Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 733
Filtrar
1.
Int J Mol Sci ; 25(20)2024 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-39456736

RESUMO

Colorectal cancer CRC remains one of the leading causes of cancer-related deaths worldwide, with chronic intestinal inflammation identified as a major risk factor. Notably, the tumor suppressor TP53 undergoes mutation at higher rates and earlier stages during human inflammation-driven colon tumorigenesis than in sporadic cases. We investigated whether deleting Trp53 affects inflammation-induced tumor growth and the expression of Lgr5+ cancer stem cells in mice. We examined azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colon tumorigenesis in wild-type Trp53 (+/+), heterozygous (+/-), and knockout (-/-) mice. Trp53-/- mice showed increased sensitivity to DSS colitis and earlier accelerated tumorigenesis with 100% incidence. All groups could develop invasive tumors, but knockouts displayed the most aggressive features. Unlike wild-type CRC, knockouts selectively showed increased populations of Lgr5+ colon cancer stem-like cells. Trp53 loss also boosted laminin, possibly facilitating the disruption of the tumor border. This study highlights how Trp53 deletion promotes the perfect storm of inflammation and stemness, driving colon cancer progression. Trp53 deletion dramatically shortened AOM/DSS latency and improved tumor induction efficiency, offering an excellent inflammation-driven CRC model.


Assuntos
Azoximetano , Carcinogênese , Colite , Neoplasias Colorretais , Sulfato de Dextrana , Camundongos Knockout , Células-Tronco Neoplásicas , Receptores Acoplados a Proteínas G , Proteína Supressora de Tumor p53 , Animais , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Camundongos , Azoximetano/toxicidade , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Colite/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/etiologia , Carcinogênese/genética , Carcinogênese/patologia , Carcinogênese/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Deleção de Genes
2.
Mar Drugs ; 22(10)2024 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-39452878

RESUMO

In this study, we investigated the protective effects of astaxanthin (AST) against oxidative stress induced by the combination of azoxymethane (AOM) and dextran sulfate sodium (DSS) in colitis-associated cancer (CAC) and TNF-α-induced human colorectal cancer cells (SW480), as well as the underlying mechanism. In vitro experiments revealed that astaxanthin reduced reactive oxygen species (ROS) generation and inhibited the expression of Phosphorylated JNK (P-JNK), Phosphorylated ERK (P-ERK), Phosphorylated p65 (P-p65), and the NF-κB downstream protein cyclooxygenase-2 (COX-2). In vivo experiments showed that astaxanthin ameliorated AOM/DSS-induced weight loss, shortened the colon length, and caused histomorphological changes. In addition, astaxanthin suppressed cellular inflammation by modulating the MAPK and NF-κB pathways and inhibiting the expression of the proinflammatory cytokines IL-6, IL-1ß, and TNF-α. In conclusion, astaxanthin attenuates cellular inflammation and CAC through its antioxidant effects.


Assuntos
Azoximetano , Neoplasias Associadas a Colite , Sulfato de Dextrana , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Fator de Necrose Tumoral alfa , Xantofilas , Xantofilas/farmacologia , Animais , Estresse Oxidativo/efeitos dos fármacos , Azoximetano/toxicidade , Fator de Necrose Tumoral alfa/metabolismo , Humanos , Camundongos , Linhagem Celular Tumoral , Neoplasias Associadas a Colite/tratamento farmacológico , Neoplasias Associadas a Colite/prevenção & controle , Neoplasias Associadas a Colite/patologia , Masculino , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/farmacologia , NF-kappa B/metabolismo , Colite/induzido quimicamente , Colite/tratamento farmacológico , Modelos Animais de Doenças
3.
Toxins (Basel) ; 16(9)2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39330861

RESUMO

The human colonic commensal enterotoxigenic Bacteroides fragilis (ETBF) is associated with chronic colitis and colon cancer. ETBF colonization induces colitis via the Bacteroides fragilis toxin (BFT). BFT secreted by ETBF cause colon inflammation via E-cadherin cleavage/NF-κB signaling. ETBF promotes colon tumorigenesis via interleukin 17A (IL-17A)/CXCL-dependent inflammation, but its bioactive therapeutics in ETBF-promoted tumorigenesis remain unexplored. In the current study, we investigated the caffeic acid phenethyl ester (CAPE) in the murine model of ETBF colitis and tumorigenesis. In this study, we observed that CAPE treatment mitigated inflammation induced by ETBF in mice. Additionally, our findings indicate that CAPE treatment offers protective effects against ETBF-enhanced colon tumorigenesis in a mouse model of colitis-associated colon cancer induced by azoxymethane (AOM) and dextran sulfate sodium. Notably, the decrease in colon tumorigenesis following CAPE administration correlates with a reduction in the expression of IL-17A and CXCL1 in the gastrointestinal tract. The molecular mechanism for CAPE-induced protection against ETBF-mediated tumorigenesis is mediated by IL-17A/CXCL1, and by NF-κB activity in intestinal epithelial cells. Our findings indicate that CAPE may serve as a preventive agent against the development of ETBF-induced colitis and colorectal cancer (CRC).


Assuntos
Bacteroides fragilis , Ácidos Cafeicos , Colite , Álcool Feniletílico , Animais , Ácidos Cafeicos/farmacologia , Álcool Feniletílico/análogos & derivados , Álcool Feniletílico/farmacologia , Bacteroides fragilis/efeitos dos fármacos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/microbiologia , Camundongos Endogâmicos C57BL , Interleucina-17/metabolismo , Camundongos , Carcinogênese/efeitos dos fármacos , Quimiocina CXCL1/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/prevenção & controle , Neoplasias do Colo/patologia , Neoplasias do Colo/microbiologia , Masculino , Colo/efeitos dos fármacos , Colo/patologia , Colo/microbiologia , Colo/metabolismo , Toxinas Bacterianas/toxicidade , Modelos Animais de Doenças , Azoximetano/toxicidade , Sulfato de Dextrana , Metaloendopeptidases/metabolismo
4.
BMC Cancer ; 24(1): 1141, 2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39267014

RESUMO

BACKGROUND: Curcumin has been reported to have activity for prevention and therapy of CRC, yet its underlying mechanisms remain largely unknown. Recently, emerging evidence suggests that the gut microbiota and its metabolites contribute to the causation and progression of Colorectal cancer (CRC). In this study, we aimed to investigate if curcumin affects the tumorigenesis of CRC by modulating gut microbiota and its metabolites. METHODS: Forty male C57BL/6JGpt mice were randomly divided into four groups: negative control (NC), curcumin control, CRC model, and curcumin treatment (CRC-Cur) groups. CRC mouse model was induced by using azoxymethane (AOM) and dextran sodium sulfate (DSS), and the mice in CRC model and curcumin treatment groups received oral PBS or curcumin (150 mg/kg/day), respectively. Additionally, fecal samples were collected. 16 S rRNA sequencing and Liquid Chromatography Mass Spectrometry (LC-MS)-based untargeted metabolomics were used to observe the changes of intestinal flora and intestinal metabolites. RESULTS: Curcumin treatment restored colon length and structural morphology, and significantly inhibited tumor formation in AOM/DSS-induced CRC model mice. The 16S rRNA sequencing analysis indicated that the diversity and richness of core and total species of intestinal microflora in the CRC group were significantly lower than those in the NC group, which were substantially restored in the curcumin treatment group. Curcumin reduced harmful bacteria, including Ileibacterium, Monoglobus and Desulfovibrio, which were elevated in CRC model mice. Moreover, curcumin increased the abundance of Clostridia_UCG-014, Bifidobacterium and Lactobacillus, which were decreased in CRC model mice. In addition, 13 different metabolites were identified. Compared to the NC group, ethosuximide, xanthosine, and 17-beta-estradiol 3-sulfate-17-(beta-D-glucuronide) were elevated in the CRC model group, whereas curcumin treatment significantly reduced their levels. Conversely, glutamylleucine, gamma-Glutamylleucine, liquiritin, ubenimex, 5'-deoxy-5'-fluorouridine, 7,8-Dihydropteroic acid, neobyakangelicol, libenzapril, xenognosin A, and 7,4'-dihydroxy-8-methylflavan were decreased in the CRC group but notably upregulated by curcumin. Kyoto Encyclopedia of Genes and Genome (KEGG) pathway analysis revealed enrichment in seven pathways, including folate biosynthesis (P < 0.05). CONCLUSIONS: The gut microecological balance was disrupted in AOM/DSS-induced CRC mice, accompanied by metabolite dysbiosis. Curcumin restored the equilibrium of the microbiota and regulated metabolites, highly indicating that curcumin may alleviate the development of AOM/DSS induced colorectal cancer in mice by regulating intestinal flora homeostasis and intestinal metabolites.


Assuntos
Neoplasias Colorretais , Curcumina , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Curcumina/farmacologia , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/induzido quimicamente , Camundongos , Masculino , Modelos Animais de Doenças , Azoximetano/toxicidade , Carcinogênese/efeitos dos fármacos , Sulfato de Dextrana/toxicidade , RNA Ribossômico 16S/genética , Metabolômica/métodos , Humanos
5.
J Mol Histol ; 55(5): 765-783, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39122895

RESUMO

Corosolic acid (CA) is a well-known natural pentacyclic triterpene found in numerous therapeutic plants that can exhibit many bioactivities including anti-inflammatory and anti-tumor actions. The current investigation explores the chemoprotective roles of CA against azoxymethane (AOM)-induced colonic aberrant crypt foci (ACF) in rats. Thirty Sprague Dawley rats were grouped in 5 cages; Group A, normal control rats inoculated subcutaneously (sc) with two doses of normal saline and fed orally on 10% tween 20; Groups B-E received two doses (sc) of azoxymethane in two weeks and treated with either 10% tween 20 (group B) or two intraperitoneal injections of 35 mg/kg 5-fluorouracil each week for one month (group C), while group D and E treated with 30 and 60 mg/kg, respectively, for 2 months. The toxicity results showed lack of any behavioral abnormalities or mortality in rats ingested with up-to 500 mg/kg of CA. The present AOM induction caused a significant initiation of ACF characterized by an increased number, larger in size, and well-matured tissue clusters in cancer controls. AOM inoculation created a bizarrely elongated nucleus, and strained cells, and significantly lowered the submucosal glands in colon tissues of cancer controls compared to 5-FU or CA-treated rats. CA treatment led to significant suppression of ACF incidence, which could be mediated by its modulatory effects on the immunohistochemical proteins (pro-apoptotic (Bax) and reduced PCNA protein expressions in colon tissues). Moreover, CA-treated rats had improved oxidative stress-mediated cytotoxicity indicated by increased endogenous antioxidants (SOD and CAT) and reduced lipid peroxidation indicators (MDA). In addition, CA ingestion (30 and 60 mg/kg) suppressed the inflammatory cascades, indicated by decreased serum TNF-α and IL-6 cytokines and increased anti-inflammatory (IL-10) cytokines consequently preventing further tumor development. CA treatment maintained liver and kidney functions in rats exposed to AOM cytotoxicity. CA could be a viable alternative for the treatment of oxidative-related human disorders including ACF.


Assuntos
Focos de Criptas Aberrantes , Antioxidantes , Azoximetano , Antígeno Nuclear de Célula em Proliferação , Ratos Sprague-Dawley , Triterpenos , Proteína X Associada a bcl-2 , Animais , Triterpenos/farmacologia , Triterpenos/uso terapêutico , Focos de Criptas Aberrantes/patologia , Focos de Criptas Aberrantes/tratamento farmacológico , Azoximetano/toxicidade , Antioxidantes/farmacologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Masculino , Proteína X Associada a bcl-2/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Estresse Oxidativo/efeitos dos fármacos , Colo/patologia , Colo/efeitos dos fármacos , Colo/metabolismo
6.
Biomed Pharmacother ; 179: 117292, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39151314

RESUMO

A type of colorectal cancer (CRC),Colitis-associated colorectal cancer (CAC), is closely associated with chronic inflammation and gut microbiota dysbiosis. Berberine (BBR) has a long history in the treatment of intestinal diseases, which has been reported to inhibit colitis and CRC. However, the mechanism of its action is still unclear. Here, this study aimed to explore the potential protective effects of BBR on azoxymethane (AOM)/dextransulfate sodium (DSS)-induced colitis and tumor mice, and to elucidate its potential molecular mechanisms by microbiota, genes and metabolic alterations. The results showed that BBR inhibited the gut inflammation and improved the function of mucosal barrier to ameliorate AOM/DSS-induced colitis. And BBR treatment significantly reduced intestinal tumor development and ki-67 expression of intestinal tissue along with promoted apoptosis. Through microbiota analysis based on the 16 S rRNA gene, we found that BBR treatment improved intestinal microbiota imbalance in AOM/DSS-induced colitis and tumor mice, which were characterized by an increase of beneficial bacteria, for instance Akkermanisa, Lactobacillus, Bacteroides uniformis and Bacteroides acidifaciens. In addition, transcriptome analysis showed that BBR regulated colonic epithelial signaling pathway in CAC mice particularly by tryptophan metabolism and Wnt signaling pathway. Notably, BBR treatment resulted in the enrichment of amino acids metabolism and microbiota-derived SCFA metabolites. In summary, our research findings suggest that the gut microbiota-amino acid metabolism-Wnt signaling pathway axis plays critical role in maintaining intestinal homeostasis, which may provide new insights into the inhibitory effects of BBR on colitis and colon cancer.


Assuntos
Azoximetano , Berberina , Neoplasias Associadas a Colite , Colite , Sulfato de Dextrana , Microbioma Gastrointestinal , Metabolômica , Transcriptoma , Berberina/farmacologia , Berberina/uso terapêutico , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Camundongos , Azoximetano/toxicidade , Colite/microbiologia , Colite/induzido quimicamente , Colite/tratamento farmacológico , Neoplasias Associadas a Colite/microbiologia , Neoplasias Associadas a Colite/tratamento farmacológico , Neoplasias Associadas a Colite/patologia , Masculino , Camundongos Endogâmicos C57BL , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/tratamento farmacológico , Disbiose , Modelos Animais de Doenças
7.
J Gastroenterol ; 59(10): 905-920, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39141107

RESUMO

BACKGROUND: The intake of high-fructose corn syrup (HFCS) may increase the risk of colorectal cancer (CRC). This study aimed to explore the potential effects and mechanisms of resistant starch (RS) in HFCS-induced colon tumorigenesis. METHODS: The azoxymethane/dextran sodium sulfate (AOM/DSS) and ApcMin/+ mice models were used to investigate the roles of HFCS and RS in CRC in vivo. An immunohistochemistry (IHC) staining analysis was used to detect the expression of proliferation-related proteins in tissues. 16S rRNA sequencing for microbial community, gas chromatography for short-chain fatty acids (SCFAs), and mass spectrometry analysis for glycolysis products in the intestines were performed. Furthermore, lactic acid assay kit was used to detect the glycolysis levels in vitro. RESULTS: RS suppressed HFCS-induced colon tumorigenesis through reshaping the microbial community. Mechanistically, the alteration of the microbial community after RS supplement increased the levels of intestinal SCFAs, especially butyrate, leading to the suppression of glycolysis and CRC cell proliferation by downregulating HK2. CONCLUSIONS: Our study identified RS as a candidate of protective factors in CRC and may provide a potential target for HFCS-related CRC treatment.


Assuntos
Carcinogênese , Proliferação de Células , Neoplasias do Colo , Ácidos Graxos Voláteis , Glicólise , Hexoquinase , Xarope de Milho Rico em Frutose , Amido Resistente , Animais , Glicólise/efeitos dos fármacos , Camundongos , Xarope de Milho Rico em Frutose/efeitos adversos , Neoplasias do Colo/prevenção & controle , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Carcinogênese/efeitos dos fármacos , Amido Resistente/farmacologia , Proliferação de Células/efeitos dos fármacos , Ácidos Graxos Voláteis/metabolismo , Hexoquinase/metabolismo , Hexoquinase/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/induzido quimicamente , Modelos Animais de Doenças , Masculino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Sulfato de Dextrana , Azoximetano/toxicidade , Camundongos Endogâmicos C57BL , Regulação para Baixo
8.
In Vivo ; 38(5): 2300-2309, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39187313

RESUMO

BACKGROUND/AIM: Inflammatory bowel diseases and colorectal cancer are a major cause of morbidity and mortality. Amine oxidase, copper-containing 3 (AOC3) is a critical enzyme in the physiological trafficking of leukocytes and the regulation of inflammation. This study aimed to examine the effects of Aoc3 deficiency in mice models of colitis and colorectal tumorigenesis. MATERIALS AND METHODS: C57BL/6 and Aoc3 knockout mice were used for Dextran Sodium Sulfate (DSS) induced acute colitis and the Azoxymethane (AOM)/DSS model of inflammation-related colon cancer. We also evaluated the effect of Aoc3 in an Apc mutant mice model of intestinal and colonic tumorigenesis. RESULTS: We observed that Aoc3 deficient mice were more prone to colitis induced by DSS in early phases and their survival was shorter. We also showed that Aoc3 deficient mice developed more tumors both in AOM/DSS and Apc mutant mice models. Furthermore, colonic tumors in the AOM/DSS groups in Aoc3 mutant mice were generally invasive type adenocarcinomas. CONCLUSION: Aoc3 deficiency promotes colitis and colonic tumorigenesis in mouse models.


Assuntos
Amina Oxidase (contendo Cobre) , Azoximetano , Colite , Neoplasias do Colo , Sulfato de Dextrana , Modelos Animais de Doenças , Animais , Camundongos , Amina Oxidase (contendo Cobre)/genética , Amina Oxidase (contendo Cobre)/metabolismo , Azoximetano/toxicidade , Carcinogênese/genética , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Neoplasias do Colo/etiologia , Suscetibilidade a Doenças , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
J Biochem Mol Toxicol ; 38(9): e23815, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39171650

RESUMO

Drug transporters play a pivotal role in modulating drug disposition and are subject to alterations under inflammatory conditions. This study aimed to elucidate the intricate expression patterns of drug transporters during both acute and chronic inflammation, which are closely linked to malignant transformation. To investigate acute inflammation, we employed an in vitro model by subjecting Caco-2 cells to various inflammatory stimuli (IL-1ß, TNF-α, or LPS) individually or in combination. The successful induction of inflammation was confirmed by robust increases in IL-6 and NO production. Notably, inflamed Caco-2 cells exhibited significantly diminished levels of ABCB1 and ABCG2, while the expression of ABCC2 was upregulated. For chronic inflammation induction in vivo, we employed the well-established AOM/DSS mouse model known for its association with colitis-driven tumorigenesis. Persistent inflammation was effectively monitored throughout the experiment via elevated IL-6 and NO levels. The sequential stages of tumorigenesis were confirmed through Ki-67 immunohistochemistry. Intriguingly, we observed gradual alterations in the expression patterns of the studied drug transporters during stepwise induction, with ABCB1, ABCG2, and ABCC1 showing downregulation and ABCC2 exhibiting upregulation. Immunohistochemistry further revealed dynamic changes in the expression of ABCB1 and ABCC2 during the induction cycles, closely paralleling the gradual increase in Ki-67 expression observed during the development of precancerous lesions. Collectively, our findings underscore the significant impact of inflammation on drug transporter expression, potentially influencing the process of malignant transformation of the colon.


Assuntos
Azoximetano , Neoplasias do Colo , Inflamação , Proteína 2 Associada à Farmacorresistência Múltipla , Humanos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Animais , Células CACO-2 , Camundongos , Azoximetano/toxicidade , Inflamação/metabolismo , Inflamação/induzido quimicamente , Inflamação/patologia , Carcinogênese/metabolismo , Carcinogênese/induzido quimicamente , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/biossíntese , Interleucina-6/metabolismo , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/induzido quimicamente , Masculino
10.
Bull Exp Biol Med ; 177(1): 162-168, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38960963

RESUMO

In adult male C57BL/6 mice with high (HR) and low (LR) resistance to hypoxia, morphological features of colon tumors and blood parameters were evaluated 70 days after intraperitoneal injection of azoxymethane and subsequent consumption of 3 cycles of dextran sulfate sodium. On macroscopic analysis, tumors were found in the distal colon in 35% (7 of 20 animals) of HR and 31% (4 of 13 animals) of LR animals. Microscopic analysis of the distal colon revealed tumors in 75% (15 of 20 animals) of HR and 69% (9 of 13 animals) of LR mice. The tumors were presented by areas of glandular intraepithelial neoplasia and adenocarcinomas; the incidence and the area of the tumors did not differ in groups of HR and LR mice. The number of neuroendocrine and goblet cells in the distal colon mucosa in the areas of tumors was similar in the compared groups. However, in both HR and LR mice of the experimental groups, the content of goblet cells in tumors was lower and the content of endocrine cells was higher than in the corresponding control groups. In the peripheral blood, the erythrocyte count and hemoglobin content decreased in HR and LR mice of the experimental groups; the relative number of monocytes increased only in HR mice and the absolute number of lymphocytes and monocytes decreased in LR mice. Thus, 70 days after azoxymethane administration and dextran sulfate sodium consumption, the tumors in mice were presented by glandular intraepithelial neoplasia and adenocarcinomas, and their incidence and area did not differ between animals with different tolerance to hypoxia.


Assuntos
Adenocarcinoma , Azoximetano , Neoplasias do Colo , Sulfato de Dextrana , Camundongos Endogâmicos C57BL , Animais , Camundongos , Neoplasias do Colo/patologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Masculino , Sulfato de Dextrana/toxicidade , Azoximetano/toxicidade , Adenocarcinoma/patologia , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/metabolismo , Hipóxia/patologia , Colo/patologia , Células Caliciformes/patologia , Células Caliciformes/metabolismo , Mucosa Intestinal/patologia , Hemoglobinas/metabolismo , Monócitos/patologia , Monócitos/metabolismo , Contagem de Eritrócitos
11.
Genes (Basel) ; 15(7)2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-39062614

RESUMO

The incidence of colorectal cancer (CRC) is closely linked to metabolic diseases. Accumulating evidence suggests the regulatory role of AMP-activated protein kinase (AMPK) in cancer metabolic reprogramming. In this study, wild-type and AMPK knockout mice were subjected to azoxymethane-induced and dextran sulfate sodium (AOM/DSS)-promoted colitis-associated CRC induction. A stable AMPK-deficient Caco-2 cell line was also established for the mechanistic studies. The data showed that AMPK deficiency accelerated CRC development, characterized by increased tumor number, tumor size, and hyperplasia in AOM/DSS-treated mice. The aggravated colorectal tumorigenesis resulting from AMPK ablation was associated with reduced α-ketoglutarate production and ten-eleven translocation hydroxylase 2 (TET2) transcription, correlated with the reduced mismatch repair protein mutL homolog 1 (MLH1) protein. Furthermore, in AMPK-deficient Caco-2 cells, the mRNA expression of mismatch repair and tumor suppressor genes, intracellular α-ketoglutarate, and the protein level of TET2 were also downregulated. AMPK deficiency also increased hypermethylation in the CpG islands of Mlh1 in both colonic tissues and Caco-2 cells. In conclusion, AMPK deficiency leads to reduced α-ketoglutarate concentration and elevates the suppressive epigenetic modifications of tumor suppressor genes in gut epithelial cells, thereby increasing the risk of colorectal tumorigenesis. Given the modifiable nature of AMPK activity, it holds promise as a prospective molecular target for the prevention and treatment of CRC.


Assuntos
Proteínas Quinases Ativadas por AMP , Azoximetano , Carcinogênese , Neoplasias Colorretais , Metilação de DNA , Dioxigenases , Animais , Humanos , Camundongos , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Azoximetano/toxicidade , Azoximetano/efeitos adversos , Células CACO-2 , Carcinogênese/genética , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/etiologia , Sulfato de Dextrana/toxicidade , Dioxigenases/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Ácidos Cetoglutáricos/metabolismo , Camundongos Knockout , Proteína 1 Homóloga a MutL/genética , Proteína 1 Homóloga a MutL/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
12.
J Ethnopharmacol ; 334: 118597, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-39034016

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Huangqin Tang (HQT), a traditional Chinese medicine formula, is commonly used in clinical practice for the treatment of inflammatory bowel diseases. It has been reported that HQT exerts antitumor effects on colitis-associated colorectal cancer (CAC). However, the mechanism by which HQT interferes with the inflammation-to-cancer transformation remains unclear. AIMS OF THE STUDY: The purpose of this study was to dynamically evaluate the efficacy of HQT in alleviating or delaying CAC and to reveal the underlying mechanism. METHODS: We established a mouse model of CAC using azoxymethane combined with 1.5% dextran sodium sulphate. The efficacy of HQT was evaluated based on pathological sections and serum biochemical indices. Subsequently, amino acids (AAs) metabolism analyses were performed using ultra-performance liquid chromatography-tandem mass spectrometry, and the phosphatidylinositol 3 kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) pathway was detected by western blotting. RESULTS: The data demonstrated that HQT could alleviate the development of CAC in the animal model. HQT effectively reduced the inflammatory response, particularly interleukin-6 (IL-6), in the inflammation induction stage, as well as in the stages of proliferation initiation and tumorigenesis. During the proliferation initiation and tumorigenesis stages, immunohistochemistry staining showed that the expression of the proliferation marker Ki67 was reduced, while apoptosis was increased in the HQT group. Accordingly, HQT substantially decreased the levels of specific AAs in the colon with CAC, including glutamic acid, glutamine, arginine, and isoleucine. Furthermore, HQT significantly inhibited the activated PI3K/AKT/mTOR pathway, which may contribute to suppression of cell proliferation and enhancement of apoptosis. CONCLUSION: HQT is effective in alleviating and delaying the colon "inflammation-to-cancer". The mechanism of action may involve HQT maintained AAs metabolism homeostasis and regulated PI3K/AKT/mTOR pathway, so as to maintain the balance between proliferation and apoptosis, and then interfere in the occurrence and development of CAC.


Assuntos
Aminoácidos , Neoplasias Associadas a Colite , Sulfato de Dextrana , Medicamentos de Ervas Chinesas , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Serina-Treonina Quinases TOR , Animais , Serina-Treonina Quinases TOR/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Masculino , Neoplasias Associadas a Colite/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Camundongos , Azoximetano/toxicidade , Modelos Animais de Doenças , Homeostase/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Camundongos Endogâmicos C57BL , Colite/tratamento farmacológico , Colite/induzido quimicamente , Colite/complicações , Colite/metabolismo , Apoptose/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Proliferação de Células/efeitos dos fármacos
13.
J Ethnopharmacol ; 334: 118541, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38992403

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Based on the core pathogenesis of hepatosplenic disorder and qi transformation disorder in ulcerative colitis, Tong-Xie-Yao-Fang (TXYF) is a classical traditional Chinese medicine commonly used to treat ulcerative colitis. Our study revealed that it has the potential to prevent colitis-associated colorectal cancer, which embodies the academic concept in traditional Chinese medicine of treating the disease before it develops. AIM OF THE STUDY: This study was aimed at evaluating the therapeutic role of TXYF in treating colitis-associated colorectal cancer and exploring its possible underlying mechanisms. MATERIALS AND METHODS: A colitis-associated colorectal cancer model was established in mice using azoxymethane and dextran sulfate sodium salt to examine the therapeutic effect of TXYF. The mouse body weights were observed. Hematoxylin-eosin staining was used to evaluate mouse colon histopathology. Colon cancer cells and colon epithelial cells were used to explore the potential molecular mechanisms. The proliferation and apoptosis of cells were detected by CCK8 and cell colony assays, flow cytometry and western blotting. The epithelial-mesenchymal transition (EMT) and mitophagy markers were examined by immunohistochemistry, western blotting, quantitative real-time PCR and immunofluorescence staining. RESULTS: TXYF inhibited the tumorigenesis of mice with colitis-associated colorectal cancer and the growth of inflammatory colon cells. TXYF induced mitophagy in colon cancer cells through the PTEN-induced putative kinase 1 (PINK1)/Parkin pathway to reverse EMT, which was consistent with the results in mice with colitis-associated colorectal cancer. CONCLUSIONS: The results of the present study demonstrated that TXYF effectively inhibited the progression of colitis-associated colorectal cancer through the PINK1/Parkin pathway, which provides new evidence for prevention strategies for this disease.


Assuntos
Neoplasias Associadas a Colite , Medicamentos de Ervas Chinesas , Células Epiteliais , Mitofagia , Animais , Mitofagia/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Camundongos , Neoplasias Associadas a Colite/tratamento farmacológico , Neoplasias Associadas a Colite/patologia , Neoplasias Associadas a Colite/prevenção & controle , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Azoximetano/toxicidade , Masculino , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sulfato de Dextrana , Colo/efeitos dos fármacos , Colo/patologia , Colo/metabolismo , Camundongos Endogâmicos C57BL , Ubiquitina-Proteína Ligases/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Colite/tratamento farmacológico , Colite/complicações , Colite/induzido quimicamente , Proteínas Quinases
14.
World J Surg Oncol ; 22(1): 192, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39054486

RESUMO

BACKGROUND: More and more studies showed that gut microbiota was closely related to the development of colorectal cancer (CRC). However, the specific pathway of gut microbiota regulating CRC development is still unknown. METHODS: We collected fecal samples from 14 CRC patients and 20 normal volunteers for 16 S sequencing analysis. At the same time, 14 CRC patients' tumors and their adjacent tissues were collected for the detection of STING pathway related protein level. Mice were injected with azoxymethane (AOM) to establish an animal model of CRC, and antibiotics were given at the same time to evaluate the influence of gut microbiota on STING pathway and whether it was involved in regulating the tumor development of CRC mice. RESULTS: The sequencing results showed that compared with the normal group, the gut microbiota gut microbiota of CRC patients changed significantly at different species classification levels. At the level of genus, Akkermansia, Ligilactobacillus and Subdoligranulum increased the most in CRC patients, while Bacteroides and Dialister decreased sharply. The expression of STING-related protein was significantly down-regulated in CRC tumor tissues. Antibiotic treatment of CRC mice can promote the development of tumor and inhibit the activation of STING pathway. CONCLUSION: Gut microbiota participates in CRC progress by mediating STING pathway activation.


Assuntos
Neoplasias Colorretais , Progressão da Doença , Microbioma Gastrointestinal , Proteínas de Membrana , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Animais , Camundongos , Humanos , Proteínas de Membrana/metabolismo , Masculino , Feminino , Estudos de Casos e Controles , Pessoa de Meia-Idade , Transdução de Sinais , Prognóstico , Azoximetano/toxicidade , Fezes/microbiologia
15.
Mol Biol Rep ; 51(1): 704, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38824233

RESUMO

BACKGROUND: Tumor modeling using organoids holds potential in studies of cancer development, enlightening both the intracellular and extracellular molecular mechanisms behind different cancer types, biobanking, and drug screening. Intestinal organoids can be generated in vitro using a unique type of adult stem cells which are found at the base of crypts and are characterized by their high Lgr5 expression levels. METHODS AND RESULTS: In this study, we successfully established intestinal cancer organoid models by using both the BALB/c derived and mouse embryonic stem cells (mESCs)-derived intestinal organoids. In both cases, carcinogenesis-like model was developed by using azoxymethane (AOM) treatment. Carcinogenesis-like model was verified by H&E staining, immunostaining, relative mRNA expression analysis, and LC/MS analysis. The morphologic analysis demonstrated that the number of generated organoids, the number of crypts, and the intensity of the organoids were significantly augmented in AOM-treated intestinal organoids compared to non-AOM-treated ones. Relative mRNA expression data revealed that there was a significant increase in both Wnt signaling pathway-related genes and pluripotency transcription factors in the AOM-induced intestinal organoids. CONCLUSION: We successfully developed simple carcinogenesis-like models using mESC-based and Lgr5 + stem cell-based intestinal organoids. Intestinal organoid based carcinogenesi models might be used for personalized cancer therapy in the future.


Assuntos
Azoximetano , Carcinogênese , Células-Tronco Embrionárias Murinas , Organoides , Via de Sinalização Wnt , Animais , Organoides/metabolismo , Organoides/patologia , Camundongos , Azoximetano/toxicidade , Carcinogênese/patologia , Carcinogênese/induzido quimicamente , Carcinogênese/genética , Células-Tronco Embrionárias Murinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Camundongos Endogâmicos BALB C , Intestinos/patologia , Neoplasias Intestinais/patologia , Neoplasias Intestinais/induzido quimicamente , Neoplasias Intestinais/genética , Neoplasias Intestinais/metabolismo , Modelos Animais de Doenças , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia
16.
J Ethnopharmacol ; 333: 118411, 2024 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-38824980

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Previous studies have revealed that a high-fat diet (HFD) promotes the progression of colorectal cancer (CRC) in close association with disturbances in the intestinal flora and metabolic disorders. Xianglian pill (XLP) is a well-established traditional prescription with unique advantages in controlling intestinal flora imbalance and inflammation. However, its therapeutic effects on HFD-related CRC remain largely unknown. AIM OF THE STUDY: The primary objective of this research was to investigate the anticancer mechanism of XLP in countering HFD-related CRC. MATERIALS AND METHODS: The protective effect of XLP was evaluated using azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced CRC model of mice exposed to a HFD. The degree of colorectal carcinogenesis, including body weight, colon length, and histopathology, was measured in mice treated with XLP and untreated mice. The effect of XLP on gut microbiota and its metabolites was detected using 16S rDNA and liquid chromatography/mass spectrometry analysis. Furthermore, a "pseudo-sterile" mouse model was constructed using antibiotics (Abx) to verify whether the gut microbiota and metabolites play a role in the pathogenesis of CRC. RESULTS: XLP inhibited colorectal tumorigenesis in a dose-dependent fashion. Our findings also highlighted that XLP protected the integrity of the intestinal barrier by reducing the expression of pro-inflammatory cytokines, such as IL-6 and TNF-α, as well as the infiltration of pro-inflammatory macrophages. Mechanistically, XLP inhibited the TLR4/MyD88 pathway. Notably, the XLP treatment increased the proportion of probiotics (particularly Akkermansia) and significantly reduced fecal deoxycholic acid (DCA), a microbiota-derived metabolite of bile acids (BA) closely related to Muribaculaceae. Furthermore, after Abx treatment, XLP showed no clear antitumor effects on CRC. Simultaneously, DCA-supplemented feedings promoted colorectal tumorigenesis and provoked obvious colonic inflammation, M1 macrophage infiltration, and colonic injury. In vitro, the results of RAW-264.7 macrophages and normal intestinal epithelial cells treated with DCA corroborated our in vivo findings, demonstrating consistent patterns in inflammatory responses and intestinal barrier protein expression. CONCLUSION: Our findings suggest that XLP inhibits colorectal cancer associated with HFD via inactivating TLR4/MyD88 by remodeling gut microbiota composition and BA metabolism.


Assuntos
Ácidos e Sais Biliares , Neoplasias Colorretais , Dieta Hiperlipídica , Medicamentos de Ervas Chinesas , Microbioma Gastrointestinal , Transdução de Sinais , Animais , Masculino , Camundongos , Azoximetano/toxicidade , Ácidos e Sais Biliares/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Sulfato de Dextrana , Dieta Hiperlipídica/efeitos adversos , Medicamentos de Ervas Chinesas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 4 Toll-Like/metabolismo
17.
Cell Mol Gastroenterol Hepatol ; 18(3): 101356, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38750899

RESUMO

BACKGROUND & AIMS: Dysregulated colonic epithelial cell (CEC) proliferation is a critical feature in the development of colorectal cancer. We show that NF-κB-inducing kinase (NIK) attenuates colorectal cancer through coordinating CEC regeneration/differentiation via noncanonical NF-κB signaling that is unique from canonical NF-kB signaling. METHODS: Initial studies evaluated crypt morphology/functionality, organoid generation, transcriptome profiles, and the microbiome. Inflammation and inflammation-induced tumorigenesis were initiated in whole-body NIK knockout mice (Nik-/-) and conditional-knockout mice following administration of azoxymethane and dextran sulfate sodium. RESULTS: Human transcriptomic data revealed dysregulated noncanonical NF-kB signaling. In vitro studies evaluating Nik-/- crypts and organoids derived from mature, nondividing CECs, and colonic stem cells exhibited increased accumulation and stunted growth, respectively. Transcriptomic analysis of Nik-/- cells revealed gene expression signatures associated with altered differentiation-regeneration. When assessed in vivo, Nik-/- mice exhibited more severe colitis with dextran sulfate sodium administration and an altered microbiome characterized by increased colitogenic microbiota. In the inflammation-induced tumorigenesis model, we observed both increased tumor burdens and inflammation in mice where NIK is knocked out in CECs (NikΔCEC). Interestingly, this was not recapitulated when NIK was conditionally knocked out in myeloid cells (NikΔMYE). Surprisingly, conditional knockout of the canonical pathway in myeloid cells (RelAΔMYE) revealed decreased tumor burden and inflammation and no significant changes when conditionally knocked out in CECs (RelAΔCEC). CONCLUSIONS: Dysregulated noncanonical NF-κB signaling is associated with the development of colorectal cancer in a tissue-dependent manner and defines a critical role for NIK in regulating gastrointestinal inflammation and regeneration associated with colorectal cancer.


Assuntos
Neoplasias Colorretais , Células Epiteliais , Camundongos Knockout , NF-kappa B , Quinase Induzida por NF-kappaB , Proteínas Serina-Treonina Quinases , Regeneração , Transdução de Sinais , Animais , Humanos , Camundongos , Azoximetano/toxicidade , Diferenciação Celular , Proliferação de Células , Colite/patologia , Colite/induzido quimicamente , Colo/patologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Mucosa Intestinal/patologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , NF-kappa B/metabolismo , Organoides/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética
18.
Cancer Lett ; 593: 216940, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38729554

RESUMO

Decreased levels of ß-hydroxybutyrate (BHB), a lipid metabolic intermediate known to slow the progression of colorectal cancer (CRC), have been observed in the colon mucosa of patients with inflammatory bowel diseases (IBD). In particular, patients with recurrent IBD present an increased risk of developing colitis-associated colorectal cancer (CAC). The role and molecular mechanism of BHB in the inflammatory and carcinogenic process of CAC remains unclear. Here, the anti-tumor effect of BHB was investigated in the Azoxymethane (AOM)/Dextran Sulfate Sodium (DSS)-induced CAC model and tumor organoids derivatives. The underlying mechanisms were studied using transcriptome and non-target metabolomic assay and further validated in colon tumor cell lineage CT26 in vitro. The tumor tissues and the nearby non-malignant tissues from colon cancer patients were collected to measure the expression levels of ketogenic enzymes. The exogenous BHB supplement lightened tumor burden and angiogenesis in the CAC model. Notably, transcriptome analysis revealed that BHB effectively decreased the expression of VEGFA in the CAC tumor mucosa. In vitro, BHB directly reduced VEGFA expression in hypoxic-treated CT26 cells by targeting transcriptional factor HIF-1α. Conversely, the deletion of HIF-1α largely reversed the inhibitory effect of BHB on CAC tumorigenesis. Additionally, decreased expression of ketogenesis-related enzymes in tumor tissues were associated with poor survival outcomes in patients with colon cancer. In summary, BHB carries out anti-angiogenic activity in CAC by regulating HIF-1α/VEGFA signaling. These findings emphasize the role of BHB in CAC and may provide novel perspectives for the prevention and treatment of colonic tumors.


Assuntos
Ácido 3-Hidroxibutírico , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neovascularização Patológica , Ácido 3-Hidroxibutírico/farmacologia , Ácido 3-Hidroxibutírico/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Animais , Camundongos , Humanos , Neoplasias Associadas a Colite/patologia , Neoplasias Associadas a Colite/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Linhagem Celular Tumoral , Carcinogênese/efeitos dos fármacos , Masculino , Azoximetano/toxicidade , Colite/complicações , Colite/metabolismo , Colite/patologia , Colite/induzido quimicamente , Sulfato de Dextrana , Modelos Animais de Doenças , Angiogênese
19.
Biomed Pharmacother ; 175: 116580, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38723513

RESUMO

Colitis-associated cancer (CAC) in inflammatory bowel diseases exhibits more aggressive behavior than sporadic colorectal cancer; however, the molecular mechanisms remain unclear. No definitive preventative agent against CAC is currently established in the clinical setting. We investigated the molecular mechanisms of CAC in the azoxymethane/dextran sulfate sodium (AOM/DSS) mouse model and assessed the antitumor efficacy of erlotinib, a small molecule inhibitor of the epidermal growth factor receptor (EGFR). Erlotinib premixed with AIN-93 G diet at 70 or 140 parts per million (ppm) inhibited tumor multiplicity significantly by 96%, with ∼60% of the treated mice exhibiting zero polyps at 12 weeks. Bulk RNA-sequencing revealed more than a thousand significant gene alterations in the colons of AOM/DSS-treated mice, with KEGG enrichment analysis highlighting 46 signaling pathways in CAC development. Erlotinib altered several signaling pathways and rescued 40 key genes dysregulated in CAC, including those involved in the Hippo and Wnt signaling. These findings suggest that the clinically-used antitumor agent erlotinib might be repurposed for suppression of CAC, and that further studies are warranted on the crosstalk between dysregulated Wnt and EGFR signaling in the corresponding patient population.


Assuntos
Azoximetano , Neoplasias Associadas a Colite , Sulfato de Dextrana , Modelos Animais de Doenças , Cloridrato de Erlotinib , Animais , Cloridrato de Erlotinib/farmacologia , Neoplasias Associadas a Colite/patologia , Neoplasias Associadas a Colite/tratamento farmacológico , Camundongos , Azoximetano/toxicidade , Receptores ErbB/metabolismo , Receptores ErbB/genética , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Camundongos Endogâmicos C57BL , Masculino , Transdução de Sinais/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Colite/tratamento farmacológico , Colite/induzido quimicamente , Colite/complicações , Colite/patologia
20.
Int J Mol Sci ; 25(9)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38731854

RESUMO

Factors that reduce the risk of developing colorectal cancer include biologically active substances. In our previous research, we demonstrated the anti-inflammatory, immunomodulatory, and antioxidant effects of oat beta-glucans in gastrointestinal disease models. The aim of this study was to investigate the effect of an 8-week consumption of a diet supplemented with low-molar-mass oat beta-glucan in two doses on the antioxidant potential, inflammatory parameters, and colonic metabolomic profile in azoxymethane(AOM)-induced early-stage colorectal cancer in the large intestine wall of rats. The results showed a statistically significant effect of AOM leading to the development of neoplastic changes in the colon. Consumption of beta-glucans induced changes in colonic antioxidant potential parameters, including an increase in total antioxidant status, a decrease in the superoxide dismutase (SOD) activity, and a reduction in thiobarbituric acid reactive substance (TBARS) concentration. In addition, beta-glucans decreased the levels of pro-inflammatory interleukins (IL-1α, IL-1ß, IL-12) and C-reactive protein (CRP) while increasing the concentration of IL-10. Metabolomic studies confirmed the efficacy of oat beta-glucans in the AOM-induced early-stage colon cancer model by increasing the levels of metabolites involved in metabolic pathways, such as amino acids, purine, biotin, and folate. In conclusion, these results suggest a wide range of mechanisms involved in altering colonic metabolism during the early stage of carcinogenesis and a strong influence of low-molar-mass oat beta-glucan, administered as dietary supplement, in modulating these mechanisms.


Assuntos
Antioxidantes , Azoximetano , Neoplasias Colorretais , beta-Glucanas , Animais , beta-Glucanas/farmacologia , Azoximetano/toxicidade , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/patologia , Ratos , Masculino , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Modelos Animais de Doenças , Avena/química , Superóxido Dismutase/metabolismo , Colo/metabolismo , Colo/patologia , Colo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar , Proteína C-Reativa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA