Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 326(1): G53-G66, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37933447

RESUMEN

Neutrophils are abundant immune cells in the colon tumor microenvironment. Studies have shown that neutrophils are recruited into hypoxic foci in colon cancer. However, the impact of hypoxia signaling on neutrophil function and its involvement in colon tumorigenesis remain unclear. To address this, we generated mice with a deletion of hypoxia-inducible factor (HIF)-1α or HIF-2α in neutrophils driven by the MRP8Cre (HIF-1αΔNeu) or (HIF-2αΔNeu) and littermate controls. In an azoxymethane (AOM)/dextran sulfate sodium (DSS) model of colon cancer, the disruption of neutrophils-HIF-1α did not result in any significant changes in body weight, colon length, tumor size, proliferation, or burden. However, the disruption of HIF-2α in neutrophils led to a slight increase in body weight, a significant decrease in the number of tumors, and a reduction in tumor size and volume compared with their littermate controls. Histological analysis of colon tissue from mice with HIF-2α-deficient neutrophils revealed notable reductions in proliferation as compared with control mice. In addition, we observed reduced levels of proinflammatory cytokines, such as TNF-α and IL-1ß, in neutrophil-specific HIF-2α-deficient mice in both the tumor tissue as well as the neutrophils. Importantly, it is worth noting that the reduced tumorigenesis associated with HIF-2α deficiency in neutrophils was not evident in already established syngeneic tumors or a DSS-induced inflammation model, indicating a potential role of HIF-2α specifically in colon tumorigenesis. In conclusion, we found that the loss of neutrophil-specific HIF-2α slows colon tumor growth and progression by reducing the levels of inflammatory mediators.NEW & NOTEWORTHY Despite the importance of hypoxia and neutrophils in colorectal cancer (CRC), the contribution of neutrophil-specific HIFs to colon tumorigenesis is not known. We describe that neutrophil HIF-1α has no impact on colon cancer, whereas neutrophil HIF-2α loss reduces CRC growth by decreasing proinflammatory and immunosuppressive cytokines. Furthermore, neutrophil HIF-2α does not reduce preestablished tumor growth or inflammation-induced colitis. The present study offers novel potential of neutrophil HIF-2α as a therapeutic target in CRC.


Asunto(s)
Neoplasias Asociadas a Colitis , Neoplasias del Colon , Animales , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Peso Corporal , Carcinogénesis/patología , Transformación Celular Neoplásica/patología , Neoplasias Asociadas a Colitis/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Citocinas , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inflamación , Neutrófilos , Microambiente Tumoral
2.
Mol Oncol ; 17(11): 2380-2395, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37341064

RESUMEN

The incidence of colitis-associated colorectal cancer (CAC) has increased due to a high-nutrient diet, increased environmental stimuli and inherited gene mutations. To adequately treat CAC, drugs should be developed by identifying novel therapeutic targets. E3 ubiquitin-protein ligase pellino homolog 3 (pellino 3; Peli3) is a RING-type E3 ubiquitin ligase involved in inflammatory signalling; however, its role in the development and progression of CAC has not been elucidated. In this study, we studied Peli3-deficient mice in an azoxymethane/dextran sulphate sodium-induced CAC model. We observed that Peli3 promotes colorectal carcinogenesis with increased tumour burden and oncogenic signalling pathways. Ablation of Peli3 reduced inflammatory signalling activation at the early stage of carcinogenesis. Mechanistic studies indicate that Peli3 enhances toll-like receptor 4 (TLR4)-mediated inflammation through ubiquitination-dependent degradation of interferon regulatory factor 4, a negative regulator of TLR4 in macrophages. Our study suggests an important molecular link between Peli3 and colonic inflammation-mediated carcinogenesis. Furthermore, Peli3 can be a therapeutic target in the prevention and treatment of CAC.


Asunto(s)
Neoplasias Asociadas a Colitis , Receptor Toll-Like 4 , Animales , Ratones , Carcinogénesis/genética , Neoplasias Asociadas a Colitis/genética , Inflamación/complicaciones , Factores Reguladores del Interferón/metabolismo , Ratones Endogámicos C57BL , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
J Immunol Res ; 2023: 7040113, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36741232

RESUMEN

Ulcerative colitis (UC) is a complex intestinal inflammation with an increasing risk of colitis-associated colorectal cancer (CAC). However, the pathogenesis is still unclear between active UC and inactive UC. Recently, it has been reported that pyroptosis-related genes (PRGs) are closely associated with inflammatory disease activity. Nevertheless, the specific roles of PRGs in the progression and treatment of UC and CAC remain unclear. In this study, we identified 30 differentially expressed PRGs based on the immune landscape of active and inactive UC samples. Meanwhile, weighted gene coexpression network analysis was applied to explore important genes associated with active UC. By intersecting with the differentially expressed PRGs, CASP5, GBP1, GZMB, IL1B, and IRF1 were selected as key PRGs to construct a pyroptosis-related signature (PR-signature). Then, logistic regression analysis was performed to validate the PR-signature and establish a pyroptosis-related score (PR-Score). We demonstrated that PR-Score had a powerful ability to distinguish active UC from inactive UC in multiple datasets. Besides, PR-Score was positively correlated with immune cell infiltration and inflammatory microenvironment in UC. Lower PR-Score was associated with a better response to anti-TNF therapy for patients with UC. Additionally, high-PR-Score was found to suppress CAC and improve the survival outcomes of patients with colorectal cancer. Finally, the levels of the PR-signature genes were validated both in vitro and in vivo. These findings can improve our understanding of PRGs in UC and provide new markers for predicting the occurrence of active UC or CAC and the treatment of UC.


Asunto(s)
Colitis Ulcerosa , Neoplasias Asociadas a Colitis , Humanos , Piroptosis , Neoplasias Asociadas a Colitis/etiología , Neoplasias Asociadas a Colitis/genética , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/genética , Inhibidores del Factor de Necrosis Tumoral , Caspasas , Microambiente Tumoral/genética
4.
Int J Mol Sci ; 23(23)2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36499472

RESUMEN

Colorectal cancer (CRC) is among the best examples for depicting the relationship between inflammation and cancer. The introduction of new therapeutics targeting inflammatory mediators showed a marked decrease in the overall risk of CRC, although their chemopreventive potential is still debated. Specifically, a monoclonal antibody that blocks tumor necrosis factor (TNF), infliximab, increases CRC risk in inflammatory bowel disease patients. To address the axis between TNF and CRC development and progression, we depleted the Tnf from our previously established murine model of colitis-associated cancer (CAC), the Winnie-ApcMin/+ line. We characterized the new Winnie-APCMin/+-TNF-KO line through macroscopical and microscopical analyses. Surprisingly, the latter demonstrated that the deletion of Tnf in Winnie-ApcMin/+ mice resulted in an initial reduction in dysplastic lesion incidence in 5-week-old mice followed by a faster disease progression at 8 weeks. Histological data were confirmed by the molecular profiling obtained from both the real-time PCR analysis of the whole tissue and the RNA sequencing of the macrodissected tumoral lesions from Winnie-APCMin/+-TNF-KO distal colon at 8 weeks. Our results highlight that TNF could exert a dual role in CAC, supporting the promotion of neoplastic lesions onset in the early stage of the disease while inducing their reduction during disease progression.


Asunto(s)
Neoplasias Asociadas a Colitis , Colitis , Neoplasias Colorrectales , Ratones , Animales , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/complicaciones , Inflamación/complicaciones , Factor de Necrosis Tumoral alfa/genética , Progresión de la Enfermedad , Neoplasias Colorrectales/genética , Colitis/complicaciones , Colitis/genética , Modelos Animales de Enfermedad
5.
JCI Insight ; 7(22)2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36509284

RESUMEN

Colitis-associated colorectal cancer (CAC) is a severe complication of inflammatory bowel disease (IBD). HIF-prolyl hydroxylases (PHD1, PHD2, and PHD3) control cellular adaptation to hypoxia and are considered promising therapeutic targets in IBD. However, their relevance in the pathogenesis of CAC remains elusive. We induced CAC in Phd1-/-, Phd2+/-, Phd3-/-, and WT mice with azoxymethane (AOM) and dextran sodium sulfate (DSS). Phd1-/- mice were protected against chronic colitis and displayed diminished CAC growth compared with WT mice. In Phd3-/- mice, colitis activity and CAC growth remained unaltered. In Phd2+/- mice, colitis activity was unaffected, but CAC growth was aggravated. Mechanistically, Phd2 deficiency (i) increased the number of tumor-associated macrophages in AOM/DSS-induced tumors, (ii) promoted the expression of EGFR ligand epiregulin in macrophages, and (iii) augmented the signal transducer and activator of transcription 3 and extracellular signal-regulated kinase 1/2 signaling, which at least in part contributed to aggravated tumor cell proliferation in colitis-associated tumors. Consistently, Phd2 deficiency in hematopoietic (Vav:Cre-Phd2fl/fl) but not in intestinal epithelial cells (Villin:Cre-Phd2fl/fl) increased CAC growth. In conclusion, the 3 different PHD isoenzymes have distinct and nonredundant effects, promoting (PHD1), diminishing (PHD2), or neutral (PHD3), on CAC growth.


Asunto(s)
Neoplasias Asociadas a Colitis , Colitis , Animales , Ratones , Azoximetano , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/metabolismo , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/metabolismo , Células Epiteliales/metabolismo , Prolil Hidroxilasas/metabolismo
6.
Int J Mol Sci ; 23(19)2022 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-36232773

RESUMEN

Chronic relapsing inflammatory bowel disease is strongly linked to an increased risk of colitis-associated cancer (CAC). One of the well-known inflammatory carcinogenesis pathways, phosphatidylinositol 3-kinase (PI3K), was identified to be a crucial mechanism in long-standing ulcerative colitis (UC). The goal of this study was to identify somatic variants in the cytokine-induced PI3K-related genes in UC, colorectal cancer (CRC) and CAC. Thirty biopsies (n = 8 long-standing UC, n = 11 CRC, n = 8 paired normal colorectal mucosa and n = 3 CAC) were subjected to targeted sequencing on 13 PI3K-related genes using Illumina sequencing and the SureSelectXT Target Enrichment System. The Genome Analysis Toolkit was used to analyze variants, while ANNOVAR was employed to detect annotations. There were 5116 intronic, 355 exonic, 172 untranslated region (UTR) and 59 noncoding intronic variations detected across all samples. Apart from a very small number of frameshifts, the distribution of missense and synonymous variants was almost equal. We discovered changed levels of IL23R, IL12Rß1, IL12Rß2, TYK2, JAK2 and OSMR in more than 50% of the samples. The IL23R variant in the UTR region, rs10889677, was identified to be a possible variant that might potentially connect CAC with UC and CRC. Additional secondary structure prediction using RNAfold revealed that mutant structures were more unstable than wildtype structures. Further functional research on the potential variants is, therefore, highly recommended since it may provide insight on the relationship between inflammation and cancer risk in the cytokine-induced PI3K pathway.


Asunto(s)
Colitis Ulcerosa , Neoplasias Asociadas a Colitis , Neoplasias Colorrectales , Citocinas , Fosfatidilinositol 3-Quinasa , Colitis Ulcerosa/genética , Neoplasias Asociadas a Colitis/genética , Neoplasias Colorrectales/genética , Citocinas/genética , Humanos , Recurrencia Local de Neoplasia/genética , Fosfatidilinositol 3-Quinasa/genética , Regiones no Traducidas
7.
Cell Mol Gastroenterol Hepatol ; 14(4): 789-811, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35809803

RESUMEN

BACKGROUND & AIMS: MUC1 is abnormally expressed in colorectal cancer, including colitis-associated colorectal cancer (CAC), but its role in tumorigenesis is unclear. This study investigated MUC1's effects in murine models of colitis and CAC and elucidated mechanisms of action. METHODS: Colitis and CAC were induced in mice by exposure to dextran sodium sulfate or azoxymethane plus dextran sodium sulphate. Clinical parameters, immune cell infiltration, and tumor development were monitored throughout disease progression. Experiments in knockout mice and bone marrow chimeras were combined with an exploration of immune cell abundance and function. RESULTS: Deficiency of Muc1 suppressed inflammation, inhibited tumor progression, increased abundance of CD8+ T lymphocytes, and reduced abundance of macrophages in colon tumors. Bone marrow chimeras showed promotion of CAC was primarily mediated by Muc1-expressing hematopoietic cells, and that MUC1 promoted a pro-tumoral immunosuppressive macrophage phenotype within tumors. Mechanistic studies revealed that Muc1 deficiency remarkably reduced interleukin-6 levels in the colonic tissues and tumors that was mainly produced by infiltrating macrophages at day 21, 42, and 85. In bone marrow-derived macrophages, MUC1 promoted responsiveness to chemoattractant and promoted activation into a phenotype with high Il6 and Ido1 expression, secreting factors which inhibited CD8+ T cell proliferation. MUC1 potently drives macrophages to produce interleukin-6, which in turn drives a pro-tumorigenic activation of signal transducer and activator of transcription 3 in colon epithelial tumor and stromal cells, ultimately increasing the occurrence and development of CAC. CONCLUSIONS: Our findings provide cellular and molecular mechanisms for the pro-tumorigenic functions of MUC1 in the inflamed colon. Therapeutic strategies to inhibit MUC1 signal transduction warrant consideration for the prevention or therapy of CAC.


Asunto(s)
Neoplasias Asociadas a Colitis , Interleucina-6 , Activación de Macrófagos , Mucina-1 , Factor de Transcripción STAT3 , Animales , Azoximetano/toxicidad , Carcinogénesis , Factores Quimiotácticos , Colitis/inducido químicamente , Colitis/genética , Colitis/inmunología , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/inmunología , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Sulfato de Dextran/toxicidad , Interleucina-6/genética , Interleucina-6/inmunología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Ratones , Ratones Noqueados , Mucina-1/genética , Mucina-1/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología
8.
BMC Genom Data ; 23(1): 48, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35733095

RESUMEN

BACKGROUND: Colitis-associated colon cancer (CAC) patients have a younger age of onset, more multiple lesions and invasive tumors than sporadic colon cancer patients. Early detection of CAC using endoscopy is challenging, and the incidence of septal colon cancer remains high. Therefore, identifying biomarkers that can predict the tumorigenesis of CAC is in urgent need. RESULTS: A total of 275 DEGs were identified in CAC. IGF1, BMP4, SPP1, APOB, CCND1, CD44, PTGS2, CFTR, BMP2, KLF4, and TLR2 were identified as hub DEGs, which were significantly enriched in the PI3K-Akt pathway, stem cell pluripotency regulation, focal adhesion, Hippo signaling, and AMPK signaling pathways. Sankey diagram showed that the genes of both the PI3K-AKT signaling and focal adhesion pathways were upregulated (e.g., SPP1, CD44, TLR2, CCND1, and IGF1), and upregulated genes were predicted to be regulated by the crucial miRNAs: hsa-mir-16-5p, hsa-mir-1-3p, et al. Hub gene-TFs network revealed FOXC1 as a core transcription factor. In ulcerative colitis (UC) patients, KLF4, CFTR, BMP2, TLR2 showed significantly lower expression in UC-associated cancer. BMP4 and IGF1 showed higher expression in UC-Ca compared to nonneoplastic mucosa. Survival analysis showed that the differential expression of SPP1, CFRT, and KLF4 were associated with poor prognosis in colon cancer. CONCLUSION: Our study provides novel insights into the mechanism underlying the development of CAC. The hub genes and signaling pathways may contribute to the prevention, diagnosis and treatment of CAC.


Asunto(s)
Colitis Ulcerosa , Neoplasias Asociadas a Colitis , Colitis Ulcerosa/complicaciones , Neoplasias Asociadas a Colitis/genética , Biología Computacional , Humanos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt
9.
BMC Gastroenterol ; 22(1): 188, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35428188

RESUMEN

BACKGROUND: Patients with ulcerative colitis are at an increased risk of developing colorectal cancer with a prolonged disease course. Many studies have shown that alterations in the immune microenvironment play a key role in ulcerative colitis-associated colorectal cancer. Additionally, competing endogenous RNAs have important functions in immunoregulation, affecting inflammation and tumorigenesis. However, the complexity and behavioral characteristics of the competing endogenous RNA immunoregulatory network in ulcerative colitis-associated colorectal cancer remain unclear. We constructed a competing endogenous RNA immunoregulatory network to discover and validate a novel competing endogenous RNA immunoregulatory axis to provide insight into ulcerative colitis-associated colorectal cancer progression. METHODS: The competing endogenous RNA immunoregulatory network was constructed using differential expression analysis, weighted gene co-expression network analysis, and immune-related genes. Cmap was used to identify small-molecule drugs with therapeutic potential in ulcerative colitis-associated colorectal cancer. The ulcerative colitis-associated colorectal cancer-related pathways were identified by gene set variation and enrichment analysis. CIBERSORT, single-sample Gene Set Enrichment Analysis, and xCell were used to evaluate the infiltration of immune cells and screen hub immunocytes. The competing endogenous RNA immunoregulatory axis was identified by correlation analysis. RESULTS: We identified 130 hub immune genes and constructed a competing endogenous RNA immunoregulatory network consisting of 56 long non-coding RNAs, four microRNAs, and six targeted hub immune genes. Four small-molecule drugs exerted potential therapeutic effects by reversing the expression of hub immune genes. Pathway analysis showed that the NF-κB pathway was significantly enriched. Neutrophils were identified as hub immunocytes, and IL6ST was significantly positively correlated with the neutrophil count. In addition, NEAT1 may serve as a competing endogenous RNA to sponge miR-1-3p and promote IL6ST expression. CONCLUSIONS: The competing endogenous RNA immunoregulatory axis may regulate neutrophil infiltration, affecting the occurrence of ulcerative colitis-associated colorectal cancer.


Asunto(s)
Colitis Ulcerosa , Neoplasias Asociadas a Colitis , MicroARNs , Colitis Ulcerosa/complicaciones , Colitis Ulcerosa/genética , Neoplasias Asociadas a Colitis/genética , Redes Reguladoras de Genes , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/genética , Microambiente Tumoral/genética
10.
J Immunol ; 208(5): 1280-1291, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35121641

RESUMEN

Inflammatory bowel disease such as chronic colitis promotes colorectal cancer, which is a common cause of cancer mortality worldwide. Hypoxia is a characteristic of inflammation as well as of solid tumors and enforces a gene expression response controlled by hypoxia-inducible factors (HIFs). Once established, solid tumors are immunosuppressive to escape their abatement through immune cells. Although HIF activity is known to 1) promote cancer development and 2) drive tumor immune suppression through the secretion of adenosine, both prolyl hydroxylases and an asparaginyl hydroxylase termed factor-inhibiting HIF (FIH) negatively regulate HIF. Thus, FIH may act as a tumor suppressor in colorectal cancer development. In this study, we examined the role of colon epithelial FIH in a mouse model of colitis-induced colorectal cancer. We recapitulated colitis-associated colorectal cancer development in mice using the azoxymethane/dextran sodium sulfate model in Vil1-Cre/FIH+f/+f and wild-type siblings. Colon samples were analyzed regarding RNA and protein expression and histology. Vil1-Cre/FIH+f/+f mice showed a less severe colitis progress compared with FIH+f/+f animals and a lower number of infiltrating macrophages in the inflamed tissue. RNA sequencing analyses of colon tissue revealed a lower expression of genes associated with the immune response in Vil1-Cre/FIH+f/+f mice. However, tumor occurrence did not significantly differ between Vil1-Cre/FIH+f/+f and wild-type mice. Thus, FIH knockout in colon epithelial cells did not modulate colorectal cancer development but reduced the inflammatory response in chronic colitis.


Asunto(s)
Neoplasias Asociadas a Colitis/patología , Colitis/patología , Neoplasias Colorrectales/patología , Mucosa Intestinal/patología , Oxigenasas de Función Mixta/metabolismo , Adenosina/metabolismo , Animales , Azoximetano/toxicidad , Hipoxia de la Célula/fisiología , Colitis/inducido químicamente , Colitis/genética , Neoplasias Asociadas a Colitis/genética , Colon/patología , Neoplasias Colorrectales/genética , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxigenasas de Función Mixta/genética , Prolil Hidroxilasas/metabolismo , Transducción de Señal/fisiología , Escape del Tumor/inmunología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
11.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35163788

RESUMEN

To better understand the role of sphingolipids in the multifactorial process of inflammatory bowel disease (IBD), we elucidated the role of CerS4 in colitis and colitis-associated cancer (CAC). For this, we utilized the azoxymethane/dextran sodium sulphate (AOM/DSS)-induced colitis model in global CerS4 knockout (CerS4 KO), intestinal epithelial (CerS4 Vil/Cre), or T-cell restricted knockout (CerS4 LCK/Cre) mice. CerS4 KO mice were highly sensitive to the toxic effect of AOM/DSS, leading to a high mortality rate. CerS4 Vil/Cre mice had smaller tumors than WT mice. In contrast, CerS4 LCK/Cre mice frequently suffered from pancolitis and developed more colon tumors. In vitro, CerS4-depleted CD8+ T-cells isolated from the thymi of CerS4 LCK/Cre mice showed impaired proliferation and prolonged cytokine production after stimulation in comparison with T-cells from WT mice. Depletion of CerS4 in human Jurkat T-cells led to a constitutively activated T-cell receptor and NF-κB signaling pathway. In conclusion, the deficiency of CerS4 in T-cells led to an enduring active status of these cells and prevents the resolution of inflammation, leading to a higher tumor burden in the CAC mouse model. In contrast, CerS4 deficiency in epithelial cells resulted in smaller colon tumors and seemed to be beneficial. The higher tumor incidence in CerS4 LCK/Cre mice and the toxic effect of AOM/DSS in CerS4 KO mice exhibited the importance of CerS4 in other tissues and revealed the complexity of general targeting CerS4.


Asunto(s)
Azoximetano/efectos adversos , Neoplasias Asociadas a Colitis/patología , Neoplasias del Colon/patología , Sulfato de Dextran/efectos adversos , Esfingosina N-Aciltransferasa/genética , Linfocitos T/metabolismo , Animales , Neoplasias Asociadas a Colitis/inducido químicamente , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/inmunología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Células Jurkat , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Especificidad de Órganos , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Carga Tumoral
12.
Biochem Biophys Res Commun ; 592: 81-86, 2022 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-35033870

RESUMEN

Studies have shown that the higher prevalence of colorectal cancers among patients with inflammatory bowel disease. Thus, proinflammatory stimulus due to a high-fat diet may impose a higher risk on the development of colorectal cancer. In the present study, we applied a transcriptomic approach to characterize the molecular mechanism(s) by which high-fat feeding aggravates colitis-associated colorectal cancer (CAC). A high-fat diet was supplied in an azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced mouse model for 10 weeks and then the severity of CAC and global gene expression in colon were assessed. Although consumption of high-fat diet did not significantly aggravate CAC, it substantially changed gene expression profile in colon. In AOM/DSS treated mice (AD group) and AD mice fed a high-fat diet (AD + HF group), 34 and 54 DEGs were enriched in 'pathways in cancer', respectively. Notably, high-fat diet upregulated the expression of genes associated with spliceosome and ribosome biogenesis, and downregulated the expression of genes associated with lipid catabolism in mice treated with AOM/DSS. In addition, we identified that DEGs between the AD and AD + HF groups, were enriched in 'metabolic pathways', especially amino acid and nucleotide metabolism. Taken together, this study provides the molecular mechanism in understanding the high-fat diet-mediated CAC development.


Asunto(s)
Neoplasias Asociadas a Colitis/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Dieta Alta en Grasa , Regulación Neoplásica de la Expresión Génica , Animales , Azoximetano , Neoplasias Asociadas a Colitis/patología , Sulfato de Dextran , Perfilación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL
13.
Sci Rep ; 12(1): 381, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013389

RESUMEN

GPR120 (encoded by FFAR4 gene) is a receptor for long chain fatty acids, activated by ω-3 Polyunsaturated Fatty Acids (PUFAs), and expressed in many cell types. Its role in the context of colorectal cancer (CRC) is still puzzling with many controversial evidences. Here, we explored the involvement of epithelial GPR120 in the CRC development. Both in vitro and in vivo experiments were conducted to mimic the conditional deletion of the receptor from gut epithelium. Intestinal permeability and integrity of mucus layer were assessed by using Evans blue dye and immunofluorescence for MUC-2 protein, respectively. Microbiota composition, presence of lipid mediators and short chain fatty acids were analyzed in the stools of conditional GPR120 and wild type (WT) mice. Incidence and grade of tumors were evaluated in all groups of mice before and after colitis-associated cancer. Finally, GPR120 expression was analyzed in 9 human normal tissues, 9 adenomas, and 17 primary adenocarcinomas. Our work for the first time highlights the role of the receptor in the progression of colorectal cancer. We observed that the loss of epithelial GPR120 in the gut results into increased intestinal permeability, microbiota translocation and dysbiosis, which turns into hyperproliferation of epithelial cells, likely through the activation of ß -catenin signaling. Therefore, the loss of GPR120 represents an early event of CRC, but avoid its progression as invasive cancer. these results demonstrate that the epithelial GPR120 receptor is essential to maintain the mucosal barrier integrity and to prevent CRC developing. Therefore, our data pave the way to GPR120 as an useful marker for the phenotypic characterization of CRC lesions and as new potential target for CRC prevention.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Asociadas a Colitis/metabolismo , Colon/metabolismo , Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Animales , Traslocación Bacteriana , Proliferación Celular , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Colon/microbiología , Colon/patología , Progresión de la Enfermedad , Disbiosis , Microbioma Gastrointestinal , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones Noqueados , Permeabilidad , Receptores Acoplados a Proteínas G/genética , Carga Tumoral
14.
Cancer Sci ; 113(2): 565-575, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34807493

RESUMEN

Colitis-associated colorectal cancer (CAC) arises due to prolonged inflammation and has distinct molecular events compared with sporadic colorectal cancer (CRC). Although inflammatory NF-κB signaling was activated by pro-inflammatory cytokines (such as TNFα) in early stages of CAC, Wnt/ß-catenin signaling later appears to function as a key regulator of CAC progression. However, the exact mechanism responsible for the cross-regulation between these 2 pathways remains unclear. Here, we found reciprocal inhibition between NF-κB and Wnt/ß-catenin signaling in CAC samples, and the Dvl2, an adaptor protein of Wnt/ß-catenin signaling, is responsible for NF-κB inhibition. Mechanistically, Dvl2 interacts with the C-terminus of tumor necrosis factor receptor 1 (TNFRI) and mediates TNFRI endocytosis, leading to NF-κB signal inhibition. In addition, increased infiltration of the pro-inflammatory cytokine interleukin-13 (IL-13) is responsible for upregulating Dvl2 expression through STAT6. Targeting STAT6 effectively decreases Dvl2 levels and restrains colony formation of cancer cells. These findings demonstrate a unique role for Dvl2 in TNFRI endocytosis, which facilitates the coordination of NF-κB and Wnt to promote CAC progression.


Asunto(s)
Neoplasias Asociadas a Colitis/metabolismo , Proteínas Dishevelled/metabolismo , FN-kappa B/metabolismo , Proteínas Wnt/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/patología , Citocinas/metabolismo , Progresión de la Enfermedad , Proteínas Dishevelled/genética , Endocitosis , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación , Ratones , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
15.
Inflamm Bowel Dis ; 28(5): 764-774, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34894222

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the third leading cause of cancer in the United States, and inflammatory bowel disease patients have an increased risk of developing CRC due to chronic intestinal inflammation with it being the cause of death in 10% to 15% of inflammatory bowel disease patients. TIPE2 (TNF-alpha-induced protein 8-like 2) is a phospholipid transporter that is highly expressed in immune cells and is an important regulator of immune cell function. METHODS: The azoxymethane/dextran sulfate sodium murine model of colitis-associated colon cancer (CAC) was employed in Tipe2 -/- and wild-type mice, along with colonoid studies, to determine the role of TIPE2 in CAC. RESULTS: Early on, loss of TIPE2 led to significantly less numbers of visible tumors, which was in line with its previously described role in myeloid-derived suppressor cells. However, as time went on, loss of TIPE2 promoted tumor progression, with larger tumors appearing in Tipe2 -/- mice. This was associated with increased interleukin-22/STAT3 phosphorylation signaling. Similar effects were also observed in primary colonoid cultures, together demonstrating that TIPE2 also directly regulated colonocytes in addition to immune cells. CONCLUSIONS: This work demonstrates that TIPE2 has dual effects in CAC. In the colonocytes, it works as a tumor suppressor. However, in the immune system, TIPE2 may promote tumorigenesis through suppressor cells or inhibit it through IL-22 secretion. Going forward, this work suggests that targeting TIPE2 for CRC therapy requires cell- and pathway-specific approaches and serves as a cautionary tale for immunotherapy approaches in general in terms of colon cancer, as intestinal inflammation can both promote and inhibit cancer.


TIPE2 (TNF-alpha-induced protein 8-like 2) regulates immune function. Here, we find that it differentially regulates the initiation and progression of its immunoregulatory properties affect murine colitis-associated colon cancer initiation and progression. Surprisingly, we found that TIPE2 a novel tumor suppressor in enterocytes, a cell compartment it was not previously known to directly regulate.


Asunto(s)
Neoplasias Asociadas a Colitis , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Azoximetano/toxicidad , Transformación Celular Neoplásica/patología , Colitis/inducido químicamente , Colitis/complicaciones , Neoplasias Asociadas a Colitis/genética , Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Inflamación/patología , Enfermedades Inflamatorias del Intestino/complicaciones , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL
16.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166288, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34628032

RESUMEN

GPR65 (TDAG8) is a proton-sensing G protein-coupled receptor predominantly expressed in immune cells. Genome-wide association studies (GWAS) have identified GPR65 gene polymorphisms as an emerging risk factor for the development of inflammatory bowel disease (IBD). Patients with IBD have an elevated risk of developing colorectal cancer when compared to the general population. To study the role of GPR65 in intestinal inflammation and colitis-associated colorectal cancer (CAC), colitis and CAC were induced in GPR65 knockout (KO) and wild-type (WT) mice using dextran sulfate sodium (DSS) and azoxymethane (AOM)/DSS, respectively. Disease severity parameters such as fecal score, colon shortening, histopathology, and mesenteric lymph node enlargement were aggravated in GPR65 KO mice compared to WT mice treated with DSS. Elevated leukocyte infiltration and fibrosis were observed in the inflamed colon of GPR65 KO when compared to WT mice which may represent a cellular mechanism for the observed exacerbation of intestinal inflammation. In line with high expression of GPR65 in infiltrated leukocytes, GPR65 gene expression was increased in inflamed intestinal tissue samples of IBD patients compared to normal intestinal tissues. Moreover, colitis-associated colorectal cancer development was higher in GPR65 KO mice than WT mice when treated with AOM/DSS. Altogether, our data demonstrate that GPR65 suppresses intestinal inflammation and colitis-associated tumor development in murine colitis and CAC models, suggesting potentiation of GPR65 with agonists may have an anti-inflammatory therapeutic effect in IBD and reduce the risk of developing colitis-associated colorectal cancer.


Asunto(s)
Neoplasias Asociadas a Colitis/genética , Colitis/genética , Inflamación/genética , Enfermedades Inflamatorias del Intestino/genética , Animales , Azoximetano/toxicidad , Colitis/inducido químicamente , Colitis/patología , Neoplasias Asociadas a Colitis/inducido químicamente , Neoplasias Asociadas a Colitis/patología , Colon/efectos de los fármacos , Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/patología , Regulación de la Expresión Génica/genética , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Enfermedades Inflamatorias del Intestino/patología , Leucocitos/patología , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Índice de Severidad de la Enfermedad
17.
Cell Commun Signal ; 19(1): 90, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34479599

RESUMEN

Ulcerative colitis (UC) has closely been associated with an increased risk of colorectal cancer. However, the exact mechanisms underlying colitis-associated cancer (CAC) development remain unclear. As a classic pattern-recognition receptor, Toll like receptor (TLR)4 is a canonical receptor for lipopolysaccharide of Gram-negative bacteria (including two CAC-associated pathogens Fusobacterium nucleatum and Salmonella), and functions as a key bridge molecule linking oncogenic infection to colonic inflammatory and malignant processes. Accumulating studies verified the overexpression of TLR4 in colitis and CAC, and the over-expressed TLR4 might promote colitis-associated tumorigenesis via facilitating cell proliferation, protecting malignant cells against apoptosis, accelerating invasion and metastasis, as well as contributing to the creation of tumor-favouring cellular microenvironment. In recent years, considerable attention has been focused on the regulation of TLR4 signaling in the context of colitis-associated tumorigenesis. MicroRNA (miR)-155 and TLR4 exhibited a similar dynamic expression change during CAC development and shared similar CAC-promoting properties. The available data demonstrated an interplay between TLR4 and miR-155 in the context of different disorders or cell lines. miR-155 could augment TLR4 signaling through targeting negative regulators SOCS1 and SHIP1; and TLR4 activation would induce miR-155 expression via transcriptional and post-transcriptional mechanisms. This possible TLR4-miR-155 positive feedback loop might result in the synergistic accelerating effect of TLR4 and miR-155 on CAC development. Video abstract.


Asunto(s)
Carcinogénesis/genética , Neoplasias Asociadas a Colitis/genética , MicroARNs/genética , Receptor Toll-Like 4/genética , Proliferación Celular/genética , Neoplasias Asociadas a Colitis/patología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Transducción de Señal/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética , Microambiente Tumoral/genética
18.
Eur J Pharmacol ; 906: 174270, 2021 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-34171392

RESUMEN

In addition to its pivotal role in purine metabolism, xanthine oxidoreductase (XOR) is one of the key enzymes involved in superoxide radical generation. Oxidative stress has been implicated in the etiology of colorectal cancer, but the contribution of XOR remains unclear. Here we investigated the role of XOR in colitis-associated colorectal cancer (CAC) and the underlying mechanisms. Using clinical samples, we demonstrated that XOR up-regulation was an early event in colonic carcinogenesis. Pharmacological inhibition of XOR effectively delayed the progression of CAC. Moreover, XOR activity positively correlated with tumor necrosis factor-alpha (TNFα) protein levels. Mechanistically, TNFα may activate XOR transcription via activator protein-1 and, thus, promote endogenous hydrogen peroxide generation, resulting in oxidative DNA damage in colon cancer cells. On the other hand, XOR may regulate the TNFα mRNA transcripts by mediating LPS-induced macrophage M1 polarization. Collectively, XOR promotes tumor development by programming the tumor microenvironment and stimulates CAC progression via DNA damage-induced genetic instability.


Asunto(s)
Neoplasias Asociadas a Colitis/inmunología , Daño del ADN/inmunología , Macrófagos/inmunología , Estrés Oxidativo/inmunología , Xantina Deshidrogenasa/metabolismo , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/inmunología , Línea Celular Tumoral , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/patología , Colon/inmunología , Colon/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Macrófagos/metabolismo , Masculino , Activación Transcripcional/inmunología , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Xantina Deshidrogenasa/genética
19.
JCI Insight ; 6(14)2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34138755

RESUMEN

Cancer cells reprogram cellular metabolism to maintain adequate nutrient pools to sustain proliferation. Moreover, autophagy is a regulated mechanism to break down dysfunctional cellular components and recycle cellular nutrients. However, the requirement for autophagy and the integration in cancer cell metabolism is not clear in colon cancer. Here, we show a cell-autonomous dependency of autophagy for cell growth in colorectal cancer. Loss of epithelial autophagy inhibits tumor growth in both sporadic and colitis-associated cancer models. Genetic and pharmacological inhibition of autophagy inhibits cell growth in colon cancer-derived cell lines and patient-derived enteroid models. Importantly, normal colon epithelium and patient-derived normal enteroid growth were not decreased following autophagy inhibition. To couple the role of autophagy to cellular metabolism, a cell culture screen in conjunction with metabolomic analysis was performed. We identified a critical role of autophagy to maintain mitochondrial metabolites for growth. Loss of mitochondrial recycling through inhibition of mitophagy hinders colon cancer cell growth. These findings have revealed a cell-autonomous role of autophagy that plays a critical role in regulating nutrient pools in vivo and in cell models, and it provides therapeutic targets for colon cancer.


Asunto(s)
Neoplasias Asociadas a Colitis/inmunología , Mitocondrias/metabolismo , Mitofagia/inmunología , Nutrientes/deficiencia , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/inmunología , Colitis/patología , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/patología , Colon/citología , Colon/inmunología , Colon/patología , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Masculino , Metabolómica , Ratones , Ratones Transgénicos , Mitocondrias/inmunología , Mitofagia/efectos de los fármacos
20.
BMC Cancer ; 21(1): 607, 2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34034704

RESUMEN

BACKGROUND: The nuclear phosphatase mitogen-activate protein kinase phosphatase-1 (MKP-1) is a key negative regulator of the innate immune response through the regulation of the biosynthesis of proinflammatory cytokines. In colorectal cancer (CRC), which is induced mainly by chronic inflammation, Mkp-1 overexpression was found in addition to disturbances in Mkp-1 functions, which may play a role in cancer development in different types of tumors. However, the potential molecular mechanisms by which Mkp-1 influences CRC development is not clear. Here, we performed global gene expression profiling of Mkp-1 KO mice using RNA sequencing (RNA-seq) to explore the role of Mkp-1 in CRC progression using transcriptome analysis. METHODS: Azoxymethane/dextran sodium sulfate (AOM/DSS) mouse models were used to examine the most dramatic molecular and signaling changes that occur during different phases of CRC development in wild-type mice and Mkp-1 KO mice. Comprehensive bioinformatics analyses were used to elucidate the molecular processes regulated by Mkp-1. Differentially expressed genes (DEGs) were identified and functionally analyzed by Gene Ontology (GO), Kyoto Enrichment of Genes and Genomes (KEGG). Then, protein-protein interaction (PPI) network analysis was conducted using the STRING database and Cytoscape software. RESULTS: Persistent DEGs were different in adenoma and carcinoma stage (238 & 251, respectively) and in WT and MKp-1 KO mice (221& 196, respectively). Mkp-1 KO modulated key molecular processes typically activated in cancer, in particular, cell adhesion, ion transport, extracellular matrix organization, response to drug, response to hypoxia, and response to toxic substance. It was obvious that these pathways are closely associated with cancer development and metastasis. From the PPI network analyses, nine hub genes associated with CRC were identified. CONCLUSION: These findings suggest that MKp-1 and its hub genes may play a critical role in cancer development, prognosis, and determining treatment outcomes. We provide clues to build a potential link between Mkp-1 and colitis-associated tumorigenesis and identify areas requiring further investigation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Asociadas a Colitis/genética , Colitis/complicaciones , Fosfatasa 1 de Especificidad Dual/metabolismo , Regulación Neoplásica de la Expresión Génica , Animales , Azoximetano/administración & dosificación , Azoximetano/toxicidad , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/patología , Biología Computacional , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Fosfatasa 1 de Especificidad Dual/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Pronóstico , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas/genética , RNA-Seq , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA