Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.485
Filtrar
1.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884748

RESUMEN

Intramedullary spinal cord tumors are a rare and understudied cancer with poor treatment options and prognosis. Our prior study used a combination of PDGF-B, HRAS, and p53 knockdown to induce the development of high-grade glioma in the spinal cords of minipigs. In this study, we evaluate the ability of each vector alone and combinations of vectors to produce high-grade spinal cord gliomas. Eight groups of rats (n = 8/group) underwent thoracolumbar laminectomy and injection of lentiviral vector in the lateral white matter of the spinal cord. Each group received a different combination of lentiviral vectors expressing PDGF-B, a constitutively active HRAS mutant, or shRNA targeting p53, or a control vector. All animals were monitored once per week for clinical deficits for 98 days. Tissues were harvested and analyzed using hematoxylin and eosin (H&E) and immunohistochemical (IHC) staining. Rats injected with PDGF-B+HRAS+sh-p53 (triple cocktail) exhibited statistically significant declines in all behavioral measures (Basso Beattie Bresnahan scoring, Tarlov scoring, weight, and survival rate) over time when compared to the control. Histologically, all groups except the control and those injected with sh-p53 displayed the development of tumors at the injection site, although there were differences in the rate of tumor growth and the histopathological features of the lesions between groups. Examination of immunohistochemistry revealed rats receiving triple cocktail displayed the largest and most significant increase in the Ki67 proliferation index and GFAP positivity than any other group. PDGF-B+HRAS also displayed a significant increase in the Ki67 proliferation index. Rats receiving PDGF-B alone and PDGF-B+ sh-p53 displayed more a significant increase in SOX2-positive staining than in any other group. We found that different vector combinations produced differing high-grade glioma models in rodents. The combination of all three vectors produced a model of high-grade glioma more efficiently and aggressively with respect to behavioral, physiological, and histological characteristics than the rest of the vector combinations. Thus, the present rat model of spinal cord glioma may potentially be used to evaluate therapeutic strategies in the future.


Asunto(s)
Glioma/etiología , Lentivirus/genética , Neoplasias de la Médula Espinal/etiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Vectores Genéticos , Glioma/patología , Glioma/fisiopatología , Mutación , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Neoplasias Experimentales/fisiopatología , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Neoplasias de la Médula Espinal/patología , Neoplasias de la Médula Espinal/fisiopatología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
2.
Int J Mol Sci ; 22(14)2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-34299314

RESUMEN

Signal transducer and activator of transcription 1 (STAT1) acts as a tumor suppressor molecule in colitis-associated colorectal cancer (CAC), particularly during the very early stages, modulating immune responses and controlling mechanisms such as apoptosis and cell proliferation. Previously, using an experimental model of CAC, we reported increased intestinal cell proliferation and faster tumor development, which were consistent with more signs of disease and damage, and reduced survival in STAT1-/- mice, compared with WT counterparts. However, the mechanisms through which STAT1 might prevent colorectal cancer progression preceded by chronic inflammation are still unclear. Here, we demonstrate that increased tumorigenicity related to STAT1 deficiency could be suppressed by IL-17 neutralization. The blockade of IL-17 in STAT1-/- mice reduced the accumulation of CD11b+Ly6ClowLy6G+ cells resembling granulocytic myeloid-derived suppressor cells (MDSCs) in both spleen and circulation. Additionally, IL-17 blockade reduced the recruitment of neutrophils into intestinal tissue, the expression and production of inflammatory cytokines, and the expression of intestinal STAT3. In addition, the anti-IL-17 treatment also reduced the expression of Arginase-1 and inducible nitric oxide synthase (iNOS) in the colon, both associated with the main suppressive activity of MDSCs. Thus, a lack of STAT1 signaling induces a significant change in the colonic microenvironment that supports inflammation and tumor formation. Anti-IL-17 treatment throughout the initial stages of CAC related to STAT1 deficiency abrogates the tumor formation possibly caused by myeloid cells.


Asunto(s)
Neoplasias Asociadas a Colitis/etiología , Granulocitos/patología , Interleucina-17/fisiología , Factor de Transcripción STAT1/fisiología , Animales , Anticuerpos Neutralizantes/administración & dosificación , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/fisiopatología , Progresión de la Enfermedad , Femenino , Granulocitos/inmunología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/patología , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Neoplasias Experimentales/fisiopatología , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Microambiente Tumoral/inmunología
3.
Cancer Prev Res (Phila) ; 14(5): 527-540, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33648941

RESUMEN

Skin cancer is the most common malignancy worldwide and is rapidly rising in incidence, representing a significant public health challenge. The ß-blocker, carvedilol, has shown promising effects in preventing skin cancer. However, as a potent ß-blocker, repurposing carvedilol to an anticancer agent is limited by cardiovascular effects. Carvedilol is a racemic mixture consisting of equimolar S- and R-carvedilol, whereas the R-carvedilol enantiomer does not possess ß-blocking activity. Because previous studies suggest that carvedilol's cancer preventive activity is independent of ß-blockade, we examined the skin cancer preventive activity of R-carvedilol compared with S-carvedilol and the racemic carvedilol. R- and S-carvedilol were equally effective in preventing EGF-induced neoplastic transformation of the mouse epidermal JB6 Cl 41-5a (JB6 P+) cells and displayed similar attenuation of EGF-induced ELK-1 activity. R-carvedilol appeared slightly better than S-carvedilol against UV-induced intracellular oxidative stress and release of prostaglandin E2 from the JB6 P+ cells. In an acute UV-induced skin damage and inflammation mouse model using a single irradiation of 300 mJ/cm2 UV, topical treatment with R-carvedilol dose dependently attenuated skin edema and reduced epidermal thickening, Ki-67 staining, COX-2 protein, and IL6 and IL1ß mRNA levels similar to carvedilol. In a chronic UV (50-150 mJ/cm2) induced skin carcinogenesis model in mice with pretreatment of test agents, topical treatment with R-carvedilol, but not racemic carvedilol, significantly delayed and reduced skin squamous cell carcinoma development. Therefore, as an enantiomer present in an FDA-approved agent, R-carvedilol may be a better option for developing a safer and more effective preventive agent for skin carcinogenesis. PREVENTION RELEVANCE: In this study, we demonstrated the skin cancer preventive activity of R-carvedilol, the non-ß-blocking enantiomer present in the racemic ß-blocker, carvedilol. As R-carvedilol does not have ß-blocking activity, such a preventive treatment would not lead to common cardiovascular side effects of ß-blockers.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carvedilol/administración & dosificación , Epidermis/efectos de los fármacos , Neoplasias Experimentales/prevención & control , Neoplasias Cutáneas/prevención & control , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Carvedilol/química , Células Epidérmicas , Factor de Crecimiento Epidérmico/toxicidad , Epidermis/patología , Epidermis/efectos de la radiación , Femenino , Células HEK293 , Humanos , Ratones , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Estereoisomerismo , Rayos Ultravioleta/efectos adversos
4.
Arch Dermatol Res ; 313(4): 263-273, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32638071

RESUMEN

Glypican-3 (GPC3) is considered as a cell surface heparan sulfate proteoglycan. It is overexpressed in skin cancer and promotes tumor progression and pathogenicity. Therefore, we aimed to find out the therapeutic effects of immuno-suppressing GPC3 in skin cancer experimentally induced in mice as well as to underline molecular mechanisms especially inflammatory and apoptotic pathways. Skin cancer was experimentally induced in mice by repeated rubbing of mice skin with 7,12-dimethylbenz (a) anthracene. Mice were injected with anti-GPC3. Skin samples were isolated to investigate the gene and protein expression of GPC3, Wnt-1, NFκB, TNF-α, IGF-1, p38 MAPK and caspase-3 using PCR, Western blot and ELISA. Moreover, skin sections were stained with hematoxylin and eosin. Treating skin cancer mice with anti-GPC3 significantly blocked GPC3, which is accompanied by amelioration of skin cancer-induced increase in the numbers of tumors and scratching behavior. Moreover, anti-GPC3 attenuated skin cancer-induced increase in the expression of Wnt-1, NFκB, TNF-α, IGF-1, p38 MAPK and caspase-3. In parallel, anti-GPC3 reduced degeneration of melanocyte cells and reduced phagocytic cells epidermal hyperplasia and dysplasia in skin sections stained with hematoxylin and eosin stain. In conclusion, anti-GPC3 produced anti-tumor effects against skin cancer, which can be explained by reduction in both inflammatory and apoptotic pathways. Targeting GPC3 is a promising therapeutic approach for skin cancer.


Asunto(s)
Antineoplásicos/farmacología , Carcinogénesis/efectos de los fármacos , Glipicanos/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , 9,10-Dimetil-1,2-benzantraceno/administración & dosificación , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Antineoplásicos/uso terapéutico , Carcinogénesis/inducido químicamente , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Neoplasias Experimentales/etiología , Piel/efectos de los fármacos , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/patología
5.
Nat Commun ; 11(1): 4653, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938923

RESUMEN

Cancer cells demand excess nutrients to support their proliferation, but how tumours exploit extracellular amino acids during systemic metabolic perturbations remain incompletely understood. Here, we use a Drosophila model of high-sugar diet (HSD)-enhanced tumourigenesis to uncover a systemic host-tumour metabolic circuit that supports tumour growth. We demonstrate coordinate induction of systemic muscle wasting with tumour-autonomous Yorkie-mediated SLC36-family amino acid transporter expression as a proline-scavenging programme to drive tumourigenesis. We identify Indole-3-propionic acid as an optimal amino acid derivative to rationally target the proline-dependency of tumour growth. Insights from this whole-animal Drosophila model provide a powerful approach towards the identification and therapeutic exploitation of the amino acid vulnerabilities of tumourigenesis in the context of a perturbed systemic metabolic network.


Asunto(s)
Azúcares de la Dieta/efectos adversos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Neoplasias Experimentales/fisiopatología , Prolina/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Animales , Animales Modificados Genéticamente , Carcinogénesis , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Hemolinfa/efectos de los fármacos , Hemolinfa/metabolismo , Larva , Debilidad Muscular/inducido químicamente , Debilidad Muscular/patología , Atrofia Muscular/inducido químicamente , Atrofia Muscular/patología , Neoplasias Experimentales/etiología , Proteínas Nucleares/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transactivadores/genética , Proteínas Señalizadoras YAP , Proteínas ras/genética
6.
Int J Mol Sci ; 21(11)2020 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-32526987

RESUMEN

Recent advances in the development of new methods of cancer immunotherapy require the production of complex cancer animal models that reliably reflect the complexity of the tumor and its microenvironment. Mice are good animals to create tumor models because they are low cost, have a short reproductive cycle, exhibit high tumor growth rates, and can be easily genetically modified. However, the obvious problem of these models is the high failure rate observed in human clinical trials after promising results obtained in mouse models. In order to increase the reliability of the results obtained in mice, the tumor model should reflect the heterogeneity of the tumor, contain components of the tumor microenvironment, in particular immune cells, to which the action of immunotherapeutic drugs are directed. This review discusses the current immunocompetent and immunocompromised mouse models of human tumors that are used to evaluate the effectiveness of immunotherapeutic agents, in particular chimeric antigen receptor (CAR) T-cells and immune checkpoint inhibitors.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Experimentales/etiología , Neoplasias Experimentales/terapia , Animales , Carcinógenos/toxicidad , Humanos , Huésped Inmunocomprometido , Isoinjertos , Ratones , Ratones Mutantes , Ratones Transgénicos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 15(4): e0226858, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32267859

RESUMEN

High-power microwaves are used to inhibit electronics of threatening military or civilian vehicles. This work aims to assess health hazards of high-power microwaves and helps to define hazard threshold levels of modulated radiofrequency exposures such as those emitted by the first generations of mobile phones. Rats were exposed to the highest possible field levels, under single acute or repetitive exposures for eight weeks. Intense microwave electric fields at 1 MV m-1 of nanoseconds duration were applied from two sources at different carrier frequencies of 10 and 3.7 GHz. The repetition rate was 100 pps, and the duration of train pulses lasted from 10 s to twice 8 min. The effects on the central nervous system were evaluated, by labelling brain inflammation marker GFAP and by performing different behavioural tests: rotarod, T-maze, beam-walking, open-field, and avoidance test. Long-time survival was measured in animals repeatedly exposed, and anatomopathological analysis was performed on animals sacrificed at two years of life or earlier in case of precocious death. Control groups were sham exposed. Few effects were observed on behaviour. With acute exposure, an avoidance reflex was shown at very high thermal level (22 W kg-1); GFAP was increased some days after exposure. Most importantly, with repeated exposures, survival time was 4-months shorter in the exposed group, with eleven animals exhibiting a large sub-cutaneous tumour, compared to two in the sham group. A residual X-ray exposure was also present in the beam (0.8 Gy), which is probably not a bias for the observed result. High power microwaves below thermal level in average, can increase cancer prevalence and decrease survival time in rats, without clear effects on behaviour. The parameters of this effect need to be further explored, and a more precise dosimetry to be performed.


Asunto(s)
Carcinogénesis/efectos de la radiación , Microondas/efectos adversos , Neoplasias Experimentales/epidemiología , Animales , Reacción de Prevención/efectos de la radiación , Conducta Animal/efectos de la radiación , Teléfono Celular , Incidencia , Masculino , Neoplasias Experimentales/etiología , Radiometría , Ratas , Ratas Sprague-Dawley , Análisis de Supervivencia , Factores de Tiempo
8.
Br J Haematol ; 190(3): 418-429, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32130729

RESUMEN

Myelodysplastic syndrome (MDS), a largely incurable hematological malignancy, is driven by complex genetic and epigenetic alterations from an aberrant clone of hematopoietic stem/progenitor cells (HSPCs). Ubiquitin-specific protease 7 (USP7) has been demonstrated to have an important oncogenic role in the development of several cancer types, but its role in MDS is unknown. Here, we demonstrate that USP7 expression is elevated in MDS cell lines and patient samples. The USP7-selective small-molecule inhibitors P5091 and P22077 inhibited cell proliferation and induced megakaryocytic differentiation in both cell lines and primary cells. Furthermore, pharmacological inhibition of USP7 markedly suppressed the growth of MDS cell lines in xenograft mouse models. To explore the mechanisms underlying the observed phenotypic changes, we employed RNA-seq to compare the differences in genes after USP7 inhibitor treatment and found that gelsolin (GSN) expression was increased significantly after USP7 inhibitor treatment. Furthermore, knockdown of GSN attenuated the proliferation inhibition, apoptosis induction and megakaryocyte differentiation induced by USP7 inhibitors in MDS cells. Collectively, our findings identify previously unknown roles of USP7 and suggest that the USP7/GSN axis may be a potential therapeutic target in MDS.


Asunto(s)
Gelsolina/fisiología , Megacariocitos/efectos de los fármacos , Síndromes Mielodisplásicos/patología , Inhibidores de Proteasas/farmacología , Tiofenos/farmacología , Trombopoyesis/efectos de los fármacos , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular/trasplante , Inducción Enzimática/efectos de los fármacos , Gelsolina/biosíntesis , Gelsolina/genética , Xenoinjertos , Humanos , Megacariocitos/patología , Ratones , Ratones Endogámicos NOD , Neoplasias Experimentales/etiología , Riesgo , Transcriptoma/efectos de los fármacos , Peptidasa Específica de Ubiquitina 7/fisiología , Regulación hacia Arriba/efectos de los fármacos
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165660, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31891805

RESUMEN

Epidemiological studies over the last few decades have shown a strong influence of obesity on colon cancer risk and its progression. These studies have primarily focussed on the role of adipokines in driving cancer progression. We investigated the incidence of cancerous polyp formation and tumor progression in presence and absence of functional leptin along with exploring the role of tumor necrosis factor α (TNFα), under obese condition. By utilizing diet induced obese and genetically obese mice, carcinogen induced colon polyp formation was investigated. Experiments were performed using tumor tissues and cell lines to delineate the inter-relationship between leptin and TNFα. Data shown in this report indicates that in leptin knockdown obese mice, AOM/DSS induced polyps are smaller and lesser in numbers as compared to AOM/DSS induced polyps in diet induced obese mice. Further in vitro experiments suggest that abrogation of leptin associated pathways promote TNFα induced apoptosis. Mechanistically, we report that TNFα induces p53 independent cell death through up regulation of p53 upregulated modulator of apoptosis (PUMA). TNFα induced PUMA was inhibited upon pre- exposure of cells to leptin, prior to TNFα treatment. Collectively these results indicate that obesity due to leptin non-functionality facilitates TNFα induced colon cancer cell death.


Asunto(s)
Neoplasias del Colon/metabolismo , Leptina/metabolismo , Neoplasias Experimentales/metabolismo , Obesidad/complicaciones , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Azoximetano/toxicidad , Neoplasias del Colon/epidemiología , Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Pólipos del Colon/inducido químicamente , Pólipos del Colon/patología , Sulfato de Dextran/toxicidad , Dieta Alta en Grasa/efectos adversos , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Incidencia , Leptina/genética , Ratones , Ratones Noqueados , Ratones Obesos , Neoplasias Experimentales/epidemiología , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Obesidad/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba
10.
Carcinogenesis ; 41(2): 194-202, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-31074772

RESUMEN

Alcohol consumption is the key risk factor for the development of esophageal squamous cell carcinoma (ESCC), and acetaldehyde, a metabolite of alcohol, is an alcohol-derived major carcinogen that causes DNA damage. Aldehyde dehydrogenase2 (ALDH2) is an enzyme that detoxifies acetaldehyde, and its activity is reduced by ALDH2 gene polymorphism. Reduction in ALDH2 activity increases blood, salivary and breath acetaldehyde levels after alcohol intake, and it is deeply associated with the development of ESCC. Heavy alcohol consumption in individuals with ALDH2 gene polymorphism significantly elevates the risk of ESCC; however, effective prevention has not been established yet. In this study, we investigated the protective effects of Alda-1, a small molecule ALDH2 activator, on alcohol-mediated esophageal DNA damage. Here, we generated novel genetically engineered knock-in mice that express the human ALDH2*1 (wild-type allele) or ALDH2*2 gene (mutant allele). Those mice were crossed, and human ALDH2*1/*1, ALDH2*1/*2 and ALDH2*2/*2 knock-in mice were established. They were given 10% ethanol for 7 days in the presence or absence of Alda-1, and we measured the levels of esophageal DNA damage, represented by DNA adduct (N2-ethylidene-2'-deoxyguanosine). Alda-1 significantly increased hepatic ALDH2 activity both in human ALDH2*1/*2 and/or ALDH2*2/*2 knock-in mice and reduced esophageal DNA damage levels after alcohol drinking. Conversely, cyanamide, an ALDH2-inhibitor, significantly exacerbated esophageal DNA adduct level in C57BL/6N mice induced by alcohol drinking. These results indicate the protective effects of ALDH2 activation by Alda-1 on esophageal DNA damage levels in individuals with ALDH2 gene polymorphism, providing a new insight into acetaldehyde-mediated esophageal carcinogenesis and prevention.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Benzamidas/administración & dosificación , Benzodioxoles/administración & dosificación , Carcinogénesis/efectos de los fármacos , Neoplasias Esofágicas/prevención & control , Carcinoma de Células Escamosas de Esófago/prevención & control , Acetaldehído/metabolismo , Acetaldehído/toxicidad , Aldehído Deshidrogenasa Mitocondrial/antagonistas & inhibidores , Aldehído Deshidrogenasa Mitocondrial/genética , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Cianamida/administración & dosificación , Aductos de ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Mucosa Esofágica/efectos de los fármacos , Mucosa Esofágica/patología , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/etiología , Carcinoma de Células Escamosas de Esófago/patología , Etanol/metabolismo , Etanol/toxicidad , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones Transgénicos , Mutación , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Polimorfismo Genético , Factores de Riesgo
11.
Bull Cancer ; 107(1): 30-40, 2020 Jan.
Artículo en Francés | MEDLINE | ID: mdl-31466696

RESUMEN

Primarily used in genetic studies of development, the zebrafish (Danio rerio) has rapidly emerged as a promising animal model of human cancer. Cancer cell transplantation in zebrafish constitutes a key platform for clinical research since it allows to study cellular and molecular events involved in various aspects of tumorigenesis and to evaluate the efficacy of therapeutic molecules in vivo. Applied to patient-derived cells, the xenotransplantation approach in zebrafish allows to define the most appropriate therapeutic strategies for specific alterations found in patients in the context of personalized medicine. This review discusses the zebrafish transplantation model for the study of cancer development and drug discovery.


Asunto(s)
Trasplante de Neoplasias , Neoplasias Experimentales/etiología , Medicina de Precisión/métodos , Investigación Biomédica Traslacional/métodos , Pez Cebra , Inmunidad Adaptativa , Animales , Animales Modificados Genéticamente , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Descubrimiento de Drogas , Genes Relacionados con las Neoplasias , Xenoinjertos , Humanos , Terapia de Inmunosupresión/métodos , Neoplasias Experimentales/genética , Oncogenes , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/inmunología
12.
Cancer Prev Res (Phila) ; 13(1): 41-52, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31796466

RESUMEN

Diets rich in fat, smoking, as well as exposure to environmental pollutants and dysbiosis of gut microbiota, increase the risk of developing colorectal cancer. Much progress has been made in combating colorectal cancer. However, options for chemoprevention from environmental insult and dysbiosis of gut microbiota remain elusive. We investigated the influence of berry-derived anthocyanidins (Anthos), with and without encapsulating them in bovine milk-derived exosomes (ExoAnthos), on the chemoprevention of bacteria-driven colon tumor development. Anthos and ExoAnthos treatment of colon cancer cells showed dose-dependent decreases in cell viability. Calculated selectivity index (SI) values for Anthos and ExoAnthos suggest that both treatments selectively targeted cancer over normal colon cells. In addition, ExoAnthos treatment yielded higher SI values than Anthos. Anthos and ExoAnthos treatment of ApcMin/+ mice inoculated with enterotoxigenic Bacteriodes fragilis (ETBF) bacteria led to significant decreases in colon tumor numbers over mice receiving vehicle treatments. Western blot analysis of normal colon, colon tumor, and liver tissue lysates showed that mice inoculated with ETBF featured increased expression of phase I enzymes in normal colon tissue and decreased expression of phase II enzymes in liver tissue. Treatment with the Anthos and ExoAnthos reverted the modulation of phase I and phase II enzymes, respectively; no significant changes in phase II enzyme expression occurred in colon tumor tissue. Treatment of HCT-116 cells with the ubiquitous carcinogen, benzo[a]pyrene (B[a]P) led to similar modulation of phase I and II enzymes, which was partially mitigated by treatment with Anthos. These results provide a promising outlook on the impact of berry Anthos for prevention and treatment of bacteria- and B[a]P-driven colorectal cancer.


Asunto(s)
Antocianinas/administración & dosificación , Neoplasias del Colon/prevención & control , Disbiosis/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Neoplasias Experimentales/prevención & control , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Antocianinas/aislamiento & purificación , Bacteroides fragilis/patogenicidad , Benzo(a)pireno/toxicidad , Carcinogénesis/inducido químicamente , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinógenos/toxicidad , Supervivencia Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Colon/patología , Neoplasias del Colon/etiología , Portadores de Fármacos/química , Disbiosis/metabolismo , Disbiosis/microbiología , Disbiosis/patología , Exosomas/química , Frutas/química , Microbioma Gastrointestinal/fisiología , Células HCT116 , Células HT29 , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones , Leche/citología , Neoplasias Experimentales/etiología , Vaccinium myrtillus/química
13.
Genesis ; 57(10): e23323, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31218818

RESUMEN

Neuroblastoma, an embryonal tumor arising from the sympathetic ganglia and adrenal medulla, is among the most intractable pediatric cancers. Although a variety of genetic changes have been identified in neuroblastoma, how they contribute to its pathogenesis remains largely unclear. Recent studies have identified alterations of the anaplastic lymphoma kinase (ALK) gene in neuroblastoma; ALK F1174L (a phenylalanine-to-leucine substitution at codon 1174) represents one of the most frequent of these somatic mutations, and is associated with amplification of the MYCN gene, the most reliable marker for the poor survival. We engineered the mouse Alk locus so that ALK F1174L is expressed by its endogenous promoter and can be induced in a spatiotemporally controlled fashion using Cre-loxP system. Although expression of ALK F1174L resulted in enhanced proliferation of sympathetic ganglion progenitors and increased the size of the sympathetic ganglia, it was insufficient to cause neuroblastoma. However, lethal neuroblastoma frequently developed in mice co-expressing ALK F1174L and MYCN, even in a genetic background where MYCN alone does not cause overt tumors. These data reveal that physiological expression of ALK F1174L significantly potentiates the oncogenic ability of MYCN in vivo. Our conditional mutant mice provide a valuable platform for investigating the pathogenesis of neuroblastoma.


Asunto(s)
Quinasa de Linfoma Anaplásico/genética , Neoplasias Experimentales/genética , Neuroblastoma/etiología , Animales , Carcinogénesis/genética , Femenino , Ganglios Simpáticos/crecimiento & desarrollo , Ingeniería Genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes/genética , Mutagénesis Insercional , Proteína Proto-Oncogénica N-Myc/biosíntesis , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/etiología , Neuroblastoma/enzimología , Neuroblastoma/genética
14.
J Cell Biochem ; 120(10): 16668-16680, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31095784

RESUMEN

Early detection of colorectal cancer and monitoring the progress in colon carcinogenesis stages is essential to reduce mortality. Therefore, there is continuous search for noninvasive biomarkers with high stability and good sensitivity and specificity. miRNAs have attracted attention as promising biomarkers as they are stably expressed in circulation. The aim of our study is to evaluate the aberrant expression of circulating miRNAs during the stepwise progress of colitis-associated colon cancer. This was accomplished through assessing the expression levels of five miRNAs (miR-141, miR-15b, miR-17-3p, miR-21, and miR-29a) in serum and their corresponding tissue samples through the different cycles of colorectal carcinogenesis cascade using the azoxymethane/dextran sulfate sodium murine model. We also compared the diagnostic performance of these selected miRNAs with the conventional tumor biomarkers CEA and CA 19-9. The results of our study revealed that the expression levels of those miRNAs were dynamically changing in accordance with the tumor development state. Moreover, their aberrant expression in serum was statistically correlated with that in tissue. Our data also revealed that serum miR-15b, miR-21, and miR-29a showed the best performance in terms of diagnostic power. Our findings highlight the efficiency of these circulating miRNAs not only for early diagnostics purposes, but also for monitoring progress in the colorectal carcinogenesis process, and therefore encouraging integrating these noninvasive biomarkers into the clinical diagnostic settings beside the traditional diagnostic markers for accurate screening of the early progress of colon carcinogenesis.


Asunto(s)
MicroARN Circulante/sangre , Colitis , Neoplasias Colorrectales , Regulación Neoplásica de la Expresión Génica , Neoplasias Experimentales , Animales , Azoximetano/toxicidad , Colitis/sangre , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/diagnóstico , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/etiología , Sulfato de Dextran/toxicidad , Masculino , Ratones , Neoplasias Experimentales/sangre , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/etiología
15.
Nat Commun ; 10(1): 2225, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31110179

RESUMEN

The effective prevention of tumor initiation, especially for potentially inoperable tumors, will be beneficial to obtain an overall higher quality of our health and life. Hence, thorough understanding of the pathophysiological mechanisms of early tumor formation arising from identifiable cellular origins is required to develop efficient preventative and early treatment options for each tumor type. Here, using genetically engineered mouse models, we provide preclinical experimental evidence for a long-standing open question regarding the pathophysiological potential of a microenvironmental and physiological stressor in tumor development, gastric acid-mediated regional microscopic injury in foregut squamous epithelia. This study demonstrates the association of gastric acid stress with Cyclooxygenase-2-dependent tumor formation originating from tumor-competent Krt5+/Krt15+ foregut basal progenitor cells. Our findings suggest that clinical management of microenvironmental stressor-mediated microscopic injury may be important in delaying tumor initiation from foregut basal progenitor cells expressing pre-existing tumorigenic mutation(s) and genetic alteration(s).


Asunto(s)
Carcinogénesis/patología , Ciclooxigenasa 2/metabolismo , Ácido Gástrico/metabolismo , Neoplasias Gastrointestinales/patología , Células Madre/patología , Animales , Diferenciación Celular/efectos de los fármacos , Sistema Digestivo/patología , Células Epiteliales/patología , Epitelio/patología , Neoplasias Gastrointestinales/etiología , Queratina-15/genética , Queratina-15/metabolismo , Queratina-5/genética , Queratina-5/metabolismo , Ratones , Ratones Transgénicos , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Inhibidores de la Bomba de Protones/farmacología , Estrés Fisiológico/efectos de los fármacos , Microambiente Tumoral
16.
Funct Integr Genomics ; 19(5): 729-742, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31044344

RESUMEN

Previous studies have demonstrated that deletion of cryptochrome (Cry) genes protects p53-/- mutant mice from the early onset of cancer and extends their median life-span by about 1.5-fold. Subsequent in vitro studies had revealed that deletion of Crys enhances apoptosis in response to UV damage through activation of p73 and inactivation of GSK3ß. However, it was not known at the transcriptome-wide level how deletion of Crys delays the onset of cancer in p53-/- mutant mice. In this study, the RNA-seq approach was taken to uncover the differentially expressed genes (DEGs) and pathways following UV-induced DNA damage in p53-/- and p53-/-Cry1-/-Cry2-/- mouse skin fibroblasts. Gene set enrichment analysis with the DEGs demonstrated enrichment in immune surveillance-associated genes regulated by IFN-γ and genes involved in TNFα signaling via NF-κB. Furthermore, protein network analysis enabled identification of DEGs p21, Sirt1, and Jun as key players, along with their interacting partners. It was also observed that the DEGs contained a high ratio of non-coding transcripts. Collectively, the present study suggests new genes in NF-κB regulation and IFN-γ response, as well as non-coding RNAs, may contribute to delaying the onset of cancer in p53-/-Cry1-/-Cry2-/- mice and increasing the life-span of these animals compared to p53-/- mice.


Asunto(s)
Apoptosis , Carcinogénesis/patología , Criptocromos/fisiología , Daño del ADN , Neoplasias Experimentales/patología , Transcriptoma , Proteína p53 Supresora de Tumor/fisiología , Animales , Carcinogénesis/metabolismo , Carcinogénesis/efectos de la radiación , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Ratones , Ratones Noqueados , Neoplasias Experimentales/etiología , Neoplasias Experimentales/metabolismo , Piel/metabolismo , Piel/patología , Piel/efectos de la radiación , Rayos Ultravioleta
17.
J Invest Dermatol ; 139(9): 2016-2028.e7, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30878676

RESUMEN

HIF-1α is constitutively expressed in mouse and human epidermis. It plays a crucial role in skin physiology, including the response of keratinocytes to UVR. However, little information is available about its role in photocarcinogenesis. Using a multistage model of UVB radiation-induced skin cancer, we show that the knockout of Hif-1α in the epidermis prevents tumorigenesis but at the same time triggers the formation of hyperkeratotic plaques. Our results indicate that the absence of oncogenic transformation in Hif-1α-ablated mice is related to increased DNA repair in keratinocytes, whereas the formation of hyperkeratotic plaques is caused by an increase in the levels of reactive oxygen species. Indeed, impairing the DNA repair machinery by ablating xeroderma pigmentosum C restored the UVB-induced neoplastic transformation of Hif-1α-ablated keratinocytes, whereas the development of hyperkeratotic plaques was blocked by chronic antioxidant treatment. We conclude that HIF-1α plays a procarcinogenic role in UVB-induced tumorigenesis.


Asunto(s)
Carcinogénesis/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Queratosis Actínica/patología , Neoplasias Cutáneas/patología , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis/genética , Apoptosis/efectos de la radiación , Carcinogénesis/efectos de la radiación , Daño del ADN/efectos de la radiación , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epidermis/patología , Epidermis/efectos de la radiación , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Queratosis Actínica/etiología , Ratones , Ratones Noqueados , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Estrés Oxidativo/genética , Estrés Oxidativo/efectos de la radiación , Neoplasias Cutáneas/etiología
18.
Oncogene ; 38(20): 3932-3945, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30692639

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with high mortality. Lack of effective treatment makes novel therapeutic discovery an urgent demand in PDAC research. By screening an epigenetic-related compound library, we identified THZ1, a covalent inhibitor of CDK7, as a promising candidate. Multiple long-established and patient-derived PDAC cell lines (PDC) were used to validate the efficacy of THZ1 in vitro. In addition, patient-derived xenograft (PDX) models and animal models of PDAC were utilized for examining THZ1 efficacy in vivo. Furthermore, RNA-Seq analyse was performed to reveal the molecular mechanism of THZ1 treatment. Finally, PDAC cell lines with primary or acquired resistance to THZ1 were investigated to explore the potential mechanism of THZ1 susceptibility. CDK7 inhibition was identified as a selective and potent therapeutic strategy for PDAC progression in multiple preclinical models. Mechanistic analyses revealed that CDK7 inhibition led to a pronounced downregulation of gene transcription, with a preferential repression of mitotic cell cycle and NF-κB signaling-related transcripts. MYC transcriptional was found to be involved in susceptibility of PDAC cells to CDK7 inhibition. In conclusion, Identification of CDK7-dependent transcriptional addiction in PDACs provides a potent therapeutic strategy that targets highly aggressive pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Quinasas Ciclina-Dependientes/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Fenilendiaminas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Animales , Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes myc , Humanos , Masculino , Ratones Endogámicos BALB C , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/etiología , Neoplasias Experimentales/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Pancreatitis/complicaciones , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Activadora de Quinasas Ciclina-Dependientes
19.
J Gerontol A Biol Sci Med Sci ; 74(2): 155-162, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29733330

RESUMEN

Caloric restriction (CR) is the most potent nonpharmacological intervention known to both protect against carcinogenesis and delay aging in laboratory animals. There is a growing number of anticarcinogens and CR mimetics that activate NAD(P)H:quinone oxidoreductase 1 (NQO1). We have previously shown that NQO1, an antioxidant enzyme that acts as an energy sensor through modulation of intracellular redox and metabolic state, is upregulated by CR. Here, we used NQO1-knockout (KO) mice to investigate the role of NQO1 in both the aging process and tumor susceptibility, specifically in the context of CR. We found that NQO1 is not essential for the beneficial effects of CR on glucose homeostasis, physical performance, metabolic flexibility, life-span extension, and (unlike our previously observation with Nrf2) chemical-induced tumorigenesis.


Asunto(s)
Composición Corporal , Restricción Calórica/efectos adversos , Longevidad , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Neoplasias Experimentales/prevención & control , Estrés Oxidativo , Animales , Biomarcadores de Tumor/metabolismo , Carcinogénesis , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/etiología , Neoplasias Experimentales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...