Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 362
Filtrar
2.
Br J Cancer ; 126(10): 1421-1431, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35091695

RESUMO

BACKGROUND: Gut microbiota dysbiosis is closely related to the progression of colorectal cancer. Our previous study revealed that early life colonisation with Lactobacillus rhamnosus GG (LGG) had long-term positive effects on health. We sought to investigate whether early life LGG colonisation could inhibit intestinal tumour formation in offspring. METHODS: Adult C57BL/6 female mice were mated with Apcmin/+ male mice. Pregnant mice with the same conception date received 108 cfu live or fixed LGG from day 18 of pregnancy until natural delivery. After genotyping, offspring mice received 107 cfu of live or fixed LGG for 0-5 days after birth. RESULTS: Early life LGG colonisation significantly promoted intestinal development, inhibited low-grade intestinal inflammation and altered the gut microbiota composition of offspring in the weaning period (3 week old). Notably, early life LGG colonisation reduced the multiplicity of intestinal tumours in adulthood (12 week old), possibly due to inhibition of Wnt signalling and promotion of tumour cell apoptosis. Importantly, at the genus level, Bifidobacterium and Anaeroplasma with potential anti-tumour effects were increased in adulthood, while Peptostreptococcus, which potentially contributes to tumour formation, was decreased. CONCLUSIONS: Early life LGG colonisation inhibited the intestinal tumour formation of offspring in adulthood.


Assuntos
Microbioma Gastrointestinal , Neoplasias Intestinais , Lacticaseibacillus rhamnosus , Probióticos , Adulto , Animais , Feminino , Humanos , Neoplasias Intestinais/prevenção & controle , Lacticaseibacillus rhamnosus/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Probióticos/farmacologia
3.
Recurso na Internet em Inglês, Português | LIS - Localizador de Informação em Saúde, LIS-controlecancer | ID: lis-48556

RESUMO

Quase 30% de todos os cânceres colorretais (cânceres de intestino) podem ser evitados com alimentação saudável, prática de atividades físicas e abandono de bebidas alcoólicas. Essas ações preventivas são importantes para redução de casos e mortes, para a manutenção da produtividade laboral e para a redução dos gastos públicos com o tratamento do câncer de intestino.


Assuntos
Neoplasias Intestinais/epidemiologia , Neoplasias Intestinais/prevenção & controle , Neoplasias Intestinais/etiologia , Dieta Saudável
4.
Bioengineered ; 12(2): 12383-12393, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34895063

RESUMO

The potential of antibodies, especially for the bispecific antibodies, are limited by high cost and complex technical process of development and manufacturing. A cost-effective and rapid platform for the endogenous antibodies expression via using the in vitro transcription (IVT) technique to produce nucleoside-modified mRNA and then encapsulated into lipid nanoparticle (LNP) may turn the body to a manufactory. Coinhibitory pathway of programmed death ligand 1 (PD-L1) and programmed cell death protein 1 receptor (PD-1) could suppress the T-cell mediated immunity. We hypothesized that the coblocking of PD-L1 and PD-1 via bispecific antibodies may achieve more potential antitumor efficacies compare with the monospecific ones. Here, we described the application of mRNA to encode a bispecific antibody with ablated Fc immune effector functions that targets both human PD-L1 and PD-1, termed XA-1, which was further assessed the in vitro functional activities and in vivo antitumor efficacies. The in vitro mRNA-encoded XA-1 held comparable abilities to fully block the PD-1/PD-L1 pathway as well as to enhance functional T cell activation compared to XA-1 protein from CHO cell source. Pharmacokinetic tests showed enhanced area under curve (AUC) of mRNA-encoded XA-1 compared with XA-1 at same dose. Chronic treatment of LNP-encapsulated XA-1 mRNA in the mouse tumor models which were reconstituted with human immune cells effectively induced promising antitumor efficacies compared to XA-1 protein. Current results collectively demonstrated that LNP-encapsulated mRNA represents the viable delivery platform for treating cancer and hold potential to be applied in the treatment of many diseases.Abbreviations: IVT: in vitro transcription; LNP: lipid nanoparticle; hPD-1: human PD-1; hPD-L1: human PD-L1; ITS-G: Insulin-Transferrin-Selenium; Pen/Strep: penicillin-streptomycin; FBS: fetal bovine serum; TGI: tumor growth inhibition; IE1: cytomegalovirus immediate early 1; SP: signal peptide; hIgLC: human immunoglobulin kappa light chain; hIgHC: human IgG1 heavy chain; AUC: area under the curve; Cl: serum clearance; Vss: steady-state distributed volume; MLR: mixed lymphocyte reaction.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Neoplasias Intestinais/prevenção & controle , Lipossomos/administração & dosagem , Nanopartículas/administração & dosagem , RNA Mensageiro/administração & dosagem , Animais , Antígeno B7-H1/metabolismo , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Cricetulus , Modelos Animais de Doenças , Feminino , Humanos , Neoplasias Intestinais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
5.
Cancer Prev Res (Phila) ; 14(9): 851-862, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34266857

RESUMO

Familial adenomatous polyposis (FAP) is a hereditary colorectal cancer syndrome, which results in the development of hundreds of adenomatous polyps carpeting the gastrointestinal tract. NSAIDs have reduced polyp burden in patients with FAP and synthetic rexinoids have demonstrated the ability to modulate cytokine-mediated inflammation and WNT signaling. This study examined the use of the combination of an NSAID (sulindac) and a rexinoid (bexarotene) as a durable approach for reducing FAP colonic polyposis to prevent colorectal cancer development. Whole transcriptomic analysis of colorectal polyps and matched normal mucosa in a cohort of patients with FAP to identify potential targets for prevention in FAP was performed. Drug-dose synergism of sulindac and bexarotene in cell lines and patient-derived organoids was assessed, and the drug combination was tested in two different mouse models. This work explored mRNA as a potential predictive serum biomarker for this combination in FAP. Overall, transcriptomic analysis revealed significant activation of inflammatory and cell proliferation pathways. A synergistic effect of sulindac (300 µmol/L) and bexarotene (40 µmol/L) was observed in FAP colonic organoids with primary targeting of polyp tissue compared with normal mucosa. This combination translated into a significant reduction in polyp development in ApcMin/+ and ApcLoxP/+-Cdx2 mice. Finally, the reported data suggest miRNA-21 could serve as a predictive serum biomarker for polyposis burden in patients with FAP. These findings support the clinical development of the combination of sulindac and bexarotene as a treatment modality for patients with FAP. PREVENTION RELEVANCE: This study identified a novel chemopreventive regimen combining sulindac and bexarotene to reduce polyposis in patients with FAP using in silico tools, ex vivo, and in vivo models. This investigation provides the essential groundwork for moving this drug combination forward into a clinical trial.


Assuntos
Polipose Adenomatosa do Colo/tratamento farmacológico , Bexaroteno/administração & dosagem , Neoplasias Intestinais/prevenção & controle , Sulindaco/administração & dosagem , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Pólipos Adenomatosos/tratamento farmacológico , Pólipos Adenomatosos/genética , Pólipos Adenomatosos/patologia , Adulto , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Estudos de Casos e Controles , Células Cultivadas , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Células HT29 , Humanos , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Transgênicos
6.
Cell Mol Gastroenterol Hepatol ; 11(5): 1483-1503, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33515805

RESUMO

BACKGROUND & AIMS: The reason why small intestinal cancer is rarer than colorectal cancer is not clear. We hypothesized that intraepithelial lymphocytes (IELs), which are enriched in the small intestine, are the closest immune cells to epithelial cells, exclude tumor cells via cell-to-cell contact. METHODS: We developed DPE-green fluorescent protein (DPE-GFP) × adenomatous polyposis coli; multiple intestinal neoplasia (APCmin ) mice, which is a T-cell-reporter mouse with spontaneous intestinal tumors. We visualized the dynamics of IELs in the intestinal tumor microenvironment and the interaction between IELs and epithelial cells, and the roles of cell-to-cell contact in anti-intestinal tumor immunity using a novel in vivo live-imaging system and a novel in vitro co-culture system. RESULTS: In the small intestinal tumor microenvironment, T-cell movement was restricted around blood vessels and the frequency of interaction between IELs and epithelial cells was reduced. Genetic deletion of CD103 decreased the frequency of interaction between IELs and epithelial cells, and increased the number of small intestinal tumors. In the co-culture system, wild-type IELs expanded and infiltrated to intestinal tumor organoids from APCmin mice and reduced the viability of them, which was cell-to-cell contact and CD103 dependent. CONCLUSIONS: The abundance of IELs in the small intestine may contribute to a low number of tumors, although this system may not work in the colon because of the sparseness of IELs. Strategies to increase the number of IELs in the colon or enhance cell-to-cell contact between IELs and epithelial cells may be effective for the prevention of intestinal tumors in patients with a high cancer risk.


Assuntos
Antígenos CD/fisiologia , Comunicação Celular , Cadeias alfa de Integrinas/fisiologia , Mucosa Intestinal/imunologia , Neoplasias Intestinais/prevenção & controle , Intestino Delgado/imunologia , Linfócitos Intraepiteliais/imunologia , Microambiente Tumoral , Animais , Técnicas de Cocultura , Feminino , Mucosa Intestinal/citologia , Neoplasias Intestinais/imunologia , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Intestino Delgado/patologia , Linfócitos Intraepiteliais/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organoides/imunologia , Organoides/patologia
7.
Oncogene ; 40(2): 369-383, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33144683

RESUMO

The Wnt/ß-catenin signaling pathway is aberrantly activated in the majority of colorectal cancer cases due to somatic mutations in the adenomatous polyposis coli (APC) gene. Prohibitin 1 (PHB1) serves pleiotropic cellular functions with dynamic subcellular trafficking, facilitating signaling crosstalk between organelles. Nuclear-localized PHB1 is an important regulator of gene transcription. Using mice with inducible intestinal epithelial cell (IEC)-specific deletion of Phb1 (Phb1iΔIEC) and mice with IEC-specific overexpression of Phb1 (Phb1Tg), we demonstrate that IEC-specific PHB1 combats intestinal tumorigenesis in the ApcMin/+ mouse model by inhibiting Wnt/ß-catenin signaling. Forced nuclear accumulation of PHB1 in human RKO or SW48 CRC cell lines increased AXIN1 expression and decreased cell viability. PHB1 deficiency in CRC cells decreased AXIN1 expression and increased ß-catenin activation that was abolished by XAV939, a pharmacological AXIN stabilizer. These results define a role of PHB1 in inhibiting the Wnt/ß-catenin pathway to influence the development of intestinal tumorigenesis. Induction of nuclear PHB1 trafficking provides a novel therapeutic option to influence AXIN1 expression and the ß-catenin destruction complex in Wnt-driven intestinal tumorigenesis.


Assuntos
Carcinogênese , Núcleo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Intestinais/prevenção & controle , Proteínas Repressoras/fisiologia , Proteínas Wnt/antagonistas & inibidores , beta Catenina/antagonistas & inibidores , Animais , Apoptose , Proteína Axina/genética , Proteína Axina/metabolismo , Proliferação de Células , Feminino , Humanos , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proibitinas , Células Tumorais Cultivadas , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
8.
Int Immunopharmacol ; 90: 107128, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33191180

RESUMO

Sunitinib is a tyrosine kinase inhibitor for many tumors. Inflammation is one of the most important factors in the development of intestinal tumors. Many inflammation-related factors are regulated by tyrosine kinase receptors. It is reasonable to hypothesize that sunitinib can regulate the development of intestinal tumors by regulating the expression and/or activity of inflammation-related factors. Here, ApcMin/+ male mouse model was used to investigate the effect and mechanism of sunitinib malate against intestinal cancer. Results show that compared to vehicle, after sunitinib malate treatment, overall survival of ApcMin/+ mice was lengthened up to 25 days, with a gain of body weight, reduction of spleen/body weight index, and RBC, WBC and HGC regulated to normal levels of wild type mice, and a number of polyps no less than 1 mm significantly reduced. Meanwhile, in the intestines, the nuclear ß-Catenin protein and c-Myc mRNA were both down-regulated, and Bcl-6 was significantly reduced with Caspase-3 up regulated. Furthermore, inflammation-related factors including IL-6, TNF-α, IL-1α, IL-1ß and IFN-γ were down-regulated at mRNA levels in the intestines. These results suggest that sunitinib malate can significantly improve the survival status and inhibit intestinal tumor development in male ApcMin/+ mice, through inhibiting inflammation-related factors, while suppressing ß-cateinin/c-Myc pathway and re-balancing protein levels of Bcl-6 and Caspase-3.


Assuntos
Anti-Inflamatórios/farmacologia , Anticarcinógenos/farmacologia , Caspase 3/metabolismo , Colo/efeitos dos fármacos , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Neoplasias Intestinais/prevenção & controle , Pólipos Intestinais/prevenção & controle , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sunitinibe/farmacologia , beta Catenina/metabolismo , Animais , Colo/enzimologia , Colo/patologia , Citocinas/genética , Regulação da Expressão Gênica , Genes APC , Neoplasias Intestinais/enzimologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Pólipos Intestinais/enzimologia , Pólipos Intestinais/genética , Pólipos Intestinais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/genética , Transdução de Sinais
9.
Cancer Prev Res (Phila) ; 14(3): 325-336, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33277315

RESUMO

A clinical trial in patients with familial adenomatous polyposis (FAP) demonstrated that sulindac plus erlotinib (SUL+ERL) had good efficacy in the duodenum and colon, but toxicity issues raised concerns for long-term prevention. We performed a biomarker study in the polyposis in rat colon (Pirc) model, observing phosphorylated Erk inhibition in colon polyps for up to 10 days after discontinuing ERL+SUL administration. In a follow-up study lasting 16 weeks, significant reduction of colon and small intestine (SI) tumor burden was detected, especially in rats given 250 ppm SUL in the diet plus once-a-week intragastric dosing of ERL at 21 or 42 mg/kg body weight (BW). A long-term study further demonstrated antitumor efficacy in the colon and SI at 52 weeks, when 250 ppm SUL was combined with once-a-week intragastric administration of ERL at 10, 21, or 42 mg/kg BW. Tumor-associated matrix metalloproteinase-7 (Mmp7), tumor necrosis factor (Tnf), and early growth response 1 (Egr1) were decreased at 16 weeks by ERL+SUL, and this was sustained in the long-term study for Mmp7 and Tnf. Based on the collective results, the optimal dose combination of ERL 10 mg/kg BW plus 250 ppm SUL lacked toxicity, inhibited molecular biomarkers, and exhibited effective antitumor activity. We conclude that switching from continuous to once-per-week ERL, given at one-quarter of the current therapeutic dose, will exert good efficacy with standard-of-care SUL against adenomatous polyps in the colon and SI, with clinical relevance for patients with FAP before or after colectomy. PREVENTION RELEVANCE: This investigation concludes that switching from continuous to once-per-week erlotinib, given at one-quarter of the current therapeutic dose, will exert good efficacy with standard-of-care sulindac against adenomatous polyps in the colon and small intestine, with clinical relevance for patients with FAP before or after colectomy.


Assuntos
Polipose Adenomatosa do Colo/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias do Colo/prevenção & controle , Pólipos do Colo/prevenção & controle , Genes APC , Neoplasias Intestinais/prevenção & controle , Mutação , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/normas , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Pólipos do Colo/genética , Pólipos do Colo/metabolismo , Pólipos do Colo/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Cloridrato de Erlotinib/administração & dosagem , Neoplasias Intestinais/genética , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Masculino , Ratos , Sulindaco/administração & dosagem
10.
Nutrients ; 12(7)2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32630805

RESUMO

Inflammatory bowel disease (IBD), which affects millions of people worldwide, includes two separate diseases: Crohn's disease (CD) and ulcerative colitis (UC). Although the background (chronic inflammatory state) and some of the symptoms of CD and UC are similar, both diseases differ from each other. It is becoming clear that a combination of many factors, in particular genetic background, host immune response and microbial reduced diversity status are associated with IBD. One potential strategy to prevent/treat IBD is gut modulation by probiotics. Over the last twenty years, many publications have focused on the role of probiotics in the course of IBD. The review discusses the utility of different strains of probiotics, especially Bifidobacterium spp., in all factors potentially involved in the etiology of IBD. The probiotic modulatory properties among different study models (cell lines, animal models of colitis, clinical study) are discussed and probiotic usefulness is assessed in relation to the treatment, prevention, and remission of diseases.


Assuntos
Doenças Inflamatórias Intestinais/terapia , Probióticos/uso terapêutico , Animais , Bifidobacterium/fisiologia , Colite/terapia , Colite Ulcerativa/terapia , Doença de Crohn/terapia , Modelos Animais de Doenças , Microbioma Gastrointestinal/fisiologia , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Neoplasias Intestinais/prevenção & controle
11.
Cancer Lett ; 469: 456-467, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31734354

RESUMO

Gut microbiota dysbiosis is closely involved in intestinal carcinogenesis. A marked reduction in butyrate-producing bacteria has been observed in patients with colorectal cancer (CRC); nevertheless, the potential benefit of butyrate-producing bacteria against intestinal tumor development has not been fully investigated. We found that Clostridium butyricum (C. butyricum, one of the commonly used butyrate-producing bacteria in clinical settings) significantly inhibited high-fat diet (HFD)-induced intestinal tumor development in Apcmin/+ mice. Moreover, intestinal tumor cells treated with C. butyricum exhibited decreased proliferation and increased apoptosis. Additionally, C. butyricum suppressed the Wnt/ß-catenin signaling pathway and modulated the gut microbiota composition, as demonstrated by decreases in some pathogenic bacteria and bile acid (BA)-biotransforming bacteria and increases in some beneficial bacteria, including short-chain fatty acid (SCFA)-producing bacteria. Accordingly, C. butyricum decreased the fecal secondary BA contents, increased the cecal SCFA quantities, and activated G-protein coupled receptors (GPRs), such as GPR43 and GPR109A. The anti-proliferative effect of C. butyricum was blunted by GPR43 gene silencing using small interfering RNA (siRNA). The analysis of clinical specimens revealed that the expression of GPR43 and GPR109A gradually decreased from human normal colonic tissue to adenoma to carcinoma. Together, our results show that C. butyricum can inhibit intestinal tumor development by modulating Wnt signaling and gut microbiota and thus suggest the potential efficacy of butyrate-producing bacteria against CRC.


Assuntos
Clostridium butyricum/metabolismo , Neoplasias Intestinais/metabolismo , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/genética , Butiratos/metabolismo , Proliferação de Células/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos Voláteis/biossíntese , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Neoplasias Intestinais/microbiologia , Neoplasias Intestinais/prevenção & controle , Probióticos/metabolismo , Probióticos/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
12.
Mol Nutr Food Res ; 63(4): e1800824, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30447137

RESUMO

SCOPE: The previous study shows that obesity-promoted inflammation is responsible for the activation of the intestinal tumorigenic Wnt-signaling. The present study aims to test a dietary strategy, dietary supplementation with a high dose of vitamin D (VD) or its combination with sulforaphane (SFN) to inhibit intestinal inflammation and obesity-associated tumorigenesis. METHODS AND RESULTS: Apc1638N mice are randomly divided into four groups: LF, a low-fat diet (10 kcal% fat) with 200 IU VD; HF, a high-fat diet (60 kcal% fat) with 200 IU VD; HFD, a high-fat diet with 5000 IU VD; and HFDS, a high-fat diet plus 5000 IU VD and 0.23 g SFN per ≈4000 kcal. VD administration decreased tumor incidence and size, and the co-administration with SFN (HFDS) magnified the effects. Inflammation and Wnt-signaling are suppressed by VD. The addition of SFN decreased the activity of histone deacetylase (HDAC) and increased autophagy. CONCLUSION: The administration of VD, at 5000 IU level, exerts an anti-inflammatory property and leads to suppressed intestinal Wnt-signaling and tumorigenesis in obese mice. The molecular function of SFN on a high dose of VD supplementation, although displayed on the inhibition of HDAC and the activation of autophagy, needs further investigation.


Assuntos
Antineoplásicos/farmacologia , Dieta Hiperlipídica/efeitos adversos , Neoplasias Intestinais/prevenção & controle , Isotiocianatos/farmacologia , Vitamina D/farmacologia , Animais , Anticarcinógenos/farmacologia , Autofagia/efeitos dos fármacos , Enterite/prevenção & controle , Feminino , Histona Desacetilases/metabolismo , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Masculino , Camundongos Mutantes , Obesidade/complicações , Sulfóxidos , Vitamina D/análogos & derivados , Vitamina D/sangue , beta Catenina/metabolismo
13.
Gastroenterology ; 156(1): 145-159.e19, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30273559

RESUMO

BACKGROUND & AIMS: RNase H2 is a holoenzyme, composed of 3 subunits (ribonuclease H2 subunits A, B, and C), that cleaves RNA:DNA hybrids and removes mis-incorporated ribonucleotides from genomic DNA through ribonucleotide excision repair. Ribonucleotide incorporation by eukaryotic DNA polymerases occurs during every round of genome duplication and produces the most frequent type of naturally occurring DNA lesion. We investigated whether intestinal epithelial proliferation requires RNase H2 function and whether RNase H2 activity is disrupted during intestinal carcinogenesis. METHODS: We generated mice with epithelial-specific deletion of ribonuclease H2 subunit B (H2bΔIEC) and mice that also had deletion of tumor-suppressor protein p53 (H2b/p53ΔIEC); we compared phenotypes with those of littermate H2bfl/fl or H2b/p53fl/fl (control) mice at young and old ages. Intestinal tissues were collected and analyzed by histology. We isolated epithelial cells, generated intestinal organoids, and performed RNA sequence analyses. Mutation signatures of spontaneous tumors from H2b/p53ΔIEC mice were characterized by exome sequencing. We collected colorectal tumor specimens from 467 patients, measured levels of ribonuclease H2 subunit B, and associated these with patient survival times and transcriptome data. RESULTS: The H2bΔIEC mice had DNA damage to intestinal epithelial cells and proliferative exhaustion of the intestinal stem cell compartment compared with controls and H2b/p53ΔIEC mice. However, H2b/p53ΔIEC mice spontaneously developed small intestine and colon carcinomas. DNA from these tumors contained T>G base substitutions at GTG trinucleotides. Analyses of transcriptomes of human colorectal tumors associated lower levels of RNase H2 with shorter survival times. CONCLUSIONS: In analyses of mice with disruption of the ribonuclease H2 subunit B gene and colorectal tumors from patients, we provide evidence that RNase H2 functions as a colorectal tumor suppressor. H2b/p53ΔIEC mice can be used to study the roles of RNase H2 in tissue-specific carcinogenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Células Epiteliais/enzimologia , Instabilidade Genômica , Neoplasias Intestinais/prevenção & controle , Intestino Delgado/enzimologia , Ribonuclease H/metabolismo , Animais , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Colite/induzido quimicamente , Colite/enzimologia , Colite/genética , Colite/patologia , Dano ao DNA , Sulfato de Dextrana , Modelos Animais de Doenças , Células Epiteliais/patologia , Feminino , Predisposição Genética para Doença , Humanos , Neoplasias Intestinais/enzimologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Intestino Delgado/patologia , Masculino , Camundongos Knockout , Fenótipo , Ribonuclease H/deficiência , Ribonuclease H/genética , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
14.
World J Gastroenterol ; 24(37): 4254-4262, 2018 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-30310258

RESUMO

AIM: To investigate the effects of VSL#3 on tumor formation, and fecal and intestinal mucosal microbiota in azoxymethane/dextran sulfate sodium (AOM/DSS) induced mice model. METHODS: C57BL/6 mice were administered AOM/DSS to develop the ulcerative colitis (UC) carcinogenesis model. Mice were treated with 5-ASA (75 mg/kg/d), VSL#3 (1.5 × 109 CFU/d), or 5-ASA combined with VSL#3 by gavage from the day of AOM injection for three months (five days/week). The tumor load was compared in each group, and tumor necrosis factor (TNF-α) and interleukin (IL)-6 levels were evaluated in colon tissue. The stool and intestinal mucosa samples were collected to analyze the differences in the intestinal microbiota by 16s rDNA sequencing method. RESULTS: VSL#3 significantly reduced the tumor load in AOM/DSS-induced mice model and decreased the level of TNF-α and IL-6 in colon tissue. The model group had a lower level of Lactobacillus and higher level of Oscillibacter and Lachnoclostridium in fecal microbiota than the control group. After the intervention with 5-ASA and VSL#3, Bacillus and Lactococcus were increased, while Lachnoclostridium and Oscillibacter were reduced. 5-ASA combined with VSL#3 increased the Lactobacillus and decreased the Oscillibacter. The intestinal mucosal microbiota analysis showed a lower level of Bifidobacterium and Ruminococcaceae_UCG-014 and higher level of Alloprevotella in the model group as compared to the control group. After supplementation with VSL#3, Bifidobacterium was increased. 5-ASA combined with VSL#3 increased the level of both Lachnoclostridium and Bifidobacterium. CONCLUSION: VSL#3 can prevent UC-associated carcinogenesis in mice, reduce the colonic mucosal inflammation levels, and rebalance the fecal and mucosal intestinal microbiota.


Assuntos
Colite Ulcerativa/complicações , Microbioma Gastrointestinal , Neoplasias Intestinais/prevenção & controle , Probióticos/uso terapêutico , Animais , Bifidobacterium , Carcinogênese , Colite Ulcerativa/metabolismo , Colo/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Interleucina-6/metabolismo , Neoplasias Intestinais/complicações , Lactobacillus , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S/isolamento & purificação , Ruminococcus , Análise de Sequência de DNA , Fator de Necrose Tumoral alfa/metabolismo
15.
Aust J Gen Pract ; 47(6): 343-349, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29966179

RESUMO

BACKGROUND AND OBJECTIVES: There is significant growth in demand for colonoscopies, with over 700,000 performed in Australia in 2012-13. For every one million Australians aged 50 years and older, 80,000 people at average risk of colorectal cancer are being over-screened with colonoscopy, and 29,000 people at increased risk are not having the colonoscopy they need. METHOD: Using monitoring data from the Australian National Bowel Cancer Screening Program and published data on colonoscopic screening, we have developed expected frequency trees (EFTs) to demonstrate projected outcomes of different colorectal cancer screening options for participants at different levels of colorectal cancer risk in Australia. RESULTS: The EFTs highlight the overall balance in favour of faecal occult blood screening for those at average risk in terms of fewer deaths and complications. DISCUSSION: This novel method of risk communication can be used to promote appropriate patient choice of colorectal cancer screening modality and potentially reduce the number of referrals for colonoscopy in patients who are not at increased risk of colorectal cancer.


Assuntos
Colonoscopia/psicologia , Educação de Pacientes como Assunto/métodos , Colonoscopia/normas , Detecção Precoce de Câncer/métodos , Detecção Precoce de Câncer/normas , Humanos , Neoplasias Intestinais/diagnóstico , Neoplasias Intestinais/prevenção & controle , Neoplasias Intestinais/psicologia , Programas de Rastreamento/métodos , Programas de Rastreamento/normas , Medição de Risco/métodos
16.
Cancer Prev Res (Phila) ; 11(2): 81-92, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29301746

RESUMO

The cGMP signaling axis has been implicated in the suppression of intestinal cancers, but the inhibitory mechanism and the extent to which this pathway can be targeted remains poorly understood. This study has tested the effect of cGMP-elevating agents on tumorigenesis in the ApcMin/+ mouse model of intestinal cancer. Treatment of ApcMin/+ mice with the receptor guanylyl-cyclase C (GCC) agonist linaclotide, or the phosphodiesterase-5 (PDE5) inhibitor sildenafil, significantly reduced the number of polyps per mouse (67% and 50%, respectively). Neither of the drugs affected mean polyp size, or the rates of apoptosis and proliferation. This was possibly due to increased PDE10 expression, as endogenous GCC ligands were not deficient in established polyps. These results indicated that the ability of these drugs to reduce polyp multiplicity was primarily due to an effect on nonneoplastic tissues. In support of this idea, ApcMin/+ mice exhibited reduced levels of endogenous GCC agonists in the nonneoplastic intestinal mucosa compared with wild-type animals, and this was associated with crypt hyperplasia and a loss of goblet cells. Administration of either sildenafil or linaclotide suppressed proliferation, and increased both goblet cell numbers and luminal apoptosis in the intestinal mucosa. Taken together, the results demonstrate that targeting cGMP with either PDE5 inhibitors or GCC agonists alters epithelial homeostasis in a manner that reduces neoplasia, and suggests that this could be a viable chemoprevention strategy for patients at high risk of developing colorectal cancer. Cancer Prev Res; 11(2); 81-92. ©2018 AACR.


Assuntos
Polipose Adenomatosa do Colo/prevenção & controle , Transformação Celular Neoplásica/efeitos dos fármacos , GMP Cíclico/metabolismo , Agonistas da Guanilil Ciclase C/farmacologia , Neoplasias Intestinais/prevenção & controle , Lesões Pré-Cancerosas/prevenção & controle , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Feminino , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/farmacologia , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Citrato de Sildenafila/farmacologia
17.
Am J Physiol Gastrointest Liver Physiol ; 314(1): G1-G13, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28935684

RESUMO

Previously, we showed that receptor for activated C kinase 1 (Rack1) regulates growth of colon cells in vitro, partly by suppressing Src kinase activity at key cell cycle checkpoints, in apoptotic and cell survival pathways and at cell-cell adhesions. Here, we generated mouse models of Rack1 deficiency to assess Rack1's function in intestinal epithelia in vivo. Intestinal Rack1 deficiency resulted in proliferation of crypt cells, diminished differentiation of crypt cells into enterocyte, goblet, and enteroendocrine cell lineages, and expansion of Paneth cell populations. Following radiation injury, the morphology of Rack1-deleted small bowel was strikingly abnormal with development of large polypoid structures that contained many partly formed villi, numerous back-to-back elongated and regenerating crypts, and high-grade dysplasia in surface epithelia. These abnormalities were not observed in Rack1-expressing areas of intestine or in control mice. Following irradiation, apoptosis of enterocytes was strikingly reduced in Rack1-deleted epithelia. These novel findings reveal key functions for Rack1 in regulating growth of intestinal epithelia: suppressing crypt cell proliferation and regeneration, promoting differentiation and apoptosis, and repressing development of neoplasia. NEW & NOTEWORTHY Our findings reveal novel functions for receptor for activated C kinase 1 (Rack1) in regulating growth of intestinal epithelia: suppressing crypt cell proliferation and regeneration, promoting differentiation and apoptosis, and repressing development of neoplasia.


Assuntos
Apoptose , Diferenciação Celular , Proliferação de Células , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Receptores de Quinase C Ativada/metabolismo , Regeneração , Animais , Apoptose/efeitos da radiação , Diferenciação Celular/efeitos da radiação , Linhagem da Célula , Proliferação de Células/efeitos da radiação , Células Epiteliais/patologia , Células Epiteliais/efeitos da radiação , Genótipo , Mucosa Intestinal/patologia , Mucosa Intestinal/efeitos da radiação , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Neoplasias Intestinais/prevenção & controle , Intestino Delgado/patologia , Intestino Delgado/efeitos da radiação , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Induzidas por Radiação/metabolismo , Neoplasias Induzidas por Radiação/patologia , Neoplasias Induzidas por Radiação/prevenção & controle , Fenótipo , Receptores de Quinase C Ativada/deficiência , Receptores de Quinase C Ativada/genética , Regeneração/efeitos da radiação , Transdução de Sinais
18.
Mol Nutr Food Res ; 62(2)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29125219

RESUMO

SCOPE: Evidence suggests that the dietary consumption of plant extracts containing polyphenols might help prevent the onset of cancers of the gastrointestinal tract. In the present study, the chemopreventive and antiproliferative efficacy of a grapevine shoot extract (Vineatrol®30) containing resveratrol and resveratrol oligomers is investigated in vivo and in vitro. METHODS AND RESULTS: The in vivo study is performed using ApcMin mice on a high-fat diet, which represents a model of human adenomatous polyposis, while the potential of the extract as well as some of its isolated constituents to inhibit intestinal adenoma cell proliferation in vitro is investigated using APC10.1 cells derived from an ApcMin mouse. Vineatrol®30 at a low (2.3 mg kg-1  diet) or high dose (476 mg kg-1  diet) reduces the adenoma number in male and adenoma volume in female animals. Furthermore, Vineatrol®30 as well as resveratrol and two resveratrol tetramers compromise the expansion of APC10.1 cells by reducing cell number, inducing cell cycle arrest, cellular senescence, and apoptosis. However, except for the extract, none of the isolated resveratrol oligomers is more efficacious than resveratrol in these cells. CONCLUSION: Vineatrol®30 may merit further investigation as a potential dietary gastrointestinal cancer chemopreventive agent in humans.


Assuntos
Adenoma/prevenção & controle , Anticarcinógenos/farmacologia , Neoplasias Intestinais/prevenção & controle , Fenóis/farmacologia , Resveratrol/farmacologia , Adenoma/metabolismo , Adenoma/patologia , Animais , Anticarcinógenos/química , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Feminino , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Masculino , Camundongos Mutantes , Fenóis/química , Resveratrol/química , Estilbenos/farmacologia
19.
Sci Rep ; 7: 43086, 2017 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-28225043

RESUMO

Among cancer diagnoses, colorectal cancer (CRC) is prevalent, with a lifetime risk of developing CRC being approximately 5%. Population variation surrounding the mean risk of developing CRCs has been associated with both inter-individual differences in genomic architecture and environmental exposures. Decreased risk of CRC has been associated with physical activity, but protective responses are variable. Here, we utilized a series of experiments to examine the effects of genetic background (strain), voluntary exercise (wheel running), and their interaction on azoxymethane (AOM)-induced intestinal tumor number and size in mice. Additionally, we investigated how the timing of exercise relative to AOM exposure, and amount of exercise, affected tumor number and size. Our results indicated that voluntary exercise significantly reduced tumor number in a strain dependent manner. Additionally, among strains where exercise reduced tumor number (A/J, CC0001/Unc) the timing of voluntary exercise relative to AOM exposure was crucial. Voluntary exercise prior to or during AOM treatment resulted in a significant reduction in tumor number, but exercise following AOM exposure had no effect. The results indicate that voluntary exercise should be used as a preventative measure to reduce risk for environmentally induced CRC with the realization that the extent of protection may depend on genetic background.


Assuntos
Azoximetano/metabolismo , Carcinogênese/induzido quimicamente , Carcinógenos/metabolismo , Neoplasias Intestinais/prevenção & controle , Condicionamento Físico Animal/métodos , Animais , Patrimônio Genético , Camundongos
20.
Methods Cell Biol ; 138: 241-270, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28129846

RESUMO

Although the zebrafish was initially developed as a model system to study embryonic development, it has gained increasing attention as an advantageous system to investigate human diseases, including intestinal disorders. Zebrafish embryos develop rapidly, and their digestive system is fully functional and visible by 5days post fertilization. There is a large degree of homology between the intestine of zebrafish and higher vertebrate organisms in terms of its cellular composition and function as both a digestive and immune organ. Furthermore, molecular pathways regulating injury and immune responses are highly conserved. In this chapter, we provide an overview of studies addressing developmental and physiological processes relevant to human intestinal disease. These studies include those related to congenital disorders, host-microbiota interactions, inflammatory diseases, motility disorders, and intestinal cancer. We also highlight the utility of zebrafish to functionally validate candidate genes identified through mutational analyses and genome-wide association studies, and discuss methodologies to investigate the intestinal biology that are unique to zebrafish.


Assuntos
Desenvolvimento Embrionário/genética , Imunidade Inata , Neoplasias Intestinais/genética , Intestinos/fisiopatologia , Peixe-Zebra/genética , Animais , Sistema Digestório/diagnóstico por imagem , Sistema Digestório/microbiologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/genética , Estudo de Associação Genômica Ampla , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiopatologia , Neoplasias Intestinais/patologia , Neoplasias Intestinais/prevenção & controle , Intestinos/microbiologia , Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...