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












Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
Elife ; 72018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30039798

RESUMEN

The early-life intestinal microbiota plays a key role in shaping host immune system development. We found that a single early-life antibiotic course (1PAT) accelerated type 1 diabetes (T1D) development in male NOD mice. The single course had deep and persistent effects on the intestinal microbiome, leading to altered cecal, hepatic, and serum metabolites. The exposure elicited sex-specific effects on chromatin states in the ileum and liver and perturbed ileal gene expression, altering normal maturational patterns. The global signature changes included specific genes controlling both innate and adaptive immunity. Microbiome analysis revealed four taxa each that potentially protect against or accelerate T1D onset, that were linked in a network model to specific differences in ileal gene expression. This simplified animal model reveals multiple potential pathways to understand pathogenesis by which early-life gut microbiome perturbations alter a global suite of intestinal responses, contributing to the accelerated and enhanced T1D development.


Asunto(s)
Antibacterianos/efectos adversos , Diabetes Mellitus Tipo 1/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata/efectos de los fármacos , Inmunidad Adaptativa/efectos de los fármacos , Animales , Antibacterianos/inmunología , Diabetes Mellitus Tipo 1/microbiología , Diabetes Mellitus Tipo 1/patología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Íleon/inmunología , Íleon/microbiología , Inmunidad Innata/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos NOD , Microbiota/efectos de los fármacos , Microbiota/inmunología
5.
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
7.
Nat Microbiol ; 3(2): 234-242, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29180726

RESUMEN

Antibiotic exposure in children has been associated with the risk of inflammatory bowel disease (IBD). Antibiotic use in children or in their pregnant mother can affect how the intestinal microbiome develops, so we asked whether the transfer of an antibiotic-perturbed microbiota from mothers to their children could affect their risk of developing IBD. Here we demonstrate that germ-free adult pregnant mice inoculated with a gut microbial community shaped by antibiotic exposure transmitted their perturbed microbiota to their offspring with high fidelity. Without any direct or continued exposure to antibiotics, this dysbiotic microbiota in the offspring remained distinct from controls for at least 21 weeks. By using both IL-10-deficient and wild-type mothers, we showed that both inoculum and genotype shape microbiota populations in the offspring. Because IL10-/- mice are genetically susceptible to colitis, we could assess the risk due to maternal transmission of an antibiotic-perturbed microbiota. We found that the IL10-/- offspring that had received the perturbed gut microbiota developed markedly increased colitis. Taken together, our findings indicate that antibiotic exposure shaping the maternal gut microbiota has effects that extend to the offspring, with both ecological and long-term disease consequences.


Asunto(s)
Antibacterianos/administración & dosificación , Colitis/microbiología , Susceptibilidad a Enfermedades/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/microbiología , Animales , Colitis/inducido químicamente , Colon/inmunología , Colon/microbiología , Colon/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Disbiosis/microbiología , Heces/microbiología , Femenino , Enfermedades Inflamatorias del Intestino/inducido químicamente , Interleucina-10 , Metagenoma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fenotipo , Embarazo
8.
J Exp Med ; 214(12): 3519-3530, 2017 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-29066577

RESUMEN

Regulatory T (T reg) cells are a specialized sublineage of T lymphocytes that suppress autoreactive T cells. Functional studies of T reg cells in vitro have defined multiple suppression mechanisms, and studies of T reg-deficient humans and mice have made clear the important role that these cells play in preventing autoimmunity. However, many questions remain about how T reg cells act in vivo. Specifically, it is not clear which suppression mechanisms are most important, where T reg cells act, and how they get there. To begin to address these issues, we sought to identify T reg cells in zebrafish, a model system that provides unparalleled advantages in live-cell imaging and high-throughput genetic analyses. Using a FOXP3 orthologue as a marker, we identified CD4-enriched, mature T lymphocytes with properties of T reg cells. Zebrafish mutant for foxp3a displayed excess T lymphocytes, splenomegaly, and a profound inflammatory phenotype that was suppressed by genetic ablation of lymphocytes. This study identifies T reg-like cells in zebrafish, providing both a model to study the normal functions of these cells in vivo and mutants to explore the consequences of their loss.


Asunto(s)
Linfocitos T Reguladores/inmunología , Pez Cebra/inmunología , Animales , Secuencia de Bases , Enfermedad Crónica , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Hematopoyesis , Inflamación/patología , Linfocitos/metabolismo , Mutación/genética , Filogenia , Esplenomegalia/patología , Análisis de Supervivencia , Timocitos/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
9.
Nat Microbiol ; 1(11): 16140, 2016 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-27782139

RESUMEN

The early life microbiome plays important roles in host immunological and metabolic development. Because the incidence of type 1 diabetes (T1D) has been increasing substantially in recent decades, we hypothesized that early-life antibiotic use alters gut microbiota, which predisposes to disease. Using non-obese diabetic mice that are genetically susceptible to T1D, we examined the effects of exposure to either continuous low-dose antibiotics or pulsed therapeutic antibiotics (PAT) early in life, mimicking childhood exposures. We found that in mice receiving PAT, T1D incidence was significantly higher, and microbial community composition and structure differed compared with controls. In pre-diabetic male PAT mice, the intestinal lamina propria had lower Th17 and Treg proportions and intestinal SAA expression than in controls, suggesting key roles in transducing the altered microbiota signals. PAT affected microbial lipid metabolism and host cholesterol biosynthetic gene expression. These findings show that early-life antibiotic treatments alter the gut microbiota and its metabolic capacities, intestinal gene expression and T-cell populations, accelerating T1D onset in non-obese diabetic mice.


Asunto(s)
Antibacterianos/efectos adversos , Diabetes Mellitus Tipo 1/etiología , Microbioma Gastrointestinal/efectos de los fármacos , Penicilina V/efectos adversos , Animales , Antibacterianos/administración & dosificación , Colesterol/biosíntesis , Esquema de Medicación , Heces/microbiología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Expresión Génica/efectos de los fármacos , Predisposición Genética a la Enfermedad , Metabolismo de los Lípidos/efectos de los fármacos , Metaboloma/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/inmunología , Obesidad , Penicilina V/administración & dosificación , ARN Ribosómico 16S , Linfocitos T Reguladores , Células Th17
10.
Oncotarget ; 7(43): 69136-69148, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27655701

RESUMEN

OBJECTIVE: Chronic Helicobacter pylori (H. pylori) infection promotes non-cardia gastric cancer. Some mouse models suggest that bone marrow derived cells (BMDC) contribute to Helicobacter-associated gastric carcinogenesis. We determined whether this increased susceptibility to Helicobacter-induced gastric carcinogenesis of p27-deficient mice is dependent upon their p27-null BMDC or their p27-null gastric epithelial cells. DESIGN: Female mice (recipients) were irradiated and transplanted with BMDC from male donors. Wild type (WT) mice in group 1 (control) received BMDC from male GFP-transgenic mice. Female WT and p27 KO mice were engrafted with male p27KO mice BMDC (Group 2) or GFP-transgenic WT BMDC (Group 3). Recipients were infected with H. pylori SS1 for one year. RESULTS: Mice lacking p27 in either the BM pool or gastric epithelium developed significantly more advanced gastric pathology, including high-grade dysplasia. Co-staining of donor BMDC in dysplastic gastric glands was confirmed by immunofluorescence. Gastric expression of IL-1 beta protein was reduced in groups 2 and 3 (p < 0.05 vs control) whereas expression of IFN-γ and chemokines MIP-1 beta, MIG, IP-10 and RANTES in group 2 were significantly higher than group 3. CONCLUSIONS: Both bone marrow-derived and gastric epithelial cells contribute to the increased gastric cancer susceptibility of p27-deficient H. pylori-infected mice.


Asunto(s)
Células de la Médula Ósea/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Infecciones por Helicobacter/metabolismo , Neoplasias Gástricas/metabolismo , Animales , Células de la Médula Ósea/microbiología , Trasplante de Médula Ósea/métodos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Citocinas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mosaicismo , Neoplasias Gástricas/microbiología
11.
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
12.
Nat Med ; 21(8): 906-13, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26107252

RESUMEN

The inflammasome activates caspase-1 and the release of interleukin-1ß (IL-1ß) and IL-18, and several inflammasomes protect against intestinal inflammation and colitis-associated colon cancer (CAC) in animal models. The absent in melanoma 2 (AIM2) inflammasome is activated by double-stranded DNA, and AIM2 expression is reduced in several types of cancer, but the mechanism by which AIM2 restricts tumor growth remains unclear. We found that Aim2-deficient mice had greater tumor load than Asc-deficient mice in the azoxymethane/dextran sodium sulfate (AOM/DSS) model of colorectal cancer. Tumor burden was also higher in Aim2(-/-)/Apc(Min/+) than in APC(Min/+) mice. The effects of AIM2 on CAC were independent of inflammasome activation and IL-1ß and were primarily mediated by a non-bone marrow source of AIM2. In resting cells, AIM2 physically interacted with and limited activation of DNA-dependent protein kinase (DNA-PK), a PI3K-related family member that promotes Akt phosphorylation, whereas loss of AIM2 promoted DNA-PK-mediated Akt activation. AIM2 reduced Akt activation and tumor burden in colorectal cancer models, while an Akt inhibitor reduced tumor load in Aim2(-/-) mice. These findings suggest that Akt inhibitors could be used to treat AIM2-deficient human cancers.


Asunto(s)
Neoplasias del Colon/prevención & control , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/fisiología , Inflamasomas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Colitis/complicaciones , Femenino , Células HCT116 , Humanos , Pólipos Intestinales/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación
13.
Cancer Lett ; 359(2): 345-51, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25853150

RESUMEN

H. pylori infection causes gastritis, peptic ulcers and gastric cancer. Eradicating H. pylori prevents ulcers, but to what extent this prevents cancer remains unknown, especially if given after intestinal metaplasia has developed. H. pylori infected wild-type (WT) mice do not develop cancer, but mice lacking the tumor suppressor p27 do so, thus providing an experimental model of H. pylori-induced cancer. We infected p27-deficient mice with H. pylori strain SS1 at 6-8 weeks of age. Persistently H. pylori-infected WT C57BL/6 mice served as controls. Mice in the eradication arms received antimicrobial therapy (omeprazole, metronidazole and clarithromycin) either "early" (at 15 weeks post infection, WPI) or "late" at 45 WPI. At 70 WPI, mice were euthanized for H. pylori determination, histopathology and cytokine/chemokine expression. Persistently infected mice developed premalignant lesions including high-grade dysplasia, whereas those given antibiotics did not. Histologic activity scores in the eradication groups were similar to each other, and were significantly decreased compared with controls for inflammation, epithelial defects, hyperplasia, metaplasia, atrophy and dysplasia. IP-10 and MIG levels in groups that received antibiotics were significantly lower than controls. There were no significant differences in expression of IFN-γ, TNF-α, IL-1ß, RANTES, MCP-1, MIP-1α or MIP-1ß among the three groups. Thus, H. pylori eradication given either early or late after infection significantly attenuated gastric inflammation, gastric atrophy, hyperplasia, and dysplasia in the p27-deficient mice model of H. pylori-induced gastric cancer, irrespective of the timing of antibiotic administration. This was associated with reduced expression of IP-10 and MIG.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones por Helicobacter/complicaciones , Neoplasias Gástricas/microbiología , Animales , Antibacterianos/farmacología , Quimiocina CXCL10/sangre , Quimiocina CXCL10/genética , Quimiocina CXCL9/sangre , Quimiocina CXCL9/genética , Claritromicina/farmacología , Claritromicina/uso terapéutico , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Expresión Génica , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/inmunología , Humanos , Metronidazol/farmacología , Metronidazol/uso terapéutico , Ratones Endogámicos C57BL , Omeprazol/farmacología , Omeprazol/uso terapéutico , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/microbiología
14.
Gastrointest Cancer ; 5: 61-71, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25844041

RESUMEN

BACKGROUND: PTEN loss contributes to the development of liver diseases including hepatic steatosis and both hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC). The factors that influence the penetrance of these conditions are unclear. We explored the influence of sustained hypoxia signaling through co-deletion of Pten and Vhl in a murine model. METHODS: We used a CreER-linked Keratin 18 mouse model to conditionally delete Pten, Vhl or both in somatic cells of adult mice, evaluating the resultant tumors by histology and gene expression microarray. Existing sets of gene expression data for human HCC and CC were examined for pathways related to those observed in the murine tumors, and a cohort of human CC samples was evaluated for relationships between HIF-1α expression and clinical outcomes. RESULTS: Both Pten deletion genotypes developed liver tumors, but with differing phenotypes. Pten deletion alone led to large hepatic tumors with widespread hepatosteatosis. Co-deletion of Pten and Vhl with the Keratin 18 promoter resulted in reduced steatosis and a reduced tumor burden that was characterized by a trabecular architecture similar to CC. Genes associated with hepatic steatosis were coordinately expressed in the human HCC dataset, while genes involved in hypoxia response were upregulated in tumors from the human CC dataset. HIF-1α expression and overall survival were examined in an independent cohort of human CC tumors with no statistical differences uncovered. CONCLUSION: Pten deletion in Keratin 18 expressing cells leads to aggressive tumor formation and widespread steatosis in mouse livers. Co-deletion of Vhl and Pten results in lower tumor burden with gene expression profiling suggesting a switch from a profile of lipid deposition to an expression profile more consistent with upregulation of the hypoxia response pathway. A relationship between tumor hypoxia signaling and altered hepatic steatotic response suggests that competing influences may alter tumor phenotypes.

15.
Clin Cancer Res ; 20(23): 6083-95, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25231403

RESUMEN

PURPOSE: Tumor cells are surrounded by a complex microenvironment. The purpose of our study was to evaluate the role of heterogeneity of the tumor microenvironment in the variability of nanoparticle (NP) delivery and efficacy. EXPERIMENTAL DESIGNS: C3(1)-T-Antigen genetically engineered mouse model (C3-TAg) and T11/TP53(Null) orthotopic syngeneic murine transplant model (T11) representing human breast tumor subtypes basal-like and claudin-low, respectively, were evaluated. For the pharmacokinetic studies, non-liposomal doxorubicin (NL-doxo) or polyethylene glycol tagged (PEGylated) liposomal doxorubicin (PLD) was administered at 6 mg/kg i.v. x1. Area under the concentration versus time curve (AUC) of doxorubicin was calculated. Macrophages, collagen, and the amount of vasculature were assessed by IHC. Chemokines and cytokines were measured by multiplex immunochemistry. NL-doxo or PLD was administered at 6 mg/kg i.v. weekly x6 in efficacy studies. Analyses of intermediary tumor response and overall survival were performed. RESULTS: Plasma AUC of NL-doxo and PLD encapsulated and released doxorubicin was similar between two models. However, tumor sum total AUC of PLD was 2-fold greater in C3-TAg compared with T11 (P < 0.05). T11 tumors showed significantly higher expression of CC chemokine ligand (CCL) 2 and VEGF-a, greater vascular quantity, and decreased expression of VEGF-c compared with C3-TAg (P < 0.05). PLD was more efficacious compared with NL-doxo in both models. CONCLUSION: The tumor microenvironment and/or tumor cell features of breast cancer affected NP tumor delivery and efficacy, but not the small-molecule drug. Our findings reveal the role of the tumor microenvironment in variability of NP delivery and therapeutic outcomes.


Asunto(s)
Neoplasias de la Mama/patología , Nanopartículas/metabolismo , Microambiente Tumoral , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Quimiocina CCL2/sangre , Quimiocina CCL2/metabolismo , Quimiocina CCL5/sangre , Quimiocina CCL5/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacocinética , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Nanopartículas/administración & dosificación , Neovascularización Patológica , Polietilenglicoles/administración & dosificación , Polietilenglicoles/farmacocinética , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Lett ; 355(1): 106-12, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25218349

RESUMEN

H. pylori infection causes gastritis, peptic ulcers and gastric cancer. Eradicating H. pylori prevents ulcers, but to what extent this prevents cancer remains unknown, especially if given after intestinal metaplasia has developed. H. pylori infected wild-type (WT) mice do not develop cancer, but mice lacking the tumor suppressor p27 do so, thus providing an experimental model of H. pylori-induced cancer. We infected p27-deficient mice with H. pylori strain SS1 at 6-8 weeks of age. Persistently H. pylori-infected WT C57BL/6 mice served as controls. Mice in the eradication arms received antimicrobial therapy (omeprazole, metronidazole and clarithromycin) either "early" (at 15 weeks post infection, WPI) or "late" at 45 WPI. At 70 WPI, mice were euthanized for H. pylori determination, histopathology and cytokine/chemokine expression. Persistently infected mice developed premalignant lesions including high-grade dysplasia, whereas those given antibiotics did not. Histologic activity scores in the eradication groups were similar to each other, and were significantly decreased compared with controls for inflammation, epithelial defects, hyperplasia, metaplasia, atrophy and dysplasia. IP-10 and MIG levels in groups that received antibiotics were significantly lower than controls. There were no significant differences in expression of IFN-γ, TNF-α, IL-1ß, RANTES, MCP-1, MIP-1α or MIP-1ß among the three groups. Thus, H. pylori eradication given either early or late after infection significantly attenuated gastric inflammation, gastric atrophy, hyperplasia, and dysplasia in the p27-deficient mice model of H. pylori-induced gastric cancer, irrespective of the timing of antibiotic administration. This was associated with reduced expression of IP-10 and MIG.


Asunto(s)
Antibacterianos/administración & dosificación , Gastritis/prevención & control , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/efectos de los fármacos , Neoplasias Gástricas/prevención & control , Estómago/efectos de los fármacos , Animales , Atrofia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Esquema de Medicación , Quimioterapia Combinada , Mucosa Gástrica/metabolismo , Gastritis/genética , Gastritis/metabolismo , Gastritis/microbiología , Gastritis/patología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Hiperplasia , Mediadores de Inflamación/metabolismo , Metaplasia , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de la Bomba de Protones/administración & dosificación , Estómago/microbiología , Estómago/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Factores de Tiempo
17.
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
18.
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
19.
Crit Rev Oncog ; 19(6): 469-81, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25597356

RESUMEN

Helicobacter pylori is a helical bacterium that colonizes the stomach in over half of the world's population. Infection with this bacterium has been linked to peptic ulcer disease and gastric cancer. The bacterium has been shown to affect regulatory pathways in its host cells through specific virulence factors that control gene expression. Infection with H. pylori increases levels of phosphorylation of Raf kinase inhibitor protein (pRKIP) in gastric adenocarcinoma (AGS) cells in vitro and in vivo. We investigated the role of H. pylori in the phosphorylation of RKIP as a possible mechanism to downregulate pro-survival signals in gastric adenocarcinoma. pRKIP induces RKIP transcriptional activity, which serves to induce apoptosis of damaged cells to prevent further tumorigenesis. Infection of wild type and RKIP knockout mice with H. pylori for 2 months further confirmed roles of RKIP and pRKIP in the prevention of gastric cancer progression. Loss of RKIP in AGS cells results in increased expression of the Cag A virulence factor after H. pylori infection and RKIP overexpression inhibits H. pylori-mediated STAT3 phosphorylation and STAT3 and NF-κB transcriptional activity. We examined the role of mTOR (mammalian target of rapamycin) after H. pylori infection on the phosphorylation of RKIP. Cells treated with rapamycin, an inhibitor of mTOR, displayed less expression of pRKIP after H. pylori infection. Microarray antibody analysis was conducted on wild-type and RKIP-knockdown AGS cells and showed that in the absence of RKIP, there was increased expression of pro-tumorigenic proteins such as EGFR, Raf-1, and MAPKs. Although further work is needed to confirm the interaction of RKIP and mTOR in AGS cells as a result of H. pylori infection, we hypothesize that H. pylori-mediated induction of pro-survival signaling in gastric epithelial cells induces a feedback response through the activation of RKIP. The phosphorylated, or active, form of RKIP is important in protecting gastric epithelial cells from tumorigenesis after H. pylori infection.


Asunto(s)
Infecciones por Helicobacter/complicaciones , Helicobacter pylori/fisiología , Proteínas de Unión a Fosfatidiletanolamina/fisiología , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología , Animales , Carcinogénesis/genética , Infecciones por Helicobacter/genética , Humanos , Ratones , Ratones Noqueados , Proteínas de Unión a Fosfatidiletanolamina/genética , Transducción de Señal/genética
20.
PLoS One ; 8(9): e73347, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098634

RESUMEN

Although antiretroviral treatment decreases HIV-AIDS morbidity/mortality, long-term side effects may include the onset of insulin resistance and cardiovascular diseases. However, the underlying molecular mechanisms responsible for highly active antiretroviral therapy (HAART)-induced cardio-metabolic effects are poorly understood. In light of this, we hypothesized that HIV protease inhibitor (PI) treatment (Lopinavir/Ritonavir) elevates myocardial oxidative stress and concomitantly inhibits the ubiquitin proteasome system (UPS), thereby attenuating cardiac function. Lopinavir/Ritonavir was dissolved in 1% ethanol (vehicle) and injected into mini-osmotic pumps that were surgically implanted into Wistar rats for 8 weeks vs. vehicle and sham controls. We subsequently evaluated metabolic parameters, gene/protein markers and heart function (ex vivo Langendorff perfusions). PI-treated rats exhibited increased serum LDL-cholesterol, higher tissue triglycerides (heart, liver), but no evidence of insulin resistance. In parallel, there was upregulation of hepatic gene expression, i.e. acetyl-CoA carboxylase b and 3-hydroxy-3-methylglutaryl-CoA-reductase, key regulators of fatty acid oxidation and cholesterol synthesis, respectively. PI-treated hearts displayed impaired cardiac contractile function together with attenuated UPS activity. However, there was no significant remodeling of hearts exposed to PIs, i.e. lack of ultrastructural changes, fibrosis, cardiac hypertrophic response, and oxidative stress. Western blot analysis of PI-treated hearts revealed that perturbed calcium handling may contribute to the PI-mediated contractile dysfunction. Here chronic PI administration led to elevated myocardial calcineurin, nuclear factor of activated T-cells 3 (NFAT3), connexin 43, and phosphorylated phospholamban, together with decreased calmodulin expression levels. This study demonstrates that early changes triggered by PI treatment include increased serum LDL-cholesterol levels together with attenuated cardiac function. Furthermore, PI exposure inhibits the myocardial UPS and leads to elevated calcineurin and connexin 43 expression that may be associated with the future onset of cardiac contractile dysfunction.


Asunto(s)
Terapia Antirretroviral Altamente Activa/métodos , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/farmacología , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Western Blotting , Calcineurina/metabolismo , Proteínas de Unión al Calcio/metabolismo , LDL-Colesterol/sangre , Conexina 43/metabolismo , Cartilla de ADN/genética , Combinación de Medicamentos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Lopinavir/farmacología , Masculino , Factores de Transcripción NFATC/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Ritonavir/farmacología , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...