology ; 49(6): 1935-43, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19475691

RESUMEN

UNLABELLED: Activation of the insulin (IN)/insulin receptor substrate-1 (IRS-1)/mitogen-associated protein kinase (MAPK) and the Wnt/beta-catenin signaling cascades occurs frequently in hepatocellular carcinoma (HCC) associated with persistent viral infection. The aims of this study were to provide a chronic proliferative stimulus through IRS-1 in the context of hepatitis Bx (HBx) protein expression in transgenic mice and determine if constitutive expression of these genes is sufficient to cause hepatocyte dysplasia and cellular transformation. We generated transgenic mice in which the HBx (ATX), IRS-1, or both (ATX+/IRS-1) genes were expressed under a liver-specific promoter. We also assessed histology and oxidative damage as well as up-regulation of molecules related to these signal transduction cascades in the liver by quantitative reverse-transcriptase polymerase chain reaction. Whereas mice with a single transgene (ATX or IRS-1) did not develop tumors, ATX+/IRS-1+ double transgenic livers had increased frequency of hepatocellular dysplasia and developed HCC. All three transgenic lines had significantly increased insulin growth factor 1 (IGF-1), Wnt 1 and Wnt 3 mRNA levels, and evidence of DNA damage and oxidative stress. The ATX+/IRS+ double transgenic mice were distinguished by having the highest level of activation of Wnt 3 and Frizzled 7 and selectively increased expression of IGF-II, proliferating cell nuclear antigen, and aspartyl-(asparaginyl)-beta-hydroxylase, a gene associated with increased cell migration. CONCLUSION: These results suggest that continued expression of the ATX or IRS-1 transgenes can contribute to hepatocyte transformation but are not sufficient to trigger neoplastic changes in the liver. However, dual expression that activates both the IN/IRS-1/MAPK and Wnt/beta-catenin cascades is sufficient to cause dysplasia and HCC in a previously normal liver.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Sustrato del Receptor de Insulina/genética , Neoplasias Hepáticas/genética , Lesiones Precancerosas/genética , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales/genética , Animales , Ratones , Ratones Transgénicos
19.
Stem Cells ; 27(9): 2301-11, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19591219

RESUMEN

Bone marrow mesenchymal stem cells (MSCs) have been shown to have immune modulatory effects. Despite efforts to identify these cells in vivo, to date, MSCs have been defined mainly by their in vitro cell characteristics. Here, we show that Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells make up approximately 0.5%-1% of murine whole bone marrow cells and yield nearly an equal amount of fibroblastic colony-forming units (CFU-F) as whole bone marrow. After transplantation into lethally irradiated recipients, Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells engrafted in the bone marrow long-term and demonstrated characteristics of MSCs, including capacity to differentiate into osteoblasts and adipocytes. To examine whether Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells have immune modulatory effects, in vitro coculture with activated CD4+ T-cells resulted in decreased Th17 cell differentiation by Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells. Furthermore, serial infusions with Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells reduced the progression to low-grade gastric dysplasia in mice infected with chronic Helicobacter felis (p = .038). This correlated with reduced gastric interleukin (IL)-17F, IL-22, and ROR-gammat gene expression in responding mice (p < .05). These data suggest that bone marrow derived Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells have characteristics of MSCs and reduce progression of early gastric tumorigenesis induced by chronic H. felis infection. The prevention of dysplastic changes may occur through inhibition of Th17-dependent pathways.


Asunto(s)
Células de la Médula Ósea/fisiología , Helicobacter felis/fisiología , Células Madre Mesenquimatosas/fisiología , Neoplasias Gástricas/prevención & control , Neoplasias Gástricas/terapia , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Citometría de Flujo , Infecciones por Helicobacter/patología , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Células Madre , Neoplasias Gástricas/microbiología
20.
Infect Immun ; 77(5): 2147-58, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19223483

RESUMEN

Because coinfections can alter helicobacter gastritis, we investigated whether enterohepatic Helicobacter bilis modulates Helicobacter pylori gastritis in C57BL/6 mice. Thirty mice per group were sham dosed, H. bilis or H. pylori infected, or H. bilis infected followed in 2 weeks by H. pylori and then evaluated at 6 and 11 months postinfection (mpi) for gastritis and premalignant lesions. Compared to H. pylori-infected mice, H. bilis/H. pylori-infected mice at 6 and 11 mpi had less severe gastritis, atrophy, mucous metaplasia and hyperplasia (P < 0.01) and, additionally, at 11 mpi, less severe intestinal metaplasia and dysplasia (P < 0.05). H. bilis/H. pylori-infected mice at 11 mpi exhibited less Ki67 labeling of proliferating epithelial cells, reduced numbers of FoxP3(+) T-regulatory (T(REG)) cells, and lower FoxP3(+) mRNA levels than did H. pylori-infected mice (P < 0.05). Proinflammatory interleukin-1beta (IL-1beta), gamma interferon, and tumor necrosis factor alpha mRNA levels were attenuated in H. bilis/H. pylori-infected mice at 6 and 11 mpi (P < 0.01), although anti-inflammatory IL-10, IL-13, and transforming growth factor beta1 mRNA levels were not consistently impacted by H. bilis coinfection. Decreased pathology in H. bilis/H. pylori-infected mice correlated with higher gastric H. pylori colonization at 6 mpi (P < 0.001) and lower Th1-associated immunoglobulin G2c responses to H. pylori at 6 and 10 mpi (P < 0.05). We hypothesized that reduced pathology in H. bilis/H. pylori-infected mice was due to H. bilis-primed T(REG) cells in the lower bowel that migrated to the gastric compartment and inhibited Th1 responses to subsequent H. pylori infection. Thus, H. pylori-induced gastric lesions may vary in mouse models of unknown enteric helicobacter infection status and, importantly, variable sequelae to human H. pylori infection, particularly in developing countries, may occur where coinfection with lower bowel helicobacters and H. pylori may be common.


Asunto(s)
Gastritis/microbiología , Gastritis/patología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/inmunología , Helicobacter pylori/patogenicidad , Linfocitos T Reguladores/inmunología , Animales , Citocinas/biosíntesis , Duodeno/patología , Femenino , Factores de Transcripción Forkhead/biosíntesis , Antígeno Ki-67/biosíntesis , Ratones , Ratones Endogámicos C57BL , Estómago/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA