Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Gastroenterology ; 160(7): 2395-2408, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33581124

RESUMO

BACKGROUND & AIMS: Gut microbial dysbiosis has pivotal involvement in colorectal cancer (CRC). However, the intratumoral microbiota and its association with CRC progression remain elusive. We aimed to determine the microbial community architecture within a neoplasia (CRC or adenoma) and its contribution to colorectal carcinogenesis. METHODS: We collected 436 tissue biopsies from patients with CRC (n = 36) or adenoma (n = 32) (2-6 biopsies from a neoplasia plus 2-5 biopsies from adjacent normal tissues per individual). Microbial profiling was performed using 16S ribosomal RNA gene sequencing with subsequent investigation of microbiota diversities and heterogeneity. The correlation between microbial dysbiosis and host genetic alterations (KRAS mutation and microsatellite instability) in all neoplasia biopsies was also analyzed. RESULTS: We discovered that intra-neoplasia microbial communities are heterogeneous. Abundances of some CRC-associated pathobionts (eg, Fusobacterium, Bacteroides, Parvimonas, and Prevotella) were found to be highly varied within a single neoplasia. Correlation of such heterogeneity with CRC development revealed alterations in microbial communities involving microbes with high intra-neoplasia variation in abundance. Moreover, we found that the intra-neoplasia variation in abundance of individual microbes changed along the adenoma-carcinoma sequence. We further determined that there was a significant difference in intra-neoplasia microbiota between biopsies with and without KRAS mutation (P < .001) or microsatellite instability (P < .001), and illustrated the association of intratumoral microbial heterogeneity with genetic alteration. CONCLUSIONS: We demonstrated that intra-neoplasia microbiota is heterogeneous and correlated with colorectal carcinogenesis. Our findings provide new insights on the contribution of gut microbiota heterogeneity to CRC progression.


Assuntos
Adenoma/microbiologia , Carcinogênese/genética , Neoplasias Colorretais/microbiologia , Microbioma Gastrointestinal/genética , Heterogeneidade Genética , Idoso , Biópsia , Colo/microbiologia , Colo/patologia , Feminino , Humanos , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas p21(ras)/genética , RNA Ribossômico 16S/análise
2.
Gastroenterology ; 161(5): 1423-1432.e4, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34273347

RESUMO

BACKGROUND & AIMS: Diet may contribute to the increasing incidence of colorectal cancer (CRC) before age 50 (early-onset CRC). Microbial metabolism of dietary sulfur produces hydrogen sulfide (H2S), a gastrointestinal carcinogen that cannot be easily measured at scale. As a result, evidence supporting its role in early neoplasia is lacking. METHODS: We evaluated long-term adherence to the sulfur microbial diet, a dietary index defined a priori based on increased abundance of 43 bacterial species involved with sulfur metabolism, with risk of CRC precursors among 59,013 individuals who underwent lower endoscopy in the Nurses' Health Study II (1991-2015), a prospective cohort study with dietary assessment every 4 years through validated food frequency questionnaires and an assessment of dietary intake during adolescence in 1998. The sulfur microbial diet was characterized by intake high in processed meats, foods previously linked to CRC development, and low in mixed vegetables and legumes. Multivariable logistic regression for clustered data was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS: We documented 2911 cases of early-onset adenoma. After adjusting for established risk factors, higher sulfur microbial diet scores were associated with increased risk for early-onset adenomas (ORquartile [Q]4 vs Q1, 1.31; 95% CI, 1.10-1.56, Ptrend = .02), but not serrated lesions. Compared with the lowest, women in the highest quartile of sulfur microbial diet scores had significantly increased risk of early-onset adenomas with greater malignant potential (ORQ4 vs Q1, 1.65 for villous/tubulovillous histology; 95% CI, 1.12-2.43; Ptrend = .04). Similar trends for early-onset adenoma were observed based on diet consumed during adolescence. In contrast, no clear association for adenomas was identified after age 50. CONCLUSIONS: Our findings in a cohort of young women support a role for dietary interactions with gut sulfur-metabolizing bacteria in early-onset colorectal carcinogenesis, possibly beginning in adolescence.


Assuntos
Pólipos Adenomatosos/epidemiologia , Bactérias/metabolismo , Pólipos do Colo/epidemiologia , Neoplasias Colorretais/epidemiologia , Dieta/efeitos adversos , Microbioma Gastrointestinal , Lesões Pré-Cancerosas/epidemiologia , Compostos de Enxofre/efeitos adversos , Pólipos Adenomatosos/diagnóstico , Adulto , Idade de Início , Pólipos do Colo/diagnóstico , Neoplasias Colorretais/diagnóstico , Feminino , Humanos , Sulfeto de Hidrogênio/efeitos adversos , Sulfeto de Hidrogênio/metabolismo , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/diagnóstico , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Compostos de Enxofre/administração & dosagem , Compostos de Enxofre/metabolismo , Fatores de Tempo , Estados Unidos/epidemiologia
3.
Am J Physiol Gastrointest Liver Physiol ; 320(2): G125-G135, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33084401

RESUMO

Among bacterial metabolites, hydrogen sulfide (H2S) has received increasing attention. The epithelial cells of the large intestine are exposed to two sources of H2S. The main one is the luminal source that results from specific bacteria metabolic activity toward sulfur-containing substrates. The other source in colonocytes is from the intracellular production mainly through cystathionine ß-synthase (CBS) activity. H2S is oxidized by the mitochondrial sulfide oxidation unit, resulting in ATP synthesis, and, thus, establishing this compound as the first mineral energy substrate in colonocytes. However, when the intracellular H2S concentration exceeds the colonocyte capacity for its oxidation, it inhibits the mitochondrial respiratory chain, thus affecting energy metabolism. Higher luminal H2S concentration affects the integrity of the mucus layer and displays proinflammatory effects. However, a low/minimal amount of endogenous H2S exerts an anti-inflammatory effect on the colon mucosa, pointing out the ambivalent effect of H2S depending on its intracellular concentration. Regarding colorectal carcinogenesis, forced CBS expression in late adenoma-like colonocytes increased their proliferative activity, bioenergetics capacity, and tumorigenicity; whereas, genetic ablation of CBS in mice resulted in a reduced number of mutagen-induced aberrant crypt foci. Activation of endogenous H2S production and low H2S extracellular concentration enhance cancerous colorectal cell proliferation. Higher exogenous H2S concentrations markedly reduce mitochondrial ATP synthesis and proliferative capacity in cancerous cells and enhance glycolysis but do not affect their ATP cell content or viability. Thus, it appears that, notably through an effect on colonocyte energy metabolism, endogenous and microbiota-derived H2S are involved in the host intestinal physiology and physiopathology.


Assuntos
Colo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Sulfeto de Hidrogênio/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Reto/efeitos dos fármacos , Animais , Humanos , Sulfeto de Hidrogênio/toxicidade , Mucosa Intestinal/citologia
4.
Gastroenterology ; 158(5): 1313-1325, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31972239

RESUMO

BACKGROUND & AIMS: Sulfur-metabolizing microbes, which convert dietary sources of sulfur into genotoxic hydrogen sulfide (H2S), have been associated with development of colorectal cancer (CRC). We identified a dietary pattern associated with sulfur-metabolizing bacteria in stool and then investigated its association with risk of incident CRC using data from a large prospective study of men. METHODS: We collected data from 51,529 men enrolled in the Health Professionals Follow-up Study since 1986 to determine the association between sulfur-metabolizing bacteria in stool and risk of CRC over 26 years of follow-up. First, in a subcohort of 307 healthy men, we profiled serial stool metagenomes and metatranscriptomes and assessed diet using semiquantitative food frequency questionnaires to identify food groups associated with 43 bacterial species involved in sulfur metabolism. We used these data to develop a sulfur microbial dietary score. We then used Cox proportional hazards modeling to evaluate adherence to this pattern among eligible individuals (n = 48,246) from 1986 through 2012 with risk for incident CRC. RESULTS: Foods associated with higher sulfur microbial diet scores included increased consumption of processed meats and low-calorie drinks and lower consumption of vegetables and legumes. Increased sulfur microbial diet scores were associated with risk of distal colon and rectal cancers, after adjusting for other risk factors (multivariable relative risk, highest vs lowest quartile, 1.43; 95% confidence interval 1.14-1.81; P-trend = .002). In contrast, sulfur microbial diet scores were not associated with risk of proximal colon cancer (multivariable relative risk 0.86; 95% CI 0.65-1.14; P-trend = .31). CONCLUSIONS: In an analysis of participants in the Health Professionals Follow-up Study, we found that long-term adherence to a dietary pattern associated with sulfur-metabolizing bacteria in stool was associated with an increased risk of distal CRC. Further studies are needed to determine how sulfur-metabolizing bacteria might contribute to CRC pathogenesis.


Assuntos
Bactérias/metabolismo , Neoplasias Colorretais/epidemiologia , Fezes/microbiologia , Comportamento Alimentar/fisiologia , Microbioma Gastrointestinal/fisiologia , Idoso , Bactérias/isolamento & purificação , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/prevenção & controle , Inquéritos sobre Dietas/estatística & dados numéricos , Seguimentos , Pessoal de Saúde/estatística & dados numéricos , Humanos , Incidência , Masculino , Massachusetts/epidemiologia , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco , Enxofre/metabolismo
5.
Proc Natl Acad Sci U S A ; 115(17): E4061-E4070, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29632181

RESUMO

Colorectal cancer (CRC) is one of the most common tumor entities, which is causally linked to DNA repair defects and inflammatory bowel disease (IBD). Here, we studied the role of the DNA repair protein poly(ADP-ribose) polymerase-1 (PARP-1) in CRC. Tissue microarray analysis revealed PARP-1 overexpression in human CRC, correlating with disease progression. To elucidate its function in CRC, PARP-1 deficient (PARP-1-/-) and wild-type animals (WT) were subjected to azoxymethane (AOM)/ dextran sodium sulfate (DSS)-induced colorectal carcinogenesis. Miniendoscopy showed significantly more tumors in WT than in PARP-1-/- mice. Although the lack of PARP-1 moderately increased DNA damage, both genotypes exhibited comparable levels of AOM-induced autophagy and cell death. Interestingly, miniendoscopy revealed a higher AOM/DSS-triggered intestinal inflammation in WT animals, which was associated with increased levels of innate immune cells and proinflammatory cytokines. Tumors in WT animals were more aggressive, showing higher levels of STAT3 activation and cyclin D1 up-regulation. PARP-1-/- animals were then crossed with O6-methylguanine-DNA methyltransferase (MGMT)-deficient animals hypersensitive to AOM. Intriguingly, PARP-1-/-/MGMT-/- double knockout (DKO) mice developed more, but much smaller tumors than MGMT-/- animals. In contrast to MGMT-deficient mice, DKO animals showed strongly reduced AOM-dependent colonic cell death despite similar O6-methylguanine levels. Studies with PARP-1-/- cells provided evidence for increased alkylation-induced DNA strand break formation when MGMT was inhibited, suggesting a role of PARP-1 in the response to O6-methylguanine adducts. Our findings reveal PARP-1 as a double-edged sword in colorectal carcinogenesis, which suppresses tumor initiation following DNA alkylation in a MGMT-dependent manner, but promotes inflammation-driven tumor progression.


Assuntos
Neoplasias Colorretais/enzimologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Camundongos , Camundongos Knockout , Poli(ADP-Ribose) Polimerase-1/genética , Proteínas Supressoras de Tumor/genética
6.
Int J Med Sci ; 17(16): 2505-2510, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33029093

RESUMO

Background and purpose: The discovery of chemical substances with carcinogenic properties has allowed the development of several experimental models of colorectal cancer (CRC). Classically, experimental models of CRC in mice have been evaluated through clinical or serial euthanasia. The present study aims to investigate the role of low endoscopy in the analysis of carcinogenesis induced by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Methods: Thirty C57BL6 mice were divided into two groups: a control group with fifteen animals that underwent rectal instillation of saline solution on day 0 and a carcinogen group with fifteen animals that underwent a 100 mg/kg MNNG rectal instillation on day 0. In both groups, low endoscopies were performed on weeks 4 and 8. We used a validated endoscopic scoring system to evaluate the severity of colitis and colorectal tumor. Euthanasia was carried out at week 12. Results: We observed higher inflammation scores (p <0.001) and a higher number of tumors (p <0.05) in the MNNG group than the control group, both at weeks 4 and 8. A worsening of inflammation scores from the first to the second endoscopy was also noticeable in the MNNG group. There were no bowel perforations related to the procedure, and there was one death in the control group. Conclusion: Low endoscopy in experimental animals allows safe macroscopic evaluation of colorectal carcinogenesis without the need for euthanasia.


Assuntos
Metilnitronitrosoguanidina/toxicidade , Neoplasias Experimentais/induzido quimicamente , Neoplasias Retais/induzido quimicamente , Administração Retal , Animais , Carcinogênese/induzido quimicamente , Carcinogênese/patologia , Colonoscopia/métodos , Feminino , Humanos , Metilnitronitrosoguanidina/administração & dosagem , Camundongos , Neoplasias Experimentais/diagnóstico , Neoplasias Experimentais/patologia , Neoplasias Retais/diagnóstico , Neoplasias Retais/patologia , Reto/diagnóstico por imagem , Reto/efeitos dos fármacos , Reto/patologia
7.
Crit Rev Food Sci Nutr ; 59(1): 59-71, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-28799785

RESUMO

Colorectal cancer (CRC) is one of the most common cause of cancer death. Phytochemicals, especially anthocyanins/anthocyanidins (A/A), have gathered attention of the scientific community owing to their anti-inflammatory, antioxidant, and cancer-inhibitory properties. In this review, we discussed the possible mechanisms whereby A/A exhibit intestinal anticarcinogenic characteristics. Anthocyanins/anthocyanidins inhibit the pro-inflammatory NF-κB pathway, attenuate Wnt signaling and suppress abnormal epithelial cell proliferation. In addition, A/A induce mitochondrial-mediated apoptosis and downregulate Akt/mTOR (mammalian target of rapamycin) pathway. Furthermore, activation of AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1) also contributes to the anti-carcinogenic effects of A/A. Finally, downregulation of metalloproteinases (MMPs) by A/A inhibits tumor invasion and metastasis. In conclusion, A/A exert their anti-tumor effects against colorectal carcinogenesis via multiple mechanisms, providing insights into the use of A/A as a natural chemopreventive intervention on major colorectal carcinogenesis.


Assuntos
Antocianinas/farmacologia , Neoplasias Colorretais/prevenção & controle , Compostos Fitoquímicos/farmacologia , Antocianinas/química , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Compostos Fitoquímicos/química , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
8.
Int J Cancer ; 142(1): 121-132, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28875496

RESUMO

The putative cannabinoid receptor GPR55 has been shown to play a tumor-promoting role in various cancers, and is involved in many physiological and pathological processes of the gastrointestinal (GI) tract. While the cannabinoid receptor 1 (CB1 ) has been reported to suppress intestinal tumor growth, the role of GPR55 in the development of GI cancers is unclear. We, therefore, aimed at elucidating the role of GPR55 in colorectal cancer (CRC), the third most common cancer worldwide. Using azoxymethane (AOM)- and dextran sulfate sodium (DSS)-driven CRC mouse models, we found that GPR55 plays a tumor-promoting role that involves alterations of leukocyte populations, i.e. myeloid-derived suppressor cells and T lymphocytes, within the tumor tissues. Concomitantly, expression levels of COX-2 and STAT3 were reduced in tumor tissue of GPR55 knockout mice, indicating reduced presence of tumor-promoting factors. By employing the experimental CRC models to CB1 knockout and CB1 /GPR55 double knockout mice, we can further show that GPR55 plays an opposing role to CB1 . We report that GPR55 and CB1 mRNA expression are differentially regulated in the experimental models and in a cohort of 86 CRC patients. Epigenetic methylation of CNR1 and GPR55 was also differentially regulated in human CRC tissue compared to control samples. Collectively, our data suggest that GPR55 and CB1 play differential roles in colon carcinogenesis where the former seems to act as oncogene and the latter as tumor suppressor.


Assuntos
Carcinogênese/metabolismo , Neoplasias Colorretais/patologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Neoplasias Colorretais/metabolismo , Humanos , Camundongos , Camundongos Knockout , Receptor CB1 de Canabinoide/metabolismo , Receptores de Canabinoides/metabolismo
9.
Gastroenterology ; 152(3): 546-549.e3, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27816598

RESUMO

We investigated the presence and patterns of mosaicism in the APC gene in patients with colon neoplasms not associated with any other genetic variants; we performed deep sequence analysis of APC in at least 2 adenomas or carcinomas per patient. We identified mosaic variants in APC in adenomas from 9 of the 18 patients with 21 to approximately 100 adenomas. Mosaic variants of APC were variably detected in leukocyte DNA and/or non-neoplastic intestinal mucosa of these patients. In a comprehensive sequence analysis of 1 patient, we found no evidence for mosaicism in APC in non-neoplastic intestinal mucosa. One patient was found to carry a mosaic c.4666dupA APC variant in only 10 of 16 adenomas, indicating the importance of screening 2 or more adenomas for genetic variants.


Assuntos
Adenoma/genética , Polipose Adenomatosa do Colo/genética , Carcinoma/genética , Neoplasias Colorretais/genética , Genes APC , Mosaicismo , Neoplasias Primárias Múltiplas/genética , Adulto , Idoso , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência de DNA , Via de Sinalização Wnt
10.
Gastroenterology ; 153(4): 1040-1053.e4, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28716720

RESUMO

BACKGROUND & AIMS: Resistance of metastatic human colorectal cancer cells to drugs that block epidermal growth factor (EGF) receptor signaling could be caused by aberrant activity of other receptor tyrosine kinases, activating overlapping signaling pathways. One of these receptor tyrosine kinases could be MET, the receptor for hepatocyte growth factor (HGF). We investigated how MET signaling, and its interaction with CD44 (a putative MET coreceptor regulated by Wnt signaling and highly expressed by intestinal stem cells [ISCs] and adenomas) affects intestinal homeostasis, regeneration, and adenoma formation in mini-gut organoids and mice. METHODS: We established organoid cultures from ISCs stimulated with HGF or EGF and assessed intestinal differentiation by immunohistochemistry. Mice with total epithelial disruption of MET (AhCre/Metfl/fl/LacZ) or ISC-specific disruption of MET (Lgr5Creert2/Metfl/fl/LacZ) and control mice (AhCre/Met+/+/LacZ, Lgr5Creert2/Met+/+/LacZ) were exposed to 10 Gy total body irradiation; intestinal tissues were collected, and homeostasis and regeneration were assessed by immunohistochemistry. We investigated adenoma organoid expansion stimulated by HGF or EGF using adenomas derived from Lgr5Creert2/Metfl/fl/Apcfl/fl and Lgr5Creert2/Met+/+/Apcfl/fl mice. The same mice were evaluated for adenoma prevalence and size. We also quantified adenomas in AhCre/Metfl/fl/Apcfl/+ mice compared with AhCre/Met+/+/Apcfl/+ control mice. We studied expansion of organoids generated from crypts and adenomas, stimulated by HGF or EGF, that were derived from mice expressing different CD44 splice variants (Cd44+/+, Cd44-/-, Cd44s/s, or Cd44v4-10/v4-10 mice). RESULTS: Crypts incubated with EGF or HGF expanded into self-organizing mini-guts with similar levels of efficacy and contained all differentiated cell lineages. MET-deficient mice did not have defects in intestinal homeostasis. Total body irradiation reduced numbers of proliferating crypts in AhCre/Metfl/fl/LacZ mice. Lgr5Creert2/Metfl/fl/LacZ mice had impaired regeneration of MET-deficient ISCs. Adenoma organoids stimulated with EGF or HGF expanded to almost twice the size of nonstimulated organoids. MET-deficient adenoma organoids did not respond to HGF stimulation, but did respond to EGF. ISC-specific disruption of Met (Lgr5Creert2/Metfl/fl/Apcfl/fl mice) caused a twofold increase in apoptosis in microadenomas, resulting in an approximately 50% reduction of microadenoma numbers and significantly reduced average adenoma size. Total epithelial disruption of Met (AhCre/Metfl/fl/Apcfl/+ mice) resulted in an approximate 50% reduction in (micro)adenoma numbers. Intestinal crypts from Cd44-/- mice did not expand to the same extent as crypts from Cd44+/+ mice on stimulation with HGF, but had the same response to EGF. The negative effect on HGF-mediated growth was overcome by expression of CD44v4-10, but not by CD44s. Similarly, HGF-mediated expansion of adenoma organoids required CD44v4-10. CONCLUSIONS: In studies of intestinal organoid cultures and mice with inducible deletion of MET, we found HGF receptor signaling to regulate intestinal homeostasis and regeneration, as well as adenoma formation. These activities of MET are promoted by the stem cell CD44 isoform CD44v4-10. Our findings provide rationale for targeting signaling via MET and CD44 during anti-EGF receptor therapy of patients with colorectal cancer or in patients resistant to EGF receptor inhibitors.


Assuntos
Adenoma/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Receptores de Hialuronatos/metabolismo , Neoplasias Intestinais/metabolismo , Intestinos/enzimologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Regeneração , Células-Tronco/enzimologia , Adenoma/genética , Adenoma/patologia , Animais , Diferenciação Celular , Linhagem da Célula , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Células Cultivadas , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Genótipo , Fator de Crescimento de Hepatócito/farmacologia , Homeostase , Receptores de Hialuronatos/genética , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Intestinos/efeitos dos fármacos , Intestinos/patologia , Intestinos/efeitos da radiação , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Proteínas Proto-Oncogênicas c-met/genética , Regeneração/efeitos dos fármacos , Regeneração/efeitos da radiação , Transdução de Sinais , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Células-Tronco/efeitos da radiação , Fatores de Tempo , Técnicas de Cultura de Tecidos , Carga Tumoral
11.
Anaerobe ; 49: 63-70, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29277623

RESUMO

The colon rectal portion of gastrointestinal tract (GI) is full of microorganisms with different complex community that plays important role in maintaining homeostasis. But now-a-days different literature indicated that microbiota cause development of colorectal cancer (CRC) with a disease and ultimately aggravates to death. The mechanism inside the colo-rectal portion of GI tract is not fully well-known and bacterial contribution inside it is also fully unclear. Therefore, there is certain evidence trying a target about the unclear mechanism between intestinal microbiota and CRC. Different reports revealed that colo-rectal microorganisms is playing a great role in inducing the onset and progression of CRC with different dynamic mechanisms viz. acceleration of chronic inflammatory state, the biosynthesis of genotoxins that interfere with cell cycle regulation, the production of toxic metabolites, or heterocyclic amine activation of pro-diet carcinogenic compounds. There is growing evidence that individuals with colonic adenomas and carcinomas harbor a distinct microbiota. Alterations to the gut microbiota may allow the outgrowth of bacterial populations that induce genomic mutations or exacerbate tumor-promoting inflammation. While cancer is largely considered to be a disease of genetic and environmental factors, increasing evidence has demonstrated a role for the microbiota in shaping inflammatory environments and promoting tumor growth and spread. Despite all these advances, different studies depicted the relationship between microbiota and CRC in humans and animal models and aid in developing alternate therapeutic approach based on gut microbiota manipulations. Alteration of the microbiota may be a useful to preventing and altering the trajectory of colorectal cancer. Therefore, the aim of the study is to identify the possible mechanistic mechanism regarding host-microbiota interaction in colorectal carcinogenesis.


Assuntos
Bactérias/isolamento & purificação , Neoplasias Colorretais/microbiologia , Microbioma Gastrointestinal , Animais , Bactérias/classificação , Bactérias/genética , Carcinogênese , Neoplasias Colorretais/patologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Humanos
12.
Int J Mol Sci ; 18(5)2017 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-28448444

RESUMO

The presence of chronic inflammation in the colonic mucosa leads to an increased risk of cancer. Among proteins involved in the regulation of mucosal inflammation and that may contribute both to structural damage of the intestinal mucosa and to intestinal carcinogenesis, there are myeloperoxidase (MPO) and vanins. The infiltration of colonic mucosa by neutrophils may promote carcinogenesis through MPO, a key enzyme contained in the lysosomes of neutrophils that regulates local inflammation and the generation of reactive oxygen species (ROS) and mutagenic species. The human vanin gene family consists of three genes: vanin-1, vanin-2 and vanin-3. All vanin molecules are pantetheinases, that hydrolyze pantetheine into pantothenic acid (vitamin B5), and cysteamine, a sulfhydryl compound. Vanin-1 loss confers an increased resistance to stress and acute intestinal inflammation, while vanin-2 regulates adhesion and transmigration of activated neutrophils. The metabolic product of these enzymes has a prominent role in the inflammation processes by affecting glutathione levels, inducing ulcers through a reduction in mucosal blood flow and oxygenation, decreasing local defense mechanisms, and in carcinogenesis by damaging DNA and regulating pathways involved in cell apoptosis, metabolism and growth, as Nrf2 and HIF-1α.


Assuntos
Amidoidrolases/metabolismo , Neoplasias Colorretais/patologia , Peroxidase/metabolismo , Amidoidrolases/antagonistas & inibidores , Amidoidrolases/genética , Carcinogênese , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Cisteamina/metabolismo , Inibidores Enzimáticos/uso terapêutico , Humanos , Inflamação , Peroxidase/antagonistas & inibidores , Peroxidase/genética , Espécies Reativas de Oxigênio/metabolismo
13.
Biochim Biophys Acta ; 1851(1): 30-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25139561

RESUMO

The nuclear Farnesoid X Receptor (FXR) is a transcription factor critically involved in metabolic homeostasis in the gut-liver axis. FXR activity is mediated by hormonal and dietary signals and driven by bile acids (BAs), which are the natural FXR ligands. Given the great physiological importance in BA homeostasis, as well as in the regulation of glucose and lipid metabolism, FXR plays a pivotal role in the pathogenesis of a wide range of disease of the liver, biliary tract and intestine, including hepatic and colorectal cancer. In the last years several studies have shown the relative FXR tissue-specific importance, highlighting synergism and additive effects in the liver and intestine. Gain- and loss-of-FXR-function mouse models have been generated in order to identify the biological processes and the molecular FXR targets. Taking advantage of the knowledge on the structure-activity relationship of BAs for FXR, semi-synthetic and synthetic molecules have been generated to obtain more selective and powerful FXR activators than BAs. This article is part of a Special Issue entitled: Linking transcription to physiology in lipodomics.


Assuntos
Neoplasias/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Glucose/metabolismo , Homeostase , Humanos , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Relação Estrutura-Atividade
14.
Int J Mol Sci ; 17(11)2016 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-27801847

RESUMO

Hypercholesterolemia resulting in atherosclerosis is associated with an increased risk of ischemic heart disease and colorectal cancer (CRC). However, the roles of apoliprotein (Apo) E (Apoe) and low-density lipoprotein (Ldl) receptor (Ldlr) in colorectal carcinogenesis have not yet been investigated. In this study, we examined the susceptibility of Apoe-deficient and Ldlr-deficient mice, which are genetic animal models of atherosclerosis to azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colorectal carcinogenesis. In Experiment 1, male Apoe-deficient (n = 20) and wild type (WT) mice (C57BL/6J, n = 21) were treated with a single intraperitoneal (i.p.) injection of AOM (10 mg/kg body weight) and then given 1.5% DSS in drinking water for seven days. They were maintained up to week 20 and sacrificed for the histopathological examination of colorectal tumors. The mRNA expression of cyclooxygenase (Cox)-2, inducible nitric oxide synthase (Nos2), tumor necrosis factor (Tnf)-α interleukin (Il)-1ß, and Il-6 was assayed in the colorectal mucosa. In Experiment 2, male Ldlr-deficient (n = 14) and WT mice (C57BL/6J, n = 10) were given a single i.p. injection of AOM (10 mg/kg body weight) and then given 2% DSS in drinking water for seven days. They were sacrificed at week 20 to evaluate their colorectum histopathologically. In Experiment 1, the multiplicity of CRCs was significantly higher in the Apoe-deficient mice (2.75 ± 1.48) than in the WT mice (0.62 ± 0.67). The serum lipoprotein levels in the Apoe-deficient mice were also significantly higher than in the WT mice. In Experiment 2, the incidence (29%) and multiplicity (0.50 ± 0.94) of CRCs in the Ldlr mice were significantly lower than in the WT mice (80% incidence and 3.10 ± 2.38 multiplicity). The mRNA expression of two inducible enzymes and certain pro-inflammatory cytokines in the colorectum of each genotype was greater than in the respective WT mice. The values in the Apoe-deficient mice were much greater than in the Ldlr mice. These findings suggest that Apoe-deficient mice showed increased susceptibility to inflammation-associated colorectal carcinogenesis due to their high reactivity to inflammatory stimuli.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Neoplasias Colorretais/genética , Inflamação/genética , Receptores de LDL/genética , Animais , Apolipoproteínas E/deficiência , Aterosclerose/patologia , Azoximetano/toxicidade , Carcinogênese/genética , Neoplasias Colorretais/sangue , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/patologia , Ciclo-Oxigenase 2/biossíntese , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Inflamação/sangue , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Lipoproteínas/sangue , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/biossíntese , Receptores de LDL/deficiência , Fator de Necrose Tumoral alfa/biossíntese
15.
Phytother Res ; 28(6): 892-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24142591

RESUMO

Patients suffering from inflammatory bowel disease are at a high risk of developing colorectal cancer. To assess the anticancer potential of botanicals, in this study, we evaluated the effects of Panax notoginseng on azoxymethane/dextran sulfate sodium (DSS)-induced colitis. One week after A/J mice received azoxymethane, the animals received DSS for 8 days or were supplemented with P. notoginseng extract, at 30 or 90 mg/kg. DSS-induced colitis was scored with the disease activity index. The severity of the inflammatory lesions was evaluated by a colon tissue histological assessment. The expression of inducible nitric oxide synthase and cyclooxygenase-2 (COX-2) were also explored. We observed that the effects of P. notoginseng on the reduction of colon inflammation, expressed in disease activity index score, were in a dose-related manner (p < 0.01). P. notoginseng inhibited the reduction of the colon length and the loss of bodyweight in dose-related manner (all p < 0.05). The histological assessment of the colitis and inflammatory-related immunohistochemical data also supported the pharmacological observations. Our data suggest that P. notoginseng is a promising candidate in preventing and treating colitis and inflammation-associated colon carcinogenesis.


Assuntos
Colite/tratamento farmacológico , Panax notoginseng/química , Extratos Vegetais/farmacologia , Animais , Azoximetano , Colite/induzido quimicamente , Colite/patologia , Colo/efeitos dos fármacos , Colo/patologia , Ciclo-Oxigenase 2/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Masculino , Camundongos , Óxido Nítrico Sintase Tipo II/metabolismo , Extratos Vegetais/química , Saponinas/química
16.
Tissue Cell ; 87: 102293, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38244400

RESUMO

The current study investigated the potential effects of probiotic supplementation on colorectal carcinogenesis chemically induced with 1,2-dimethylhydrazine (DMH) and treated with 5-fluorouracil (5FU)-based chemotherapy in mice. Animals were randomly allocated in five different groups: Control: which not receive any treatment throughout the experimental course; Colitis model group (DMH): treated with DMH; DMH+ 5FU: animals received I.P. (intraperitoneal) dose of chemotherapy on a weekly basis; DMH+PROB: animals received daily administrations (via gavage) of probiotics (Lactobacillus: acidophilus and paracasei, Bifidobacterium lactis and bifidum); and DMH+ PROB+ 5FU: animals received the same treatment as the previous groups. After ten-week treatment, mice's large intestine was collected and subjected to colon length, histopathological, periodic acid-schiff (PAS) staining and immunohistochemistry (TLR2, MyD88, NF-κB, IL-6, TLR4, TRIF, IRF-3, IFN-γ, Ki-67, KRAS, p53, IL-10, and TGF-ß) analyzes. Variance (ANOVA) and Kruskal-Wallis tests were used for statistical analysis, at significance level p 0.05. Probiotics' supplementation has increased the production of Ki-67 cell-proliferation marker, reduced body weight, and colon shortening, as well as modulated the chronic inflammatory process in colorectal carcinogenesis by inhibiting NF-κB expression and mitigating mucin depletion. Thus, these findings lay a basis for guide future studies focused on probiotics' action mechanisms in tumor microenvironment which might have implications in clinical practice.


Assuntos
Neoplasias Colorretais , Probióticos , Camundongos , Animais , 1,2-Dimetilidrazina/toxicidade , NF-kappa B , Antígeno Ki-67 , Carcinogênese/patologia , Probióticos/farmacologia , Probióticos/uso terapêutico , Inflamação/tratamento farmacológico , Inflamação/patologia , Neoplasias Colorretais/patologia , Fluoruracila/farmacologia , Colo/microbiologia , Colo/patologia , Microambiente Tumoral
17.
Chemosphere ; 359: 142332, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38754493

RESUMO

Perfluorooctanesulfonic acid (PFOS) is a widely recognized environment pollutant known for its high bioaccumulation potential and a long elimination half-life. Several studies have shown that PFOS can alter multiple biological pathways and negatively affect human health. Considering the direct exposure to the gastrointestinal (GI) tract to environmental pollutants, PFOS can potentially disrupt intestinal homeostasis. However, there is limited knowledge about the effect of PFOS exposure on normal intestinal tissues, and its contribution to GI-associated diseases remains to be determined. In this study, we examined the effect of PFOS exposure on the gene expression profile of intestinal tissues of C57BL/6 mice using RNAseq analysis. We found that PFOS exposure in drinking water significantly downregulates mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), a rate-limiting ketogenic enzyme, in intestinal tissues of mice. We found that diets containing the soluble fibers inulin and pectin, which are known to be protective against PFOS exposure, were ineffective in reversing the downregulation of HMGCS2 expression in vivo. Analysis of intestinal tissues also demonstrated that PFOS exposure leads to upregulation of proteins implicated in colorectal carcinogenesis, including ß-catenin, c-MYC, mTOR and FASN. Consistent with the in vivo results, PFOS exposure leads to downregulation of HMGCS2 in mouse and human normal intestinal organoids in vitro. Furthermore, we show that shRNA-mediated knockdown of HMGCS2 in a human normal intestinal cell line resulted in increased cell proliferation and upregulation of key proliferation-associated proteins such as cyclin D, survivin, ERK1/2 and AKT, along with an increase in lipid accumulation. In summary, our results suggest that PFOS exposure may contribute to pathological changes in normal intestinal cells via downregulation of HMGCS2 expression and upregulation of pro-carcinogenic signaling pathways that may increase the risk of colorectal cancer development.


Assuntos
Ácidos Alcanossulfônicos , Carcinogênese , Regulação para Baixo , Fluorocarbonos , Hidroximetilglutaril-CoA Sintase , Camundongos Endogâmicos C57BL , Animais , Ácidos Alcanossulfônicos/toxicidade , Fluorocarbonos/toxicidade , Hidroximetilglutaril-CoA Sintase/metabolismo , Hidroximetilglutaril-CoA Sintase/genética , Camundongos , Regulação para Baixo/efeitos dos fármacos , Neoplasias Intestinais/induzido quimicamente , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Regulação para Cima/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Intestinos/efeitos dos fármacos , Humanos , Mucosa Intestinal/metabolismo
18.
Asian Pac J Cancer Prev ; 25(2): 617-626, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38415549

RESUMO

OBJECTIVE: This study aimed to evaluate the expression of Beclin 1 and HER2 proteins using immunohistochemistry in CRC tissues compared to colonic adenoma, and to investigate the correlation of their expression with clinicopathological parameters and survival outcomes in CRC patients. METHODS: The study utilized paraffin-embedded blocks from 17 colonic adenoma and 81 CRC cases. Immunohistochemical analysis was performed to assess the expression of Beclin 1 and HER2 proteins. RESULTS: The cytoplasmic expression of Beclin 1 was significantly higher in CRC tissues compared to adenoma specimens (P=0.051). High Beclin 1 expression was significantly associated with distal colon location (P=0.028). High HER2 cytoplasmic expression was significantly associated with vascular invasion (P=0.05), perineural invasion (P=0.03), and shorter overall survival (P=0.035). CONCLUSIONS: The findings suggest that Beclin 1 plays a role in colorectal carcinogenesis, with higher expression observed in CRC cases compared to adenoma cases.  Furthermore, HER2 carries poor prognostic impact in CRC cases.


Assuntos
Adenoma , Neoplasias do Colo , Neoplasias Colorretais , Humanos , Proteína Beclina-1 , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/patologia , Prognóstico , Adenoma/patologia
19.
J Gastroenterol Hepatol ; 28 Suppl 4: 41-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24251703

RESUMO

Colorectal cancer is the third leading cause of cancer death in Japan and the United States and is strongly associated with obesity, especially visceral obesity. Several metabolic mediators, such as adiponectin, have been suspected to play a role in obesity-related carcinogenesis. In a previous human study, the existence of a significant correlation between the number of human dysplastic aberrant crypt foci (ACF) and the visceral fat area was demonstrated, and also that of a significant inverse correlation between the number of dysplastic ACF and the plasma adiponectin level. Other studies have investigated the effect of adiponectin under the normal and high-fat diet conditions in a mouse model of azoxymethane-induced colon cancer. Enhanced formation of both ACF and tumors was observed in the adiponectin-deficient mice, as compared with that in the wild-type, under the high-fat diet condition but not under the normal diet condition. Furthermore, that the 5'-AMP-activated kinase/mammalian target of rapamycin pathway is involved in the promotion of colorectal carcinogenesis in adiponectin-deficient mice under the high-fat diet condition was shown. Therefore, that the 5'-AMP-activated kinase/mammalian target of rapamycin signaling pathway may play an important role in colorectal carcinogenesis was speculated. Metformin, a biguanide derivative widely used in the treatment of diabetes mellitus, has been shown to exert a suppressive effect on ACF formation in both mouse models and humans. Therefore, metformin might be a promising candidate as a safe drug for chemoprevention of colorectal carcinogenesis. Further studies with high evidence levels, such as randomized, controlled studies, are needed to clarify these relationships.


Assuntos
Focos de Criptas Aberrantes/etiologia , Colo/patologia , Neoplasias Colorretais/etiologia , Dieta Hiperlipídica/efeitos adversos , Obesidade Abdominal/etiologia , Obesidade Abdominal/patologia , Proteínas Quinases Ativadas por AMP/fisiologia , Focos de Criptas Aberrantes/sangue , Focos de Criptas Aberrantes/patologia , Focos de Criptas Aberrantes/prevenção & controle , Adiponectina/sangue , Adiponectina/fisiologia , Animais , Proliferação de Células , Transformação Celular Neoplásica , Neoplasias Colorretais/sangue , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Modelos Animais de Doenças , Células Epiteliais/patologia , Humanos , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Camundongos , Obesidade Abdominal/sangue , Fatores de Risco , Transdução de Sinais/fisiologia , Sirolimo
20.
Cancer Manag Res ; 15: 435-448, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37250384

RESUMO

The Wnt/ß-catenin signaling pathway is a growth control pathway involved in various biological processes as well as the development and progression of cancer. Colorectal cancer (CRC) is one of the most common malignancies in the world. The hyperactivation of Wnt signaling is observed in almost all CRC and plays a crucial role in cancer-related processes such as cancer stem cell (CSC) propagation, angiogenesis, epithelial-mesenchymal transition (EMT), chemoresistance, and metastasis. This review will discuss how the Wnt/ß-catenin signaling pathway is involved in the carcinogenesis and progression of CRC and related therapeutic approaches.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa