Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Carcinog ; 61(7): 702-716, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35452563

RESUMEN

Emerging evidence suggests an important role for SIRT1, a nicotinamide adenine dinucleotide (NAD)-dependent deacetylase in cancer development, progression and therapeutic resistance; making it a viable therapeutic target. Here, we examined the impact of resveratrol-mediated pharmacological activation of SIRT1 on the progression of HGPIN lesions (using the Pten-/- mouse model) and on prostate tumor development (using an orthotopic model of prostate cancer cells stably silenced for SIRT1). We show that precise SIRT1 modulation could benefit both cancer prevention and treatment. Positive effect of SIRT1 activation can prevent Pten deletion-driven development of HGPIN lesions in mice if resveratrol is administered early (pre-cancer stage) with little to no benefit after the establishment of HGPIN lesions or tumor cell implantation. Mechanistically, our results show that under androgen deprivation conditions, SIRT1 inhibition induces senescence as evidenced by decreased gene signature associated with negative regulators of senescence and increased senescence-associated ß-galactosidase activity. Furthermore, pharmacological inhibition of SIRT1 potentiated growth inhibitory effects of clinical androgen receptor blockade agents and radiation. Taken together, our findings provide an explanation for the discrepancy regarding the role of SIRT1 in prostate tumorigenesis. Our results reveal that the bifurcated roles for SIRT1 may occur in stage and context-dependent fashion by functioning in an antitumor role in prevention of early-stage prostate lesion development while promoting tumor development and disease progression post-lesion development. Clinically, these data highlight the importance of precise SIRT1 modulation to provide benefits for cancer prevention and treatment including sensitization to conventional therapeutic approaches.


Asunto(s)
Neoplasia Intraepitelial Prostática , Neoplasias de la Próstata , Antagonistas de Andrógenos/farmacología , Animales , Senescencia Celular , Humanos , Masculino , Ratones , Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/prevención & control , Resveratrol/farmacología , Sirtuina 1/genética
2.
Cell Immunol ; 366: 104383, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34111646

RESUMEN

For over 70 years experimental autoimmune encephalomyelitis (EAE) has been induced with myelin autoantigens emulsified in complete Freund's adjuvant (CFA) which has significant side effects such as pain, inflammation, and tissue necrosis at the injection site. ß-1,3-d-glucan particles (GPs) are hollow microcapsules prepared from Saccharomyces cerevisiae cell walls that induce potent Th17 cell responses without causing strong injection site tissue reactions. We evaluated the potential of GPs complexed with neuroantigens to induce EAE while avoiding undesirable side effects. GPs loaded with myelin oligodendrocyte glycoprotein 35-55 (MOG35-55) or proteolipid protein 139-151 (PLP139-151) peptides effectively induced EAE in C57BL/6 mice and SJL mice. Disease severity, CNS pathology and immune responses were comparable between GP- and CFA-immunized mice. Importantly, injection with GPs resulted in significantly decreased inflammation compared with CFA. We posit that use of GPs provides an alternative means for inducing EAE that results in comparable disease, but less discomfort to animals.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Cápsulas/metabolismo , Pared Celular/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Proteoglicanos/metabolismo , Saccharomyces cerevisiae/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Adyuvante de Freund , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Proteína Proteolipídica de la Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Proteoglicanos/inmunología , Células Th17/inmunología
3.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33673346

RESUMEN

2-Methoxyestradiol (2-ME2) possesses anti-tumorigenic activities in multiple tumor models with acceptable tolerability profile in humans. Incomplete understanding of the mechanism has hindered its development as an anti-tumorigenic compound. We have identified for the first-time macrophage stimulatory protein 1 receptor (MST1R) as a potential target of 2-ME2 in prostate cancer cells. Human tissue validation studies show that MST1R (a.k.a RON) protein levels are significantly elevated in prostate cancer tissues compared to adjacent normal/benign glands. Serum levels of macrophage stimulatory protein (MSP), a ligand for RON, is not only associated with the risk of disease recurrence, but also significantly elevated in samples from African American patients. 2-ME2 treatment inhibited mechanical properties such as adhesion and elasticity that are associated with epithelial mesenchymal transition by downregulating mRNA expression and protein levels of MST1R in prostate cancer cell lines. Intervention with 2-ME2 significantly reduced tumor burden in mice. Notably, global metabolomic profiling studies identified significantly higher circulating levels of bile acids in castrated animals that were decreased with 2-ME2 intervention. In summary, findings presented in this manuscript identified MSP as a potential marker for predicting biochemical recurrence and suggest repurposing 2-ME2 to target RON signaling may be a potential therapeutic modality for prostate cancer.


Asunto(s)
2-Metoxiestradiol/farmacología , Reposicionamiento de Medicamentos , Proteínas de Neoplasias , Neoplasias de la Próstata , Proteínas Tirosina Quinasas Receptoras , Animales , Humanos , Masculino , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Células PC-3 , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo
4.
Mol Carcinog ; 58(6): 854-861, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30859654

RESUMEN

Published evidence shows a correlation between several molecular markers and prostate cancer (PCa) progression including in African Americans (AAs) who are disproportionately affected. Our early detection efforts led to the identification of elevated levels of antiapoptotic protein, c-FLIP and its upstream regulatory factors such as androgen receptor (AR), recepteur d'origine nantais (RON), a receptor tyrosine kinase in human prostate tumors. The primary objective of this study was to explore whether these markers play a role in racial disparities using immunohistochemistry in prostatectomy samples from a cohort of AA, Hispanic Whites (HWs), and non-Hispanic Whites (NHWs). Bivariable and multivariable logistic regression analyses were used to identify a statistical association between molecular markers, possible correlation with risk factors including race, obesity, prostate-specific antigen (PSA) and disease aggressiveness. Further, changes in the levels and expression of these molecular markers were also evaluated using human PCa cell lines. We found significantly elevated levels of RON ( P = 0.0082), AR ( P = 0.0001), c-FLIP ( P = 0.0071) in AAs compared with HWs or NHWs. Furthermore, a higher proportion of HW and NHWs had a high Gleason score (>6) but not PSA as compared to AAs ( P = 0.032). In summary, our findings suggest that PSA was important in predicting aggressive disease for the cohort overall; however, high levels of RON may play a role in predisposing AA men to develop aggressive disease. Future research is needed using large datasets to confirm these findings and to explore whether all or any of these markers could aid in race-specific stratification of patients for treatment.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Próstata/etnología , Neoplasias de la Próstata/cirugía , Proteínas Tirosina Quinasas Receptoras/metabolismo , Regulación hacia Arriba , Adulto , Negro o Afroamericano , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular Tumoral , Estudios de Cohortes , Regulación Neoplásica de la Expresión Génica , Hispánicos o Latinos , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Prostatectomía , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Androgénicos/metabolismo , Población Blanca
5.
Mol Carcinog ; 58(3): 309-320, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30365185

RESUMEN

Hepatocellular carcinoma (HCC) remains a deadly cancer, underscoring the need for relevant preclinical models. Male C3HeB/FeJ mice model spontaneous HCC with some hepatocarcinogenesis susceptibility loci corresponding to syntenic regions of human chromosomes altered in HCC. We tested other properties of C3HeB/FeJ tumors for similarity to human HCC. C3HeB/FeJ tumors were grossly visible at 4 months of age, with prevalence and size increasing until about 11 months of age. Histologic features shared with human HCC include hepatosteatosis, tumor progression from dysplasia to poorly differentiated, vascular invasion, and trabecular, oncocytic, vacuolar, and clear cell variants. More tumor cells displayed cytoplasmic APE1 staining versus normal liver. Ultrasound effectively detected and monitored tumors, with 85.7% sensitivity. Over 5000 genes were differentially expressed based on the GSE62232 and GSE63898 human HCC datasets. Of these, 158 and 198 genes, respectively, were also differentially expressed in C3HeB/FeJ. Common cancer pathways, cell cycle, p53 signaling and other molecular aspects, were shared between human and mouse differentially expressed genes. We established eigengenes that distinguish HCC from normal liver in the C3HeB/FeJ model and a subset of human HCC. These features extend the relevance and improve the utility of the C3HeB/FeJ line for HCC studies.


Asunto(s)
Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Neoplasias Hepáticas/patología , Animales , Apoptosis , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Células Tumorales Cultivadas
6.
Int J Mol Sci ; 19(7)2018 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-30036938

RESUMEN

COBRA1 (co-factor of BRCA1) is one of the four subunits of the negative elongation factor originally identified as a BRCA1-interacting protein. Here, we provide first-time evidence for the oncogenic role of COBRA1 in prostate pathogenesis. COBRA1 is aberrantly expressed in prostate tumors. It positively influences androgen receptor (AR) target gene expression and promoter activity. Depletion of COBRA1 leads to decreased cell viability, proliferation, and anchorage-independent growth in prostate cancer cell lines. Conversely, overexpression of COBRA1 significantly increases cell viability, proliferation, and anchorage-independent growth over the higher basal levels. Remarkably, AR-positive androgen dependent (LNCaP) cells overexpressing COBRA1 survive under androgen-deprivation conditions. Remarkably, treatment of prostate cancer cells with well-studied antitumorigenic agent, 2-methoxyestradiol (2-ME2), caused significant DNA methylation changes in 3255 genes including COBRA1. Furthermore, treatment of prostate cancer cells with 2-ME2 downregulates COBRA1 and inhibition of prostate tumors in TRAMP (transgenic adenocarcinomas of mouse prostate) animals with 2-ME2 was also associated with decreased COBRA1 levels. These observations implicate a novel role for COBRA1 in progression to CRPC and suggest that COBRA1 downregulation has therapeutic potential.


Asunto(s)
Proteína BRCA1/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/metabolismo , Factores de Transcripción/metabolismo , 2-Metoxiestradiol , Animales , Proteína BRCA1/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Estradiol/análogos & derivados , Estradiol/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Proteínas Nucleares/genética , Neoplasias de la Próstata/genética , Unión Proteica , Proteínas de Unión al ARN , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Factores de Transcripción/genética
7.
Radiographics ; 36(4): 963-83, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27399236

RESUMEN

Myriad infectious organisms can infect the endocardium, myocardium, and pericardium, including bacteria, fungi, parasites, and viruses. Significant cardiac infections are rare in the general population but are associated with high morbidity and mortality as well as increased risk in certain populations, such as the elderly, those undergoing cardiac instrumentation, and intravenous drug abusers. Diagnostic imaging of cardiac infections plays an important role despite its variable sensitivity and specificity, which are due in part to the nonspecific manifestations of the central inflammatory process of infection and the time of onset with respect to the time of imaging. The primary imaging modality remains echocardiography. However, cardiac computed tomography and magnetic resonance (MR) imaging have emerged as the modalities of choice wherever available, especially for diagnosis of complex infectious complications including abscesses, infected prosthetic material, central lines and instruments, and the cryptic manifestations of viral and parasitic diseases. MR imaging can provide functional, morphologic, and prognostic value in a single examination by allowing characterization of inflammatory changes from the acute to chronic stages, including edema and the patterns and extent of delayed gadolinium enhancement. We review the heterogeneous and diverse group of cardiac infections based on their site of primary cardiac involvement with emphasis on their cross-sectional imaging manifestations. Online supplemental material is available for this article. (©)RSNA, 2016.


Asunto(s)
Cardiopatías/diagnóstico por imagen , Cardiopatías/fisiopatología , Infecciones/diagnóstico por imagen , Infecciones/fisiopatología , Diagnóstico Diferencial , Diagnóstico por Imagen , Cardiopatías/microbiología , Humanos , Infecciones/microbiología
8.
Lab Invest ; 95(10): 1092-104, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26214584

RESUMEN

The balance of nitric oxide (NO) versus superoxide generation has a major role in the initiation and progression of endothelial dysfunction. Under conditions of high glucose, endothelial nitric oxide synthase (eNOS) functions as a chief source of superoxide rather than NO. In order to improve NO bioavailability within the vessel wall in type-1 diabetes, we investigated treatment strategies that improve eNOS phosphorylation and NO-dependent vasorelaxation. We evaluated methods to increase the eNOS activity by (1) feeding Ins2(Akita) spontaneously diabetic (type-1) mice with l-arginine in the presence of sepiapterin, a precursor of tetrahydrobiopterin; (2) preventing eNOS/NO deregulation by the inclusion of inhibitor kappa B kinase beta (IKKß) inhibitor, salsalate, in the diet regimen in combination with l-arginine and sepiapterin; and (3) independently increasing eNOS expression to improve eNOS activity and associated NO production through generating Ins2(Akita) diabetic mice that overexpress human eNOS predominantly in vascular endothelial cells. Our results clearly demonstrated that diet supplementation with l-arginine, sepiapterin along with salsalate improved phosphorylation of eNOS and enhanced vasorelaxation of thoracic/abdominal aorta in type-1 diabetic mice. More interestingly, despite the overexpression of eNOS, the in-house generated transgenic eNOS-GFP (TgeNOS-GFP)-Ins2(Akita) cross mice showed an unanticipated effect of reduced eNOS phosphorylation and enhanced superoxide production. Our results demonstrate that enhancement of endogenous eNOS activity by nutritional modulation is more beneficial than increasing the endogenous expression of eNOS by gene therapy modalities.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Suplementos Dietéticos , Endotelio Vascular/metabolismo , Hipoglucemiantes/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Animales , Aorta/citología , Aorta/metabolismo , Aorta/fisiopatología , Arginina/metabolismo , Arginina/uso terapéutico , Bovinos , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Endotelio Vascular/citología , Endotelio Vascular/fisiopatología , Femenino , Heterocigoto , Humanos , Hipoglucemiantes/metabolismo , Insulina/genética , Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/química , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Inhibidores de Proteínas Quinasas/metabolismo , Pterinas/metabolismo , Pterinas/uso terapéutico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Salicilatos/metabolismo , Salicilatos/uso terapéutico , Destete
9.
Infect Immun ; 82(7): 2688-96, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24711570

RESUMEN

Lower genital tract infection with Chlamydia trachomatis and C. muridarum can induce long-lasting hydrosalpinx in the upper genital tract of women and female mice, respectively. However, A/J mice were highly resistant to induction of long-lasting hydrosalpinx by C. muridarum. We further compared host inflammatory responses and chlamydial infection courses between the hydrosalpinx-resistant A/J mice and CBA/J mice known to be susceptible to hydrosalpinx induction. Both mouse strains developed robust pyosalpinx during the acute phase followed by hydrosalpinx during the chronic phase. However, the hydrosalpinges disappeared in A/J mice by day 60 after infection, suggesting that some early hydrosalpinges are reversible. Although the overall inflammatory responses were indistinguishable between CBA/J and A/J mice, we found significantly more neutrophils in oviduct lumen of A/J mice on days 7 and 10, which correlated with a rapid but transient oviduct invasion by C. muridarum with a peak infection on day 7. In contrast, CBA/J mice developed a delayed and extensive oviduct infection. These comparisons have revealed an important role of the interactions of oviduct infection with inflammatory responses in chlamydial induction of long-lasting hydrosalpinx, suggesting that a rapid but transient invasion of oviduct by chlamydial organisms can prevent the development of the long-lasting hydrosalpinges.


Asunto(s)
Infecciones por Chlamydia/microbiología , Chlamydia muridarum/fisiología , Enfermedades de las Trompas Uterinas/microbiología , Animales , Infecciones por Chlamydia/patología , Enfermedades de las Trompas Uterinas/patología , Trompas Uterinas/microbiología , Trompas Uterinas/patología , Femenino , Inflamación/microbiología , Inflamación/patología , Ratones , Ratones Endogámicos , Infecciones del Sistema Genital/microbiología , Infecciones del Sistema Genital/patología
10.
Proc Natl Acad Sci U S A ; 108(21): 8779-84, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555558

RESUMEN

Diabetes and the development of its complications have been associated with mitochondrial DNA (mtDNA) dysfunction, but causal relationships remain undetermined. With the objective of testing whether increased mtDNA mutations exacerbate the diabetic phenotype, we have compared mice heterozygous for the Akita diabetogenic mutation (Akita) with mice homozygous for the D257A mutation in mitochondrial DNA polymerase gamma (Polg) or with mice having both mutations (Polg-Akita). The Polg-D257A protein is defective in proofreading and increases mtDNA mutations. At 3 mo of age, the Polg-Akita and Akita male mice were equally hyperglycemic. Unexpectedly, as the Polg-Akita males aged to 9 mo, their diabetic symptoms decreased. Thus, their hyperglycemia, hyperphagia and urine output declined significantly. The decrease in their food intake was accompanied by increased plasma leptin and decreased plasma ghrelin, while hypothalamic expression of the orexic gene, neuropeptide Y, was lower and expression of the anorexic gene, proopiomelanocortin, was higher. Testis function progressively worsened with age in the double mutants, and plasma testosterone levels in 9-mo-old Polg-Akita males were significantly reduced compared with Akita males. The hyperglycemia and hyperphagia returned in aged Polg-Akita males after testosterone administration. Hyperglycemia-associated distal tubular damage in the kidney also returned, and Polg-D257A-associated proximal tubular damage was enhanced. The mild diabetes of female Akita mice was not affected by the Polg-D257A mutation. We conclude that reduced diabetic symptoms of aging Polg-Akita males results from appetite suppression triggered by decreased testosterone associated with damage to the Leydig cells of the testis.


Asunto(s)
Apetito/genética , ADN Mitocondrial/genética , ADN Polimerasa Dirigida por ADN/genética , Diabetes Mellitus/genética , Ratones Endogámicos/genética , Mutación , Envejecimiento , Animales , ADN Polimerasa gamma , Diabetes Mellitus/patología , Diabetes Mellitus/fisiopatología , Femenino , Hiperglucemia , Células Intersticiales del Testículo/patología , Masculino , Ratones , Fenotipo , Edición de ARN/genética , Testículo/patología , Testosterona/sangre
11.
Mol Carcinog ; 52(4): 275-85, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22213062

RESUMEN

The prevalence of hepatocellular carcinoma (HCC) was diminished from 60% to 18% at 15 months of age in C3HeB/FeJ male transgenic mice expressing hMGMT in our previous studies. To directly test if the methyltransferase activity is required for diminished tumor prevalence, two separate lines of transgenic mice bearing an enzymatically inactive form of hMGMT were used. In these lines, cysteine 145 was substituted with alanine (C145A). Expression of the hMGMT C145A transgene in liver was demonstrated by Northern blots and Western blots. Immunohistochemistry revealed predominantly nuclear localization of the hMGMT C145A protein. hMGMT C145A transgenic mice were crossed with lacI transgenic mice to assess mutant frequencies in the presence of the mutant protein. Mutant frequencies were similar among livers of lacI × hMGMT C145A bi-transgenic mice and lacI × wild-type (WT) mice. DNA sequence analysis of recovered lacI mutants revealed similar mutation spectra for hMGMT C145A and WT mice. The prevalence of HCC was also similar for the two tested lines of hMGMT C145A mice, 45% and 48% prevalence with median tumor sizes of 11 and 8 mm, and WT mice, 40% prevalence and median tumor size of 10 mm. These results provide evidence that residue C145 in hMGMT is required to reduce the prevalence of HCC in C3HeB/FeJ mice transgenic for hMGMT.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Hígado/patología , O(6)-Metilguanina-ADN Metiltransferasa/genética , Sustitución de Aminoácidos , Animales , Carcinoma Hepatocelular/enzimología , Activación Enzimática , Humanos , Hígado/enzimología , Hígado/metabolismo , Neoplasias Hepáticas/enzimología , Masculino , Ratones , Ratones Transgénicos , O(6)-Metilguanina-ADN Metiltransferasa/análisis , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Transgenes
12.
PLoS One ; 17(12): e0279694, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36584001

RESUMEN

A perfluorocarbon (PFC) investigated for treatment of traumatic brain injury (TBI) delivers oxygen to support brain function, but causes transient thrombocytopenia. TBI can cause acute inflammation with resulting thrombocytopenia; an interaction between the PFC effects and TBI inflammation might exacerbate thrombocytopenia. Therefore, PFC effects on platelet (PLT) function and hemostasis in a lipopolysaccharide (LPS) model of inflammation in the baboon were studied. Animals were randomized to receive saline ±LPS, and ± one of two doses of PFC. PLT count, transmission electron microscopy, and microparticle populations were quantified at baseline (BL) and at 2, 24, 48, 72, and 96 hours; hemostatic parameters for aggregometry and for blood clotting were measured at baseline (BL) and days 3 and 4. Injection of vehicle and LPS caused thrombocytopenia within hours; PFCs caused delayed thrombocytopenia beginning 48 hours post-infusion. LPS+PFC produced a more prolonged PLT decline and decreased clot strength. LPS+PFC increased ADP-stimulated aggregation, but PFC alone did not. Microparticle abundance was greatest in the LPS+PFC groups. LPS+PFC caused diffuse microvascular hemorrhage and death in 2 of 5 baboons in the low dose LPS-PFC group and 2 of 2 in the high dose LPS-PFC group. Necropsy and histology suggested death was caused by shock associated with hemorrhage in multiple organs. Abnormal morphology of platelets and red blood cells were notable for PFC inclusions. In summary, PFC infusion caused clinically significant thrombocytopenia and exacerbated LPS-induced platelet activation. The interaction between these effects resulted in decreased hemostatic capacity, diffuse bleeding, shock and death.


Asunto(s)
Fluorocarburos , Inflamación , Animales , Modelos Animales de Enfermedad , Eritrocitos/efectos de los fármacos , Eritrocitos/patología , Fluorocarburos/envenenamiento , Hemorragia/inducido químicamente , Hemostáticos , Inflamación/tratamiento farmacológico , Lipopolisacáridos , Trombocitopenia/inducido químicamente
13.
J Cutan Pathol ; 38(5): 420-31, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21251037

RESUMEN

Trichodysplasia spinulosa (TS) is a folliculocentric and clinically papular dermatological disorder occurring in the setting of immunosuppression typically in association with solid organ transplantation or hematolymphoid malignancies. We report the occurrence of TS in a 7-year-old girl with Down syndrome and pre-B-acute lymphoblastic leukemia who was completing chemotherapy at onset. The patient's affected follicles were dilated by an expansion of a dystrophic follicular inner root sheath cell population displaying enlarged trichohyaline cytoplasmic granules and progressing centrally to keratotic and parakeratotic debris, and superficially demonstrating some diminutive hair shaft-like material within the keratotic spicules. Electron microscopic studies of a follicular lesion showed extracellular viral particles suggestive of a polyomavirus within the central follicular keratotic debris. DNA polymerase chain reaction (PCR) and gene sequencing studies, performed on the tissue of the microscopic slide and paraffin block, for the recently identified TS-associated polyomavirus (TSPyV) resulted as positive for TSPyV. PCR for the Merkel cell polyomavirus was negative. To date, this case is unique in representing the first case of TS confirmed by electron microscopy in which a related viral pathogen has been molecularly identified. An additional 19 reported cases classifiable as TS are tabulated and reviewed.


Asunto(s)
Enfermedades del Cabello/patología , Enfermedades del Cabello/virología , Folículo Piloso/patología , Folículo Piloso/virología , Infecciones por Polyomavirus/patología , Infecciones por Polyomavirus/virología , Poliomavirus , Niño , ADN Viral/metabolismo , Síndrome de Down/complicaciones , Síndrome de Down/metabolismo , Síndrome de Down/patología , Síndrome de Down/terapia , Femenino , Enfermedades del Cabello/metabolismo , Folículo Piloso/metabolismo , Humanos , Infecciones por Polyomavirus/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología
14.
J Gerontol A Biol Sci Med Sci ; 75(4): 647-653, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-30423026

RESUMEN

Nuclear factor (NF)κB is a transcription factor that controls immune and inflammatory signaling pathways. In skeletal muscle, NFκB has been implicated in the regulation of metabolic processes and tissue mass, yet its affects on mitochondrial function in this tissue are unclear. To investigate the role of NFκB on mitochondrial function and its relationship with muscle mass across the life span, we study a mouse model with muscle-specific NFκB suppression (muscle-specific IκBα super-repressor [MISR] mice). In wild-type mice, there was a natural decline in muscle mass with aging that was accompanied by decreased mitochondrial function and mRNA expression of electron transport chain subunits. NFκB inactivation downregulated expression of PPARGC1A, and upregulated TFEB and PPARGC1B. NFκB inactivation also decreased gastrocnemius (but not soleus) muscle mass in early life (1-6 months old). Lower oxygen consumption rates occurred in gastrocnemius and soleus muscles from young MISR mice, whereas soleus (but not gastrocnemius) muscles from old MISR mice displayed increased oxygen consumption compared to age-matched controls. We conclude that the NFκB pathway plays an important role in muscle development and growth. The extent to which NFκB suppression alters mitochondrial function is age dependent and muscle specific. Finally, mitochondrial function and muscle mass are tightly associated in both genotypes and across the life span.


Asunto(s)
Mitocondrias Musculares/fisiología , Desarrollo de Músculos/fisiología , FN-kappa B/fisiología , Envejecimiento/genética , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Citrato (si)-Sintasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Musculares/genética , Desarrollo de Músculos/genética , Músculo Esquelético/fisiología , Músculo Esquelético/ultraestructura , FN-kappa B/antagonistas & inhibidores , Consumo de Oxígeno , Transducción de Señal
15.
Commun Biol ; 3: 12, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31909204

RESUMEN

NAD[P]H:quinone oxidoreductase 1 (NQO1) regulates cell fate decisions in response to stress. Oxidative stress supports cancer maintenance and progression. Previously we showed that knockdown of NQO1 (NQO1low) prostate cancer cells upregulate pro-inflammatory cytokines and survival under hormone-deprived conditions. Here, we tested the ability of NQO1low cells to form tumors. We found NQO1low cells form aggressive tumors compared with NQO1high cells. Biopsy specimens and circulating tumor cells showed biochemical recurrent prostate cancer was associated with low NQO1. NQO1 silencing was sufficient to induce SMAD-mediated TGFß signaling and mesenchymal markers. TGFß treatment decreased NQO1 levels and induced molecular changes similar to NQO1 knockdown cells. Functionally, NQO1 depletion increased migration and sensitivity to oxidative stress. Collectively, this work reveals a possible new gatekeeper role for NQO1 in counteracting cellular plasticity in prostate cancer cells. Further, combining NQO1 with TGFß signaling molecules may serve as a better signature to predict biochemical recurrence.


Asunto(s)
Plasticidad de la Célula/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , Estrés Oxidativo , Neoplasias de la Próstata/fisiopatología , Factor de Crecimiento Transformador beta/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Neoplasias de la Próstata/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/fisiología
16.
Prostate ; 69(7): 755-60, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19170049

RESUMEN

BACKGROUND: Transgenic adenocarcinoma of mouse prostate (TRAMP) mice, derived by prostate specific expression of SV40 large T antigen using the rat probasin promoter, all develop prostate tumors akin to human prostate cancers. More recently, epithelial-stromal (ES) tumors resembling phyllodes tumors have been described in the seminal vesicles of TRAMP mice. We report malignancy arising in these ES tumors of the seminal vesicles in TRAMP mice. METHODS: H&E stained sections from 28-week-old TRAMP mice autopsies were examined. Immunostains (cytokeratin, vimentin, desmin, and MIB-1) and electron microscopy were performed on selected blocks of the genitourinary system and metastatic tumor nodules. RESULTS: The seminal vesicles frequently develop tumors containing broad papillae, with bland epithelium and a cellular spindled stroma just beneath the epithelium. The stromal cells have high nuclear to cytoplasmic ratio, frequent apoptotic cells and mitoses. In some cases, the stromal cells become large mass lesions that overgrow the prostate. The epithelium can also proliferate and become malignant. The tumors have high proliferation indices by MIB-1. Some metastatic tumors have characteristics similar to the seminal vesicle ES tumor. CONCLUSIONS: Metastatic tumors in TRAMP mice show three patterns: (1) A definite adenocarcinoma pattern metastatic from the prostate; (2) poorly differentiated tumor without epithelial differentiation; (3) carcinosarcomatous pattern. The carcinosarcomatous pattern and some of the poorly differentiated tumors likely arise from seminal vesicle ES tumors.


Asunto(s)
Adenocarcinoma/patología , Neoplasias de la Próstata/patología , Vesículas Seminales/patología , Adenocarcinoma/genética , Adenocarcinoma/ultraestructura , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/ultraestructura , Vesículas Seminales/ultraestructura
17.
PLoS One ; 14(12): e0226187, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31856170

RESUMEN

The purpose of this investigation was to compare the antitumorigenic effects of the natural product Nexrutine to voluntary wheel running (VWR) in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Forty-five, 10-week old TRAMP mice were randomized to either receive free access to the running wheel, Nexrutine pelleted into chow at 600 mg/kg or no treatment control. Mice were serially sacrificed at weeks 4, 8,12 and 20 weeks. Palpable tumors, body weight, food consumption and running wheel activity were monitored weekly. At necropsy, tumors and serum were harvested and stored for analysis. Serum was used to quantify circulating cytokines in 4 and 20 week time points. Nexrutine supplementation led to a 66% protection against high grade tumors. Exercise resulted in a 60% protection against high grade tumors. Both interventions reduced concentrations of IL-1α. Exercise also significantly lowered concentrations of eotaxin, IL-5, IL-12(p40) and VEGF. While there were no significant differences at baseline, exercise mice had significantly lower IL-5 and VEGF compared to control at the 20 week time point. Nexrutine also significantly reduced circulating IL-9 concentrations. No significant differences were observed when compared to the control group. Immunohistochemistry of tumor sections showed significantly lower expression of pAkt in Nexrutine fed mice with no visible differences for NFκB. In conclusion, both Nexrutine and exercise suppressed tumor growth. Though similar outcomes were seen in this comparative effectiveness study, the mechanisms by which exercise and Nexrutine exert this benefit may focus on different pathways.


Asunto(s)
Condicionamiento Físico Animal , Extractos Vegetales/farmacología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/prevención & control , Animales , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Clasificación del Tumor , Fosfoproteínas/metabolismo , Neoplasias de la Próstata/fisiopatología , Proteínas Proto-Oncogénicas c-akt/metabolismo
18.
J Diabetes Complications ; 33(1): 23-32, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30424931

RESUMEN

Previous studies demonstrated that global deficiency of eNOS in diabetic mice exacerbated renal lesions and that overexpression of eNOS may protect against tissue injury. Our study revealed for the first time overexpression of eNOS leads to disease progression rather than protection. Transgenic mice selectively expressing eNOS in endothelial cells (eNOSTg) were cross bred with Ins2Akita type-1 (AK) diabetic mice to generate eNOS overexpressing eNOSTg/AK mice. Wild type, eNOSTg, AK and eNOSTg/AK mice were assessed for kidney function and blood glucose levels. Remarkably, overexpressing eNOSTg mice showed evidence of unpredicted glomerular injury with segmental mesangiolysis and occasional microaneurysms. Notably, in eNOSTg/AK mice overexpression of eNOS led to increased glomerular/endothelial injury that was associated with increased superoxide levels and renal dysfunction. Results indicate for the first time that overexpressing eNOS in endothelial cells cannot ameliorate diabetic lesions, but paradoxically leads to progression of nephropathy likely due to eNOS uncoupling and superoxide upsurge. This novel finding has a significant impact on current therapeutic strategies to improve endothelial function and prevent progression of diabetic renal disease. Further, the eNOSTg/AK model developed in this study has significant translational potentials for elucidating the underlying mechanism implicated in the deflected function of eNOS in diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Endotelio Vascular/metabolismo , Glomérulos Renales/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Animales , Nefropatías Diabéticas/diagnóstico por imagen , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endotelio Vascular/diagnóstico por imagen , Endotelio Vascular/patología , Insulina/genética , Glomérulos Renales/diagnóstico por imagen , Glomérulos Renales/patología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Microscopía Electrónica
19.
Nat Commun ; 10(1): 4720, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31624249

RESUMEN

Understanding the molecular components of insulin signaling is relevant to effectively manage insulin resistance. We investigated the phenotype of the TMEM127 tumor suppressor gene deficiency in vivo. Whole-body Tmem127 knockout mice have decreased adiposity and maintain insulin sensitivity, low hepatic fat deposition and peripheral glucose clearance after a high-fat diet. Liver-specific and adipose-specific Tmem127 deletion partially overlap global Tmem127 loss: liver Tmem127 promotes hepatic gluconeogenesis and inhibits peripheral glucose uptake, while adipose Tmem127 downregulates adipogenesis and hepatic glucose production. mTORC2 is activated in TMEM127-deficient hepatocytes suggesting that it interacts with TMEM127 to control insulin sensitivity. Murine hepatic Tmem127 expression is increased in insulin-resistant states and is reversed by diet or the insulin sensitizer pioglitazone. Importantly, human liver TMEM127 expression correlates with steatohepatitis and insulin resistance. Our results suggest that besides tumor suppression activities, TMEM127 is a nutrient-sensing component of glucose/lipid homeostasis and may be a target in insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Genes Supresores de Tumor , Resistencia a la Insulina/genética , Hígado/metabolismo , Proteínas de la Membrana/genética , Adipogénesis/genética , Animales , Dieta Alta en Grasa , Perfilación de la Expresión Génica/métodos , Gluconeogénesis/genética , Humanos , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos/genética
20.
Lab Invest ; 88(5): 515-28, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18391994

RESUMEN

Functional consequences of impaired endothelial nitric oxide synthase (eNOS) activity causing organ-specific abnormalities on a diabetic setting are not completely understood. In this study, we extensively characterized a diabetic mouse model (lepr(db/db)) in which eNOS expression is genetically disrupted (eNOS-/-). The eNOS-/-/ lepr(db/db) double-knockout (DKO) mice developed obesity, hyperglycemia, hyperinsulinemia and hypertension. Analysis of tissues from DKO mice showed large islets in the pancreas and fat droplets in hepatocytes. Interestingly, the aorta was normal and atherogenic lesions were not observed. Abnormalities in the aorta including poor re-endothelialization and increased medial wall thickness were evident only in response to deliberate injury. In contrast, significant glomerular capillary damage in the kidney was identified, with DKO mice demonstrating a robust diabetic nephropathy similar to human disease. The vascular and renal impairments in DKO mice were pronounced despite lower fasting plasma glucose levels compared to lepr(db/db) mice, indicating that eNOS is a critical determinant of hyperglycemia-induced organ-specific complications and their severity in diabetes. Results provide the first evidence that absence of eNOS in diabetes has a greater deleterious effect on the renal microvasculature than on the larger aortic vessel. The DKO model may suggest novel therapeutic strategies to prevent both vascular and renal complications of diabetes.


Asunto(s)
Complicaciones de la Diabetes , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Angiopatías Diabéticas/etiología , Óxido Nítrico Sintasa de Tipo III/deficiencia , Albuminuria/fisiopatología , Animales , Aorta Abdominal/lesiones , Aorta Abdominal/patología , Peso Corporal , Creatinina/orina , Diabetes Mellitus/patología , Diabetes Mellitus/fisiopatología , Angiopatías Diabéticas/patología , Angiopatías Diabéticas/fisiopatología , Endotelio Vascular/patología , Genotipo , Hipertensión/etiología , Resistencia a la Insulina , Riñón/irrigación sanguínea , Riñón/patología , Glomérulos Renales/patología , Glomérulos Renales/ultraestructura , Macrófagos/patología , Ratones , Ratones Noqueados , Ratones Mutantes , Microcirculación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA