Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.358
Filtrar
1.
Sci Rep ; 11(1): 22085, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34764423

RESUMO

Bladder cancer (BCa) is the most common malignancy of the urinary system with increasing incidence, mortality, and limited treatment options. Therefore, it is imperative to validate preclinical models that faithfully represent BCa cellular, molecular, and metabolic heterogeneity to develop new therapeutics. We performed metabolomic profiling of premalignant and non-muscle invasive bladder cancer (NMIBC) that ensued in the chemical carcinogenesis N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN) mouse model. We identified the enriched metabolic signatures that associate with premalignant and NMIBC. We found that enrichment of lipid metabolism is the forerunner of carcinogen-induced premalignant and NMIBC lesions. Cross-species analysis revealed the prognostic value of the enzymes associated with carcinogen-induced enriched metabolic in human disease. To date, this is the first study describing the global metabolomic profiles associated with early premalignant and NMIBC and provide evidence that these metabolomic signatures can be used for prognostication of human disease.


Assuntos
Butilidroxibutilnitrosamina/toxicidade , Carcinógenos/toxicidade , Metaboloma , Neoplasias da Bexiga Urinária/induzido quimicamente , Urotélio/efeitos dos fármacos , Animais , Linhagem Celular , Humanos , Masculino , Metaboloma/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Urotélio/metabolismo , Urotélio/patologia
2.
Eur J Pharmacol ; 913: 174626, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34774852

RESUMO

Disruption in the nerve-tumor interaction is now considered as a possible anticancer strategy for treating various cancer types, particularly colorectal cancer. However, the underlying mechanisms are not still fully understood. Therefore, the present study aimed to evaluate the effects of sympathetic and parasympathetic denervation on the inhibition of colorectal cancer progression in early and late phases and assess the involvement of nerve growth factor in denervation mediated anticancer effects. One-hundred and fifty male Wistar rats were assigned into 15 groups. Seven groups comprising the control group, 1,2-dimethylhydrazine (DMH) group, sympathetic denervation group (celiac-mesenteric ganglionectomy and guanethidine sulphate administration), parasympathetic denervation group (vagotomy and atropine administration), and combination group were used in the early-stage protocol. For the late-stage protocol, eight groups comprising the control, DMH, surgical and pharmacological sympathetic and parasympathetic denervation groups, combination group, and 5-flourouracil group were considered. After 8 weeks, sympathetic and parasympathetic denervation significantly reduced ACF numbers in rats receiving DMH. On the other hand, in the late stages, parasympathetic but not sympathetic denervation resulted in significant reductions in tumor incidence, tumor volume and weight, cell proliferation (indicated by reduced immunostaining of PCNA and ki-67), and angiogenesis (indicated by reduced immunostaining of CD31 and VEGF expression levels), and downregulated NGF, ß2 adrenergic, and M3 receptors. It can be concluded that parasympathetic denervation may be of high importance in colon carcinogenesis and suggested as a possible therapeutic modality in late stages of colorectal cancer.


Assuntos
Atropina/administração & dosagem , Neoplasias Colorretais/cirurgia , Neoplasias Experimentais/cirurgia , Vagotomia , 1,2-Dimetilidrazina/administração & dosagem , 1,2-Dimetilidrazina/toxicidade , Animais , Carcinogênese/induzido quimicamente , Carcinógenos/administração & dosagem , Carcinógenos/toxicidade , Colo/inervação , Colo/patologia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/patologia , Progressão da Doença , Gânglios Simpáticos/efeitos dos fármacos , Gânglios Simpáticos/cirurgia , Ganglionectomia , Guanetidina/administração & dosagem , Humanos , Masculino , Mesentério/inervação , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/patologia , Sistema Nervoso Parassimpático/efeitos dos fármacos , Sistema Nervoso Parassimpático/cirurgia , Ratos , Ratos Wistar
3.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-34638879

RESUMO

Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is the rate-limiting enzyme in the glycosphingolipid (GSL)-biosynthesis pathway and overexpressed in many human tumors. We suppressed GSL-biosynthesis using the GCS inhibitor Genz-123346 (Genz), NB-DNJ (Miglustat) or by genetic targeting of the GCS-encoding gene UDP-glucose-ceramide-glucosyltransferase- (UGCG). GCS-inhibition or GSL-depletion led to a marked arrest of the cell cycle in Lovo cells. UGCG silencing strongly also inhibited tumor spheroid growth in Lovo cells and moderately in HCT116 cells. MS/MS analysis demonstrated markedly elevated levels of sphingomyelin (SM) and phosphatidylcholine (PC) that occurred in a Genz-concentration dependent manner. Ultrastructural analysis of Genz-treated cells indicated multi-lamellar lipid storage in vesicular compartments. In mice, Genz lowered the incidence of experimentally induced colorectal tumors and in particular the growth of colorectal adenomas. These results highlight the potential for GCS-based inhibition in the treatment of CRC.


Assuntos
Ciclo Celular/efeitos dos fármacos , Neoplasias do Colo , Dioxanos/farmacologia , Glicoesfingolipídeos , Pirrolidinas/farmacologia , Esferoides Celulares , Animais , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Glucosiltransferases/antagonistas & inibidores , Glucosiltransferases/metabolismo , Glicoesfingolipídeos/biossíntese , Glicoesfingolipídeos/genética , Células HCT116 , Humanos , Camundongos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
4.
Food Funct ; 12(21): 10632-10643, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34585698

RESUMO

Safflower yellow (SY) is the main active ingredient isolated from the traditional Chinese medicine Carthamus tinctorius, which is a valuable natural edible pigment that is widely used to treat cerebrovascular and cardiovascular diseases. However, the effect of SY on hepatocellular carcinoma (HCC) remains unclear. In this study, we showed that SY decreased the degree of injury and inhibited the release of inflammatory factors in the liver of a diethylnitrosamine (DEN)-induced HCC mouse model. Flow cytometry and immunoblotting showed that SY increased the infiltration of CD8+ T cells and Gr-1+ macrophages to improve the immune microenvironment by affecting the expression of collagen fibers. Further cellular experiments showed that SY degraded the collagens in the liver cells through the TGF-ß/Smad signalling pathway. SY also regulated the gut microbiota which may contribute to the immune microenvironment. In conclusion, SY exhibited a potent effect on the development of HCC by enhancing liver immune infiltration by promoting collagen degradation and modulating the gut microbiota. This study provides novel insights into the mechanism of SY as a candidate for the treatment of HCC in the future.


Assuntos
Carcinoma Hepatocelular/induzido quimicamente , Chalcona/análogos & derivados , Dietilnitrosamina/toxicidade , Microbioma Gastrointestinal/efeitos dos fármacos , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Animais , Carcinoma Hepatocelular/prevenção & controle , Linhagem Celular Tumoral , Chalcona/farmacologia , Colágeno/metabolismo , Humanos , Fígado/imunologia , Fígado/metabolismo , Neoplasias Hepáticas/prevenção & controle , Macrófagos Peritoneais/efeitos dos fármacos , Masculino , Camundongos , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/prevenção & controle , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
5.
J Am Chem Soc ; 143(37): 15233-15242, 2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34514797

RESUMO

Taking advantage of cancer cells' endogenous characters, the responsive activation of DNA nanomachines has achieved great success in tumor therapy. Combining with extra stimuli such as external light irradiation provided spatiotemporal control of DNA nanomachine activation. However, specific activation at the cellular level is still challenging considering the macroscopic-scale exposure area of usual light sources. DNA logic gates located at the cell membrane contributed to cellular specificity, but the free diffusion of input DNA strands during the operation process would impair efficiency and result in side effects to circumjacent normal cells in solid tumors. Here we design a transmembrane DNA logical computation strategy to activate a DNA nanomachine only in cancer cells from a complex solid tumor microenvironment. The DNA nanomachine multishell UCNPs-DNA is prepared by modifying DNA strands on upconversion nanoparticles. LA-apt, a DNA strand anchoring to a cancer cell membrane overexpressed receptor, and intracellular miRNA-21 served as inputs 1 and 2, respectively. Hybridization with input 1 at the cell membrane not only exposes the miRNA-21 recognition region at the DNA nanomachine, but also delivers it into cancer cells. The cascade hybridization with intracellular input 2 completes the "AND" gate operation and releases a DNA strand L2 as output. L2 acts as the trigger to operate the DNA nanomachine and correspondingly activates the photosensitizer Rose Bengal for reactive oxygen species generation. Through the "AND" gate operation of the DNA nanomachine across the cancer cell membrane, highly precise therapy only to cancer cells is achieved in a complex solid tumor microenvironment, which could become a promising modality for precise therapy of solid tumors.


Assuntos
Neoplasias da Mama/terapia , Engenharia Química/métodos , DNA/química , Nanoestruturas/uso terapêutico , Neoplasias/terapia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , Nanoestruturas/química , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/diagnóstico por imagem
6.
J Biochem Mol Toxicol ; 35(9): e22838, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34273909

RESUMO

Colorectal cancer (CRC) is the third most common type of cancer. Here, we studied the inhibitory effect of IRAK1 and IRAK4 as a preventive strategy using a colitis-induced tumorigenesis mouse model. CRC clinical data were obtained from the Gene Expression Omnibus (GEO). An experimental inflammation-dependent CRC model was induced by treatment with azoxymethane (AOM) and then dextran sodium sulfate (DSS) in C57BL/6 mice. Mice were administered an IRAK1/4 inhibitor by intraperitoneal injection at 3 mg/kg twice each week for 9 weeks. The IRAK1/4 inhibitor attenuated histological changes and prevented tumor growth. Tumor-associated proteins, including p65 and Ki-67, were downregulated by the IRAK1/4 inhibitor in AOM/DSS-treated mice. Additionally, IRAK1/4 inhibitor administration effectively decreased the expression of inflammatory cytokines. Furthermore, we observed that IRAK1/4 inhibitor treatment attenuated colitis-induced tumorigenesis by inhibiting epithelial-mesenchymal transition. These observations indicate that inhibition of IRAK1 and IRAK4 may suppress experimental colitis-induced tumorigenesis by inhibiting inflammatory responses and epithelial-mesenchymal transition.


Assuntos
Carcinogênese/efeitos dos fármacos , Neoplasias Associadas a Colite/tratamento farmacológico , Colite/tratamento farmacológico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Quinases Associadas a Receptores de Interleucina-1/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Experimentais/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Animais , Carcinogênese/induzido quimicamente , Carcinogênese/metabolismo , Colite/induzido quimicamente , Colite/enzimologia , Neoplasias Associadas a Colite/induzido quimicamente , Neoplasias Associadas a Colite/enzimologia , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Masculino , Camundongos , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/enzimologia
7.
J Ethnopharmacol ; 280: 114415, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34271113

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: The root of Angelica sinensis is widely used in traditional Chinese Medicine for relieving gynecological discomforts among the women population. However, its hormone-like effects have raised great attention on whether it is appropriate to use in breast cancer (BC) patients. Hence, this study aimed to investigate the tumorigenic effect of aqueous root extract of Angelica sinensis (AS) on estrogen receptor (ER)-positive BC growth through ER-induced stemness in-vitro and in-vivo. MATERIALS AND METHODS: The chemical composition of the AS was characterized by HPLC. Cell viability was detected by MTS assay. The in-vivo effect of AS was investigated by xenograft model, immunohistochemistry, histology, Western blot, and self-renewal ability assay. Target verification was used by shRNA construction and transfection. Mammosphere formation assay was performed by flow cytometry. RESULTS: AS significantly promoted the proliferation of MCF-7 cells and inhibited the growth of MDA-MB-231 cells. AS significantly induced tumor growth (2.5 mg/kg) in xenograft models and however tamoxifen treatment significantly suppressed the AS-induced tumor growth. AS induced ERα expression in both in-vivo and in-vitro and promoted cancer stem cell activity in ER-positive BC. CONCLUSION: AS shows the tumorigenic potential on ER-positive BC growth through ERα induced stemness, suggesting that the usage of AS is not recommended for BC in terms of safety measures.


Assuntos
Angelica sinensis/química , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/tratamento farmacológico , Carcinogênese/induzido quimicamente , Extratos Vegetais/efeitos adversos , Receptores de Estrogênio/metabolismo , Animais , Antineoplásicos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/tratamento farmacológico , Células-Tronco Neoplásicas , Extratos Vegetais/uso terapêutico , Raízes de Plantas/química , Receptores de Estrogênio/genética , Tamoxifeno/uso terapêutico
8.
Regul Toxicol Pharmacol ; 124: 104977, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34174380

RESUMO

The National Toxicology Program (NTP) reported that chronic dietary exposure to 4-methylimidazole (4-MeI) increased the incidence of lung adenomas/carcinomas beyond the normally high spontaneous rate in B6C3F1 mice. To examine plausible modes of action (MoAs) for mouse lung tumors (MLTs) upon exposure to high levels of 4-MeI, and their relevance in assessing human risk, a systematic approach was used to identify and evaluate mechanistic data (in vitro and in vivo) in the primary and secondary literature, along with high-throughput screening assay data. Study quality, relevance, and activity of mechanistic data identified across the evidence-base were organized according to key characteristics of carcinogens (KCCs) to identify potential key events in known or novel MLT MoAs. Integration of these evidence streams provided confirmation that 4-MeI lacks genotoxic and cytotoxic activity with some evidence to support a lack of mitogenic activity. Further evaluation of contextual and chemical-specific characteristics of 4-MeI was consequently undertaken. Due to lack of genotoxicity, along with transcriptomic and histopathological lung changes up to 28 and 90 days of exposure, the collective evidence suggests MLTs observed following exposure to high levels of 4-MeI develop at a late stage in the mouse chronic bioassay, albeit the exact MoA remains unclear.


Assuntos
Carcinógenos/toxicidade , Imidazóis/toxicidade , Neoplasias Pulmonares/epidemiologia , Neoplasias Experimentais/epidemiologia , Testes de Toxicidade Crônica/estatística & dados numéricos , Animais , Carcinógenos/administração & dosagem , Interpretação Estatística de Dados , Progressão da Doença , Relação Dose-Resposta a Droga , Imidazóis/administração & dosagem , Incidência , Pulmão/efeitos dos fármacos , Pulmão/patologia , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/patologia , Camundongos , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/patologia , Medição de Risco/métodos , Medição de Risco/estatística & dados numéricos , Testes de Toxicidade Crônica/métodos
9.
Int Immunopharmacol ; 98: 107830, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34118646

RESUMO

BACKGROUND: Colon cancer is a malignant condition that affects the lower gastrointestinal tract and has unfavorable prognosis. Its mechanisms range from enhanced production of reactive oxygen species, inflammatory changes in the colon microenvironment and affection of the apoptotic pathways. Due to the high incidence of resistance of colon cancer to the traditional chemotherapeutic agents, a need for finding safe/effective agents that can attenuate the malignant changes had emerged. OBJECTIVE: To investigate the possible immunomodulatory and antitumor effects of topiramate on azoxymethane-induced colon cancer in rats. METHODOLOGY: Fifty male Wistar rats were randomized into five equal groups as follows: Control; azoxymethane-induced colon cancer; azoxymethane + methyl cellulose; azoxymethane + topiramate small dose; and azoxymethane + topiramate large dose. The body weight gain, serum carcinoembryonic antigen (CEA), tissue antioxidant status, proinflammatory cytokines, vascular endothelial growth factor (VEGF), Nrf2/HO-1 content, p-AKT, mTOR, p38 MAP kinase, caspase 9, nerve growth factor beta and beclin-1 were measured. Also, parts of the colon had undergone histopathological and immunohistochemical evaluation. KEY FINDINGS: Topiramate improved the body weight gain, decreased serum CEA, augmented the antioxidant defenses in the colonic tissues with significant amelioration of the inflammatory changes, decline in tissue VEGF and p-AKT/mTOR/MAP kinase signaling and increased Nrf2/HO-1 content in a dose-dependent manner when compared to rats treated with azoxymethane alone. In addition, topiramate, in a dose-dependent manner, significantly enhanced apoptosis and improved the histopathological picture in comparison to animals treated with azoxymethane alone. CONCLUSION: Taking these findings together, topiramate might serve as a new effective adjuvant line of treatment of colon cancer.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Neoplasias Experimentais/tratamento farmacológico , Topiramato/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Azoximetano/administração & dosagem , Azoximetano/toxicidade , Carcinogênese/induzido quimicamente , Carcinogênese/efeitos dos fármacos , Carcinogênese/imunologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Serina-Treonina Quinases TOR/metabolismo , Topiramato/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
10.
Regul Toxicol Pharmacol ; 124: 104961, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34015422

RESUMO

Ethyl acrylate (EA) was classified by IARC as a Group-2B Carcinogen based, in part, on data suggesting increased incidence of thyroid neoplasia in rats and mice exposed chronically to EA vapors. We examined chronic exposure of rats and mice to EA vapors, evaluated the data on the incidence of thyroid follicular neoplasia, and determined the relevance of thyroid tumors to human health risk. The data revealed a small statistically significant increase in thyroid tumors in EA-exposed male rats and mice. The tumor incidences were within the range of historical controls and were not consistently dose-dependent. Most thyroid tumors in exposed animals were benign. Chronic exposure of EA to rats and mice (drinking water or gavage) and dogs (capsules) had no evidence of thyroid neoplasia. Results from chronic studies, in vivo and in vitro data, and ToxCastTM/Tox 21 HTPS did not support genotoxic/mutagenic potential for EA. This suggests that the associations between EA exposure and thyroid neoplasia represent chance or random observations rather than a compound-mediated effect. Due to species-specific physiological differences, the hypothalamic-pituitary-thyroid axis of rodents is more sensitive to endocrine disruptive chemicals than that of humans which further suggests that findings in rodents have questionable relevance to human health.


Assuntos
Acrilatos/toxicidade , Carcinógenos/toxicidade , Neoplasias Experimentais/induzido quimicamente , Neoplasias Gástricas/induzido quimicamente , Neoplasias da Glândula Tireoide/induzido quimicamente , Animais , Cães , Feminino , Humanos , Masculino , Camundongos , Neoplasias Experimentais/sangue , Neoplasias Experimentais/patologia , Ratos , Especificidade da Espécie , Estômago/efeitos dos fármacos , Estômago/patologia , Neoplasias Gástricas/patologia , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Hormônios Tireóideos/sangue , Hormônios Tireóideos/metabolismo , Neoplasias da Glândula Tireoide/sangue , Neoplasias da Glândula Tireoide/patologia , Testes de Toxicidade Crônica/estatística & dados numéricos
11.
Nat Commun ; 12(1): 2288, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863883

RESUMO

Hypothalamic tanycytes in median eminence (ME) are emerging as a crucial cell population that regulates endocrine output, energy balance and the diffusion of blood-born molecules. Tanycytes have recently been considered as potential somatic stem cells in the adult mammalian brain, but their regenerative and tumorigenic capacities are largely unknown. Here we found that Rax+ tanycytes in ME of mice are largely quiescent but quickly enter the cell cycle upon neural injury for self-renewal and regeneration. Mechanistically, Igf1r signaling in tanycytes is required for tissue repair under injury conditions. Furthermore, Braf oncogenic activation is sufficient to transform Rax+ tanycytes into actively dividing tumor cells that eventually develop into a papillary craniopharyngioma-like tumor. Together, these findings uncover the regenerative and tumorigenic potential of tanycytes. Our study offers insights into the properties of tanycytes, which may help to manipulate tanycyte biology for regulating hypothalamic function and investigate the pathogenesis of clinically relevant tumors.


Assuntos
Craniofaringioma/patologia , Células Ependimogliais/fisiologia , Eminência Mediana/fisiologia , Neoplasias Experimentais/patologia , Regeneração , Animais , Carcinogênese/patologia , Autorrenovação Celular/fisiologia , Craniofaringioma/induzido quimicamente , Craniofaringioma/genética , Proteínas do Olho/metabolismo , Feminino , Proteínas de Homeodomínio/metabolismo , Eminência Mediana/citologia , Camundongos , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Proteínas Proto-Oncogênicas B-raf/genética , RNA-Seq , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Análise de Célula Única , Fatores de Transcrição/metabolismo
12.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33762306

RESUMO

High levels of the intermediate filament protein keratin 17 (K17) are associated with poor prognoses for several human carcinomas. Studies in mouse models have shown that K17 expression is positively associated with growth, survival, and inflammation in skin and that lack of K17 delays onset of tumorigenesis. K17 occurs in the nucleus of human and mouse tumor keratinocytes where it impacts chromatin architecture, gene expression, and cell proliferation. We report here that K17 is induced following DNA damage and promotes keratinocyte survival. The presence of nuclear K17 is required at an early stage of the double-stranded break (DSB) arm of the DNA damage and repair (DDR) cascade, consistent with its ability to associate with key DDR effectors, including γ-H2A.X, 53BP1, and DNA-PKcs. Mice lacking K17 or with attenuated K17 nuclear import showed curtailed initiation in a two-step skin carcinogenesis paradigm. The impact of nuclear-localized K17 on DDR and cell survival provides a basis for the link between K17 induction and poor clinical outcomes for several human carcinomas.


Assuntos
Carcinoma/genética , Reparo do DNA , Queratina-17/metabolismo , Queratinas/metabolismo , Neoplasias Experimentais/genética , 9,10-Dimetil-1,2-benzantraceno/administração & dosagem , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Transporte Ativo do Núcleo Celular , Animais , Carcinogênese/induzido quimicamente , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma/induzido quimicamente , Carcinoma/patologia , Núcleo Celular/metabolismo , Sobrevivência Celular/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Feminino , Técnicas de Inativação de Genes , Células HeLa , Humanos , Microscopia Intravital , Queratina-17/genética , Queratinócitos , Queratinas/genética , Masculino , Camundongos Knockout , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/patologia , Imagem com Lapso de Tempo
13.
Hepatology ; 74(2): 797-815, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33650193

RESUMO

BACKGROUND AND AIMS: Intrahepatic cholangiocarcinoma (iCCA) is closely correlated with hepatic progenitor cell (HPC) expansion and liver fibrosis. Brahma-related gene 1 (Brg1), an enzymatic subunit of the switch/sucrose nonfermentable complex that is critical in stem cell maintenance and tumor promotion, is prominently up-regulated in both HPCs and iCCA; however, its role in this correlation remains undefined. APPROACH AND RESULTS: A retrospective cohort study indicated that high Brg1 expression suggests poor prognosis in patients with iCCA. In chronically injured livers induced by a 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine diet or bile duct ligation surgery, HPCs were dramatically activated, as indicated by their enhanced expression of Brg1 and a subset of stem cell markers; however, Brg1 ablation in HPCs strongly suppressed HPC expansion and liver fibrosis. Furthermore, in a chemically induced iCCA model, inhibition of Brg1 by a specific inhibitor or inducible gene ablation markedly improved histology and suppressed iCCA growth. Mechanistically, in addition to transcriptionally promoting both Wnt receptor genes and target genes, Brg1 was found to bind to the ß-catenin/transcription factor 4 transcription complex, suggesting a possible approach for regulation of Wnt/ß-catenin signaling. CONCLUSIONS: We have demonstrated the function of Brg1 in promoting HPC expansion, liver cirrhosis, and, ultimately, iCCA development in chronically injured livers, which is largely dependent on Wnt/ß-catenin signaling. Our data suggest that therapies targeting Brg1-expressing HPCs are promising for the treatment of liver cirrhosis and iCCA.


Assuntos
Neoplasias dos Ductos Biliares/genética , Colangiocarcinoma/genética , DNA Helicases/genética , Cirrose Hepática/genética , Recidiva Local de Neoplasia/epidemiologia , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Adulto , Idoso , Animais , Compostos Azabicíclicos/farmacologia , Compostos Azabicíclicos/uso terapêutico , Neoplasias dos Ductos Biliares/induzido quimicamente , Neoplasias dos Ductos Biliares/mortalidade , Neoplasias dos Ductos Biliares/terapia , Ductos Biliares Intra-Hepáticos/patologia , Ductos Biliares Intra-Hepáticos/cirurgia , Colangiocarcinoma/induzido quimicamente , Colangiocarcinoma/mortalidade , Colangiocarcinoma/terapia , DNA Helicases/antagonistas & inibidores , DNA Helicases/metabolismo , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fígado/patologia , Fígado/cirurgia , Cirrose Hepática/diagnóstico , Cirrose Hepática/patologia , Cirrose Hepática/prevenção & controle , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Prognóstico , Piridinas/farmacologia , Piridinas/uso terapêutico , Estudos Retrospectivos , Células-Tronco/metabolismo , Células-Tronco/patologia , Tioacetamida/administração & dosagem , Tioacetamida/toxicidade , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Regulação para Cima , Via de Sinalização Wnt/genética
14.
J Biochem Mol Toxicol ; 35(5): e22723, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33511709

RESUMO

Gastric carcinoma is one of the most aggressive types of cancer that ranks fifth among all cancer incidences and third in cancer mortality. As it exhibits a prolonged asymptomatic condition and high recurrence rate, it is a great challenge to treat gastric cancer. Traditional medicine that utilizes herbal phytochemicals to treat various diseases is a potent alternative for current allopathic treatment. Hence, we evaluated the potency of a phytochemical bilobalide for treating gastric cancer in in vitro and in vivo models. Bilobalide, a sesquiterpenoid, is present in the Ginkgo biloba plant that belongs to the family of Ginkgoaceae. The cytotoxicity effect of bilobalide was evaluated in both gastric cancer (AGS) cells and normal gastric epithelial cells. Apoptosis-inducing property of bilobalide against the AGS cell line was analyzed with different fluorescent staining techniques and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, and cell cycle analysis was carried out by flow cytometry. The in vivo studies were assessed with N-methyl-N-nitrosourea (MNU)-induced gastric cancer in rats. Serum-specific gastric markers were quantified and histopathological analysis of stomach tissue was performed. The expression of target-signaling molecules was analyzed by a reverse-transcription polymerase chain reaction. The in vitro results proved that bilobalide effectively suppressed the AGS cell growth and induced cell death by nuclear damage and apoptosis induction. The bilobalide treatment effectively arrested the cell cycle of AGS cells via inhibiting the PI3K-signaling pathway. Our in vivo results also confirmed that the bilobalide persuasively inhibited the MNU-induced gastric carcinoma via inhibiting the thioredoxin-fold family proteins and inflammatory markers' expression. Overall, our results authentically prove that bilobalide possesses therapeutic potency to cure gastric carcinoma.


Assuntos
Apoptose/efeitos dos fármacos , Bilobalídeos/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Experimentais/tratamento farmacológico , Extratos Vegetais/química , Neoplasias Gástricas/tratamento farmacológico , Animais , Bilobalídeos/química , Linhagem Celular Tumoral , Ginkgo biloba , Humanos , Masculino , Metilnitrosoureia/toxicidade , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Ratos , Ratos Wistar , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
15.
Oncol Rep ; 45(1): 202-216, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33416133

RESUMO

Long non­coding RNA growth arrest specific 5 (GAS5) exerts inhibitory effects through the modulation of several target microRNAs (miRs) in cancer. However, its potential roles and underlying relationship during colorectal cancer (CRC) progression are unclear. Therefore, we explored the role of the negative feedback loop formed by the GAS5/miR­34a axis and mammalian target of rapamycin/sirtuin 1 (mTOR/SIRT1) pathway on macroautophagy and apoptosis in CRC. Expression of GAS5, miR­34a, SIRT1 and mTOR in CRC patients and cell lines was detected by quantitative reverse transcription polymerase chain reaction. Online bioinformatic analysis was used to predict the downstream miRs of GAS5. Luciferase assay and western blotting were performed to demonstrate miR­34a as a downstream target gene of GAS5 in CRC cells. The effects of the GAS5/miR­34a axis on apoptosis, macroautophagy, and the mTOR/SIRT1 pathway were assessed by flow cytometry, transmission electron microscopy and western blotting, respectively. Our results suggested that GAS5 was downregulated and acted as a molecular sponge of miR­34a during CRC progression. miR­34a participated in regulating GAS5­suppressed CRC cell macroautophagy and induced apoptosis through the mTOR/SIRT1 pathway. GAS5­mediated macroautophagy was maintained in an equilibrium state that might have a protective effect on CRC cell apoptosis. The mTOR signaling pathway suppressed GAS5 expression and formed a negative regulation feedback loop with miR­34a in CRC cells. Our results suggested that the GAS5/miR­34a/SIRT1/mTOR negative regulatory feedback loop mediated CRC cell macroautophagy, and maintained the cells in an autonomous equilibrium state, but not excessive activation state, which functions as a strong antiapoptotic phenotype during human CRC progression.


Assuntos
Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Macroautofagia/genética , MicroRNAs/genética , RNA Longo não Codificante/metabolismo , Idoso , Animais , Azoximetano/administração & dosagem , Azoximetano/toxicidade , Linhagem Celular Tumoral , Colo/imunologia , Colo/patologia , Colo/cirurgia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Retroalimentação Fisiológica , Feminino , Humanos , Macroautofagia/efeitos dos fármacos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , RNA Longo não Codificante/genética , Ratos , Transdução de Sinais/genética , Sirolimo/farmacologia , Sirtuína 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo
16.
Mol Cell Biochem ; 476(4): 1765-1781, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33433833

RESUMO

NF-κB is the principle transcription factor and plays the central role in orchestrating chronic inflammation by regulating levels of cytokines, chemokines and growth factors. Piperlongumine (PL), a major alkaloid in the fruit of Piper longum Linn. has gained worldwide attention for its anticancer properties, however, its mechanism of action in the chemoprevention of colon cancer has not been investigated yet. Therefore, the present study was designed to elucidate the underlying molecular mechanism of PL in preventing DMH/DSS induced experimental colon cancer in mice. In the current study well established DMH/DSS induced experimental colon cancer mouse model was used to demonstrate the chemopreventive potential of PL. The expression of NF-κB and its downstream target proteins was evaluated mainly through western blotting. In addition, CAM assay, immunohistochemical staining and gelatin zymography was used to show anti-angiogenic and anti-invasive potential of PL. Additionally, important tumor biomarkers such as TSA, LASA, LDH and IL-6 levels were also estimated. The results of current study showed that PL was capable to inhibit NF-κB activation as well as its nuclear translocation. PL administration to DMH/DSS treated mice also inhibited the NF-κB downstream signaling cascades such as including COX-2 pathway, JAK/STAT pathway, ß-catenin, Notch signaling pathway, angiogenesis and epithelial to mesenchymal transition pathway. The findings of the present study have claimed PL as promising chemopreventive agent for colon cancer with pleiotropic action. The current study emphasizes that regular consumption of PL can be an effective approach in the prevention of colon cancer in humans.


Assuntos
Neoplasias do Colo/metabolismo , Dioxolanos/farmacologia , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/patologia
17.
Int J Oncol ; 58(2): 251-265, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33491745

RESUMO

The present study, to the best of our knowledge, is the first systematic study of the inhibitory effects of palmitoyl piperidinopiperidine (PPI; Japan Patent no. 5597427), on colon carcinogenesis. PPI exhibited marked growth inhibitory activity in several human colon carcinoma cell lines, with IC50 values of approximately 0.5­2.2 µM. In silico docking analysis indicated that PPI could bind to the SH2 domain of signal transducer and activator of transcription 3 (STAT3). PPI markedly inhibited the transcriptional activity of the SW837 cell line. Flowcytometric analysis demonstrated that PPI induced an increase in the number of cells in the G1 phase of the cell cycle, and induced sub­G1 fractions of cells at a higher concentration level of PPI. In the HT29 and SW837 cells, western blot analyses exhibited that in whole cell lysates, PPI induced a marked decrease in the expression levels of p­STAT3, but not in the levels of STAT3 in these cells. PPI also induced a marked decrease in the expression levels of both STAT3 and p­STAT3 in the chromatin fraction. In addition, PPI affected the protein expression levels of cyclin D1, p53, Bcl­2, Bcl­xL and vascular endothelial growth factor (VEGF). In the HT29 cells, PPI induced a marked and dose­dependent increase in the expression levels of Bax, cleaved caspase­3, cleaved caspase­7, cleaved caspase­8, cleaved caspase­9 and cleaved poly (ADP­ribose) polymerase (PARP). In animal model systems, PPI inhibited the growth of implanted carcinoma cells, and also induced a significant decrease in the multiplicity of colonic aberrant crypt foci. In addition, a marked and dose­dependent inhibition of angiogenesis of the chick chorioallantoic membrane was observed. As regards the possible molecular mechanisms, it is suggested that the inhibition of STAT3 by PPI may affect the function of molecules that are related to apoptosis, angiogenesis and cell cycle progression, eventually contributing to the PPI­induced growth inhibitory effects.


Assuntos
Antineoplásicos/farmacologia , Carcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Ácidos Graxos Monoinsaturados/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Azoximetano/administração & dosagem , Azoximetano/toxicidade , Carcinógenos/administração & dosagem , Carcinógenos/toxicidade , Carcinoma/patologia , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide , Neoplasias do Colo/patologia , Ácidos Graxos Monoinsaturados/química , Ácidos Graxos Monoinsaturados/uso terapêutico , Feminino , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Simulação de Acoplamento Molecular , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/patologia , Neovascularização Patológica/dietoterapia , Neovascularização Patológica/patologia , Ratos , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Food Funct ; 12(3): 1063-1078, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33443517

RESUMO

According to population-based studies, lung cancer has become one of the leading causes of death globally in males and is also rising in females at an alarming rate. The aim of this study was to exploit the inherent properties of eugenol to restrict the growth of cancer cells in a tobacco-related human carcinogen NDEA-induced lung carcinogenesis model in vivo as a chemopreventive agent. More precisely, by utilizing its abundance in nature, eugenol (a component of clove) was utilized to establish the molecular mechanism of chemoprevention in the NDEA-induced mouse lung carcinogenesis model in a substantial cost-effective manner and was validated in the A549 human lung cancer cell line. Our study especially targeted the tiny, drug-resistant, and most virulent subpopulation of cancer cells called CSCs by targeting their regulator molecule ß-catenin. The non-toxic dosage of eugenol was shown to enhance apoptosis, simultaneously suppressing cell proliferation in the lung tissue of carcinogen-treated mice without affecting the normal mice. Combining cellular apoptosis and proliferation, eugenol showed an exceptional chemopreventive potential in this lung carcinogenesis model. Importantly, eugenol strongly restricted the lung carcinoma in the mild dysplastic stage as a chemopreventive agent. The molecular analysis remarkably depicted the restriction of ß-catenin nuclear transportation. The minimized total ß-catenin pool and induced N-terminal Ser37 phosphorylation form after eugenol treatment resulted in its cytoplasmic degradation. Consequently, CSC markers such as CD44, Oct4, EpCAM, and Notcht1, whose expression is dependent on ß-catenin decreased significantly, as proven by IHC, ICC, and WB analysis both in vivo and in vitro. The in vitro secondary sphere formation assay also proved the remarkably repressed CSC population, and hence the virulence. In another way, eugenol was proven to significantly enhance the degradation of ß-catenin when treated with the CK1α inhibitor D4476 in vitro by Western blot. CK1α in the Wnt/ß-catenin pathway plays a crucial role for tagging with the N-terminal Ser45 phosphorylation of ß-catenin, which ultimately opens a position for the decisive phosphorylation by GSK3ß at the Ser37 residue to take place. Thus, the conclusive extermination of CSCs achieved that was associated with recurrence due to treatment failure. That can help to achieve a longer and better quality of life in a natural, economical way.


Assuntos
Eugenol/farmacologia , beta Catenina/metabolismo , Células A549 , Animais , Apoptose , Dietilnitrosamina/toxicidade , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/tratamento farmacológico , beta Catenina/genética
19.
Cell Mol Gastroenterol Hepatol ; 11(2): 639-658, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33091622

RESUMO

BACKGROUND & AIMS: The fragile X mental retardation protein (FMRP) affects multiple steps of the mRNA metabolism during brain development and in different neoplastic processes. However, the contribution of FMRP in colon carcinogenesis has not been investigated. METHODS: FMR1 mRNA transcript and FMRP protein expression were analyzed in human colon samples derived from patients with sporadic colorectal cancer (CRC) and healthy subjects. We used a well-established mouse model of sporadic CRC induced by azoxymethane to determine the possible role of FMRP in CRC. To address whether FMRP controls cancer cell survival, we analyzed cell death pathway in CRC human epithelial cell lines and in patient-derived colon cancer organoids in presence or absence of a specific FMR1 antisense oligonucleotide or siRNA. RESULTS: We document a significant increase of FMRP in human CRC relative to non-tumor tissues. Next, using an inducible mouse model of CRC, we observed a reduction of colonic tumor incidence and size in the Fmr1 knockout mice. The abrogation of FMRP induced spontaneous cell death in human CRC cell lines activating the necroptotic pathway. Indeed, specific immunoprecipitation experiments on human cell lines and CRC samples indicated that FMRP binds receptor-interacting protein kinase 1 (RIPK1) mRNA, suggesting that FMRP acts as a regulator of necroptosis pathway through the surveillance of RIPK1 mRNA metabolism. Treatment of human CRC cell lines and patient-derived colon cancer organoids with the FMR1 antisense resulted in up-regulation of RIPK1. CONCLUSIONS: Altogether, these data support a role for FMRP  in controlling RIPK1 expression and necroptotic activation in CRC.


Assuntos
Neoplasias Colorretais/genética , Proteína do X Frágil de Retardo Mental/metabolismo , Recidiva Local de Neoplasia/epidemiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Animais , Azoximetano/administração & dosagem , Azoximetano/toxicidade , Carcinogênese/genética , Estudos de Casos e Controles , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Colo/patologia , Colo/cirurgia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/cirurgia , Conjuntos de Dados como Assunto , Intervalo Livre de Doença , Proteína do X Frágil de Retardo Mental/antagonistas & inibidores , Proteína do X Frágil de Retardo Mental/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Voluntários Saudáveis , Humanos , Masculino , Camundongos , Camundongos Knockout , Necroptose/genética , Recidiva Local de Neoplasia/genética , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Organoides , Prognóstico
20.
Gastroenterology ; 160(4): 1179-1193.e14, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32920015

RESUMO

BACKGROUND & AIMS: Streptococcus thermophilus was identified to be depleted in patients with colorectal cancer (CRC) by shotgun metagenomic sequencing of 526 multicohort fecal samples. Here, we aim to investigate whether this bacterium could act as a prophylactic for CRC prevention. METHODS: The antitumor effects of S thermophilus were assessed in cultured colonic epithelial cells and in 2 murine models of intestinal tumorigenesis. The tumor-suppressive protein produced by S thermophilus was identified by mass spectrometry and followed by ß-galactosidase activity assay. The mutant strain of S thermophilus was constructed by homologous recombination. The effect of S thermophilus on the gut microbiota composition was assessed by shotgun metagenomic sequencing. RESULTS: Oral gavage of S thermophilus significantly reduced tumor formation in both Apcmin/+ and azoxymethane-injected mice. Coincubation with S thermophilus or its conditioned medium decreased the proliferation of cultured CRC cells. ß-Galactosidase was identified as the critical protein produced by S thermophilus by mass spectrometry screening and ß-galactosidase activity assay. ß-Galactosidase secreted by S thermophilus inhibited cell proliferation, lowered colony formation, induced cell cycle arrest, and promoted apoptosis of cultured CRC cells and retarded the growth of CRC xenograft. The mutant S thermophilus without functional ß-galactosidase lost its tumor-suppressive effect. Also, S thermophilus increased the gut abundance of known probiotics, including Bifidobacterium and Lactobacillus via ß-galactosidase. ß-Galactosidase-dependent production of galactose interfered with energy homeostasis to activate oxidative phosphorylation and downregulate the Hippo pathway kinases, which partially mediated the anticancer effects of S thermophilus. CONCLUSION: S thermophilus is a novel prophylactic for CRC prevention in mice. The tumor-suppressive effect of S thermophilus is mediated at least by the secretion of ß-galactosidase.


Assuntos
Proteínas de Bactérias/metabolismo , Neoplasias Colorretais/prevenção & controle , Probióticos/administração & dosagem , Streptococcus thermophilus/enzimologia , beta-Galactosidase/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Azoximetano/administração & dosagem , Azoximetano/toxicidade , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/induzido quimicamente , Colo/microbiologia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia , Humanos , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Experimentais/microbiologia , Neoplasias Experimentais/prevenção & controle , Probióticos/metabolismo , Streptococcus thermophilus/genética , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...