Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 279
Filtrar
1.
J Immunol Res ; 2024: 8273732, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39359694

RESUMEN

Oxidative stress is crucial in ulcerative colitis (UC) and colitis-associated colorectal cancer (CAC). Intestinal epithelial cells (IECs) are an important component of the intestinal barrier. In previous studies, we have demonstrated that suppressing microRNA-222-3p (miR-222-3p) can protect against oxidative stress in IECs, which ameliorates colonic injuries in UC mice and prevents the conversion of UC to CAC. In this case, we hope to explore whether moxibustion can alleviate UC and CAC by inhibiting miR-222-3p based on mouse models of UC and CAC. After herb-partitioned moxibustion (HPM) intervention, the disease activity index (DAI) and colon macroscopic damage index (CMDI) were significantly reduced in UC mice, and the number and volume of intestinal tumors were decreased considerably in CAC mice. Meanwhile, we found that HPM suppressed miR-222-3p expression and upregulated the mRNA and protein expression of Brahma-related gene 1 (BRG1), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), while inhibiting Kelch-like ECH-associated protein 1 (Keap1) expression in IECs of UC and CAC mice. With changes in reactive oxygen species (ROS), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), and inflammatory cytokines interleukin (IL)-1ß and tumor necrosis factor (TNF)-α), we verified that HPM protects against oxidative stress and inflammation in IECs of UC and CAC mice. The effect of HPM was inhibited in miR-222-3p overexpression mice, further demonstrating that the protective effect of HPM on UC and CAC mice was through inhibiting miR-222-3p. In summary, HPM regulates the BRG1/Nrf2/HO-1 pathway by inhibiting miR-222-3p to attenuate oxidative stress in IECs in UC and CAC.


Asunto(s)
Colitis Ulcerosa , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1 , MicroARNs , Moxibustión , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Transducción de Señal , Factores de Transcripción , Animales , MicroARNs/genética , MicroARNs/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Colitis Ulcerosa/terapia , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/genética , Ratones , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , ADN Helicasas/metabolismo , ADN Helicasas/genética , Neoplasias Asociadas a Colitis/etiología , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Humanos
2.
Cancer Immunol Res ; 12(10): 1311-1319, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39270036

RESUMEN

Neutrophils are the primary myeloid cells that are recruited to inflamed tissues, and they are key players during colitis, being also present within the tumor microenvironment during the initiation and growth of colon cancer. Neutrophils fundamentally serve to protect the host against microorganism invasion, but during cancer development, they can become protumoral and lead to tumor initiation, growth, and eventually, metastasis-hence, playing a dichotomic role for the host. Protumoral neutrophils in cancer patients can be immunosuppressive and serve as markers for disease progression but their characteristics are not fully defined. In this review, we explore the current knowledge on how neutrophils in the gut fluctuate between an inflammatory or immunosuppressive state and how they contribute to tumor development. We describe neutrophils' antitumoral and protumoral effects during inflammatory bowel diseases and highlight their capacity to provoke the advent of inflammation-driven colorectal cancer. We present the functional ambivalence of the neutrophil populations within the colon tumor microenvironment, which can be potentially exploited to establish therapies that will prevent, or even reverse, inflammation-dependent colon cancer incidence in high-risk patients.


Asunto(s)
Neoplasias Asociadas a Colitis , Neutrófilos , Microambiente Tumoral , Humanos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/patología , Microambiente Tumoral/inmunología , Animales , Colitis/inmunología , Colitis/complicaciones
3.
Biomed Pharmacother ; 179: 117292, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39151314

RESUMEN

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.


Asunto(s)
Azoximetano , Berberina , Neoplasias Asociadas a Colitis , Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , Metabolómica , Transcriptoma , Berberina/farmacología , Berberina/uso terapéutico , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Ratones , Azoximetano/toxicidad , Colitis/microbiología , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/patología , Masculino , Ratones Endogámicos C57BL , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/tratamiento farmacológico , Disbiosis , Modelos Animales de Enfermedad
4.
Sci Rep ; 14(1): 18094, 2024 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-39103474

RESUMEN

Ulcerative colitis (UC) is a chronic inflammatory disorder of the colon, and its pathogenesis remains unclear. Polyamine metabolic enzymes play a crucial role in UC. In this study, we aimed to identify pivotal polyamine-related genes (PRGs) and explore the underlying mechanism between PRGs and the disease status and therapeutic response of UC. We analyzed mRNA-sequencing data and clinical information of UC patients from the GEO database and identified NNMT, PTGS2, TRIM22, TGM2, and PPARG as key PRGs associated with active UC using differential expression analysis and weighted gene co-expression network analysis (WCGNA). Receiver operator characteristic curve (ROC) analysis confirmed the accuracy of these key genes in UC and colitis-associated colon cancer (CAC) diagnosis, and we validated their relationship with therapeutic response in external verification sets. Additionally, single-cell analysis revealed that the key PRGs were specific to certain immune cell types, emphasizing the vital role of intestinal tissue stem cells in active UC. The results were validated in vitro and in vivo experiments, including the colitis mice model and CAC mice model. In conclusion, these key PRGs effectively predict the progression of UC patients and could serve as new pharmacological biomarkers for the therapeutic response of UC.


Asunto(s)
Biomarcadores , Colitis Ulcerosa , Poliaminas , Análisis de la Célula Individual , Colitis Ulcerosa/genética , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/terapia , Animales , Humanos , Ratones , Biomarcadores/metabolismo , Análisis de la Célula Individual/métodos , Poliaminas/metabolismo , Modelos Animales de Enfermedad , Proteína Glutamina Gamma Glutamiltransferasa 2 , Masculino , Femenino , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/metabolismo , Transglutaminasas/genética , Transglutaminasas/metabolismo
5.
Theranostics ; 14(11): 4393-4410, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113810

RESUMEN

Rationale: The treatment of ulcerative colitis (UC) presents an ongoing clinical challenge. Emerging research has implicated that the cGAS-STING pathway promotes the progression of UC, but conflicting results have hindered the development of STING as a therapeutic target. In the current study, we aim to comprehensively elucidate the origins, downstream signaling and pathogenic roles of myeloid STING in colitis and colitis-associated carcinoma (CAC). Methods: Tmem173 fl/fl Lyz2-Cre ert2 mice were constructed for inducible myeloid-specific deletion of STING. RNA-sequencing, flow cytometry, and multiplex immunohistochemistry were employed to investigate immune responses in DSS-induced colitis or AOM/DSS-induced carcinogenesis. Colonic organoids, primary bone marrow derived macrophages and dendritic cells, and splenic T cells were used for in vitro studies. Results: We observed that myeloid STING knockout in adult mice inhibited macrophage maturation, reduced DC cell activation, and suppressed pro-inflammatory Th1 and Th17 cells, thereby protecting against both acute and chronic colitis and CAC. However, myeloid STING deletion in neonatal or tumor-present mice exhibited impaired immune tolerance and anti-tumor immunity. Furthermore, we found that TFAM-associated mtDNA released from damaged colonic organoids, rather than bacterial products, activates STING in dendritic cells in an extracellular vesicle-independent yet endocytosis-dependent manner. Both IRF3 and NF-κB are required for STING-mediated expression of IL-12 family cytokines, promoting Th1 and Th17 differentiation and contributing to excessive inflammation in colitis. Conclusions: Detection of the TFAM-mtDNA complex from damaged intestinal epithelium by myeloid STING exacerbates colitis through IL-12 cytokines, providing new evidence to support the development of STING as a therapeutic target for UC and CAC.


Asunto(s)
ADN Mitocondrial , Células Dendríticas , Interleucina-12 , Mucosa Intestinal , Proteínas de la Membrana , Ratones Noqueados , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Interleucina-12/metabolismo , Interleucina-12/genética , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/inmunología , Ratones Endogámicos C57BL , Colitis/patología , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/genética , Transducción de Señal , Colitis Ulcerosa/genética , Colitis Ulcerosa/patología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/inmunología , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/inmunología , Macrófagos/metabolismo , Macrófagos/inmunología , Modelos Animales de Enfermedad , Sulfato de Dextran
6.
Front Biosci (Landmark Ed) ; 29(8): 276, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39206897

RESUMEN

Colitis-associated cancer (CAC) is the most serious complication of inflammatory bowel disease. In recent years, the incidence of CAC has increased worldwide. Oxidative stress (OS) is involved in the development of CAC through oxidative damage to biomolecules or activation of inflammatory signaling pathways. Exosomes are extracellular vesicles that act as messengers to deliver signals and macromolecules to target cells, making them important mediators of intercellular communication and exchange of biologically active molecules between cells. MicroRNAs (miRNAs) carried by exosomes regulate the pro- and anti-inflammatory pathways of OS and play a key role in communication between OS and cancer cells. This review describes the correlation between OS and exosomal miRNAs with the goal of identifying a novel therapeutic method for CAC.


Asunto(s)
Neoplasias Asociadas a Colitis , Exosomas , MicroARNs , Estrés Oxidativo , Humanos , MicroARNs/metabolismo , MicroARNs/genética , Exosomas/metabolismo , Exosomas/genética , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/etiología , Neoplasias Asociadas a Colitis/genética , Animales , Transducción de Señal
7.
J Ethnopharmacol ; 334: 118541, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38992403

RESUMEN

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.


Asunto(s)
Neoplasias Asociadas a Colitis , Medicamentos Herbarios Chinos , Células Epiteliales , Mitofagia , Animales , Mitofagia/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Ratones , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/prevención & control , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Azoximetano/toxicidad , Masculino , Transición Epitelial-Mesenquimal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sulfato de Dextran , Colon/efectos de los fármacos , Colon/patología , Colon/metabolismo , Ratones Endogámicos C57BL , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Colitis/tratamiento farmacológico , Colitis/complicaciones , Colitis/inducido químicamente , Proteínas Quinasas
8.
J Ethnopharmacol ; 334: 118597, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39034016

RESUMEN

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.


Asunto(s)
Aminoácidos , Neoplasias Asociadas a Colitis , Sulfato de Dextran , Medicamentos Herbarios Chinos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Masculino , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Ratones , Azoximetano/toxicidad , Modelos Animales de Enfermedad , Homeostasis/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Ratones Endogámicos C57BL , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/metabolismo , Apoptosis/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Proliferación Celular/efectos de los fármacos
9.
Ecotoxicol Environ Saf ; 282: 116750, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39053045

RESUMEN

Microcystins (MCs) are secondary metabolites generated by cyanobacterial blooms, among which microcystin-LR (MC-LR) stands out as the most widely distributed variant in aquatic environments. However, the effects of MC-LR on the colorectum and its role in promoting colorectal tumor progression remain unclear. Therefore, this study aims to scrutinize the impact of MC-LR on a mice model of colitis-associated colorectal cancer and elucidate the potential underlying molecular mechanisms. In this study, we used AOM/DSS mice and orally administered MC-LR at doses of 40 µg/kg or 200 µg/kg. Exposure to MC-LR increased tumor burden, promoted tumor growth, shortened colon size, and decreased goblet cell numbers and tight junction protein levels in intestinal tissues. Additionally, exposure to MC-LR induced alterations in the structure of gut microbiota in the mouse colon, characterized by an increase in the relative abundance of Escherichia_coli and Shigella_sonnei, and a decline in the relative abundance of Akkermansia_muciniphila. Transcriptomic analysis revealed that MC-LR exposure activated the IL-17 signaling pathway in mouse colorectal tissues and participated in inflammation regulation and immune response. Immunofluorescence results demonstrated an increase in T-helper 17 (Th17) cell levels in mouse colorectal tumors following MC-LR exposure. The results from RT-qPCR revealed that MC-LR induced the upregulation of IL-6, IL-1ß, IL-10, IL-17A, TNF-α, CXCL1, CXCL2, CXCL5 and CCL20. The novelty of this study lies in its comprehensive approach to understanding the mechanisms by which MC-LR may contribute to CRC progression, offering new perspectives and valuable reference points for establishing guidance standards regarding MC-LR in drinking water. Our findings suggest that even at guideline value, MC-LR can have profound effects on susceptible mice, emphasizing the need for a reevaluation of guideline value and a deeper understanding of the role of environmental toxins in cancer progression.


Asunto(s)
Neoplasias Asociadas a Colitis , Disbiosis , Microbioma Gastrointestinal , Toxinas Marinas , Microcistinas , Animales , Microcistinas/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Disbiosis/inducido químicamente , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/inducido químicamente , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/patología , Masculino , Progresión de la Enfermedad , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Colitis/inducido químicamente , Colitis/patología , Colitis/microbiología
10.
Histopathology ; 85(4): 671-685, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39031700

RESUMEN

AIMS: Ulcerative colitis-associated neoplasia (UCAN) is characterised by multifocal tumourigenesis. A wide range of metachronous lesions have been reported to occur after endoscopic treatment of UCAN, which suggests the development of sporadic tumours in lesions treated as UCAN. Therefore, we aimed to evaluate differences of immunohistochemistry (IHC) in features and clinicopathological characteristics of intramucosal lesions in patients with ulcerative colitis (UC). METHODS AND RESULTS: We examined 35 intramucosal lesions resected for carcinoma or dysplasia by total colectomy from patients with UC and 71 sporadic adenomas (SAs) endoscopically resected from patients without UC. UC lesions were divided into the conventional UCAN group, defined as p53 mutant pattern and normal expression of ß-catenin, and the non-conventional UCAN group, defined as the rest. Ki-67 distribution, α-methylacyl-CoA racemase (AMACR) expression and mucin phenotypes were compared using IHC, and clinicopathological characteristics were investigated. Conventional and non-conventional UCAN lesions were located in the left colon and rectum. Relative to the SA lesions, UCAN lesions occurred in much younger patients and exhibited more frequent basal distribution of Ki-67 in tumour crypts. Conventional UCAN lesions tended to be non-polyploid and exhibited a higher frequency of normal AMACR expression than SA lesions. UC lesions were heterogeneous-only two of the eight patients with multiple lesions had lesions (both non-conventional UCAN lesions) exhibiting concordant IHC staining features. CONCLUSIONS: The basal pattern of Ki-67 distribution, normal expression of AMACR and a non-intestinal mucin phenotype were determined as characteristic features suggestive of UCAN. Non-polypoid growth was another a key feature of UCAN.


Asunto(s)
Colitis Ulcerosa , Antígeno Ki-67 , Mucinas , Racemasas y Epimerasas , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Colitis Ulcerosa/patología , Colitis Ulcerosa/complicaciones , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/etiología , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Mucinas/metabolismo , Fenotipo , Racemasas y Epimerasas/metabolismo
11.
Int J Mol Sci ; 25(14)2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39063041

RESUMEN

One of the factors contributing to colorectal cancer (CRC) development is inflammation, which is mostly hypoxia-associated. This study aimed to characterize the morphological and molecular biological features of colon tumors in mice that were tolerant and susceptible to hypoxia based on colitis-associated CRC (CAC). Hypoxia tolerance was assessed through a gasping time evaluation in a decompression chamber. One month later, the animals were experimentally modeled for colitis-associated CRC by intraperitoneal azoxymethane administration and three dextran sulfate sodium consumption cycles. The incidence of tumor development in the distal colon in the susceptible to hypoxia mice was two times higher and all tumors (100%) were represented by adenocarcinomas, while in the tolerant mice, only 14% were adenocarcinomas and 86% were glandular intraepithelial neoplasia. The tumor area assessed on serially stepped sections was statistically significantly higher in the susceptible animals. The number of macrophages, CD3-CD19+, CD3+CD4+, and NK cells in tumors did not differ between animals; however, the number of CD3+CD8+ and vimentin+ cells was higher in the susceptible mice. Changes in the expression of genes regulating the response to hypoxia, inflammation, cell cycle, apoptosis, and epithelial barrier functioning in tumors and the peritumoral area depended on the initial mouse's hypoxia tolerance, which should be taken into account for new CAC diagnostics and treatment approaches development.


Asunto(s)
Apoptosis , Ciclo Celular , Neoplasias Asociadas a Colitis , Inflamación , Animales , Ratones , Apoptosis/genética , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/etiología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ciclo Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/etiología , Regulación Neoplásica de la Expresión Génica , Hipoxia/metabolismo , Hipoxia/genética , Hipoxia/complicaciones , Colitis/genética , Colitis/metabolismo , Colitis/complicaciones , Colitis/inducido químicamente , Colitis/patología , Masculino
12.
Discov Med ; 36(186): 1363-1369, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39054707

RESUMEN

BACKGROUND: Ulcerative colitis is a well-known inflammatory bowel disease. Patients have an increased risk of developing colitis associated carcinoma (CAC). It is important for patient management to be able to distinguish between ulcerative colitis associated carcinoma and sporadic carcinoma (sCRC). However, this distinction is frequently very challenging. It is not readily possible to differentiate this histologically. However, the diagnosis is crucial for the patient's further treatment and follow-up. An attempt was therefore made to develop a diagnostic regime that would enable a reliable distinction between sCRC and CAC. METHODS: We screened 96 patients analyzing more than 850,000 methylation hotspots, to detect distinct epigenetic patterns between both types of carcinomas. Patients with sporadic carcinoma and colitis-associated carcinoma as well as patients with normal colon and patients with confirmed ulcerative colitis without neoplasia were used for the analysis. By extensively filtering the results, methylation sites relevant to distinguish between CAC and sCRC were identified. RESULTS: After the results were filtered, three methylation sites relevant to distinguish between CAC and sCRC were identified. For this purpose, methylation limit values were defined, which favor the samples as CAC or sCRC up to a certain methylation value of the methylation sites. The combination of three methylation sites allows a correct assignment to CAC or sCRC in 94.5% of the cases. CONCLUSION: The results show that these three methylation sites are promising markers in the diagnosis of CAC vs sCRC. Nevertheless, the diagnosis should always be made in conjunction with histomorphological analyses.


Asunto(s)
Colitis Ulcerosa , Neoplasias Asociadas a Colitis , Neoplasias Colorrectales , Metilación de ADN , Humanos , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/genética , Colitis Ulcerosa/complicaciones , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/diagnóstico , Masculino , Femenino , Epigénesis Genética
13.
Gut ; 73(10): 1749-1762, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-38851294

RESUMEN

Mounting evidence underscores the pivotal role of the intestinal barrier and its convoluted network with diet and intestinal microbiome in the pathogenesis of inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CRC). Moreover, the bidirectional association of the intestinal barrier with the liver and brain, known as the gut-brain axis, plays a crucial role in developing complications, including extraintestinal manifestations of IBD and CRC metastasis. Consequently, barrier healing represents a crucial therapeutic target in these inflammatory-dependent disorders, with barrier assessment predicting disease outcomes, response to therapy and extraintestinal manifestations.New advanced technologies are revolutionising our understanding of the barrier paradigm, enabling the accurate assessment of the intestinal barrier and aiding in unravelling the complexity of the gut-brain axis. Cutting-edge endoscopic imaging techniques, such as ultra-high magnification endocytoscopy and probe-based confocal laser endomicroscopy, are new technologies allowing real-time exploration of the 'cellular' intestinal barrier. Additionally, novel advanced spatial imaging technology platforms, including multispectral imaging, upconversion nanoparticles, digital spatial profiling, optical spectroscopy and mass cytometry, enable a deep and comprehensive assessment of the 'molecular' and 'ultrastructural' barrier. In this promising landscape, artificial intelligence plays a pivotal role in standardising and integrating these novel tools, thereby contributing to barrier assessment and prediction of outcomes.Looking ahead, this integrated and comprehensive approach holds the promise of uncovering new therapeutic targets, breaking the therapeutic ceiling in IBD. Novel molecules, dietary interventions and microbiome modulation strategies aim to restore, reinforce, or modulate the gut-brain axis. These advancements have the potential for transformative and personalised approaches to managing IBD.


Asunto(s)
Neoplasias Asociadas a Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Medicina de Precisión , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/patología , Medicina de Precisión/métodos , Microbioma Gastrointestinal/fisiología , Neoplasias Asociadas a Colitis/etiología , Neoplasias Asociadas a Colitis/patología , Mucosa Intestinal/patología , Eje Cerebro-Intestino/fisiología
14.
Toxicol Appl Pharmacol ; 489: 117018, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38945373

RESUMEN

Colitis-associated cancer (CAC) is an aggressive subtype of colorectal cancer that can develop in ulcerative colitis patients and is driven by chronic inflammation and oxidative stress. Current chemotherapy for CAC, based on 5-fluorouracil and oxalipltin, is not fully effective and displays severe side effects, prompting the search for alternative therapies. Dimethylfumarate (DMF), an activator of the nuclear factor erythroid 2-related factor 2 (NRF2), is a potent antioxidant and immunomodelatrory drug used in the treatment of multiple sclerosis and showed a strong anti-inflammatory effect on experimental colitis. Here, we investigated the chemotherapeutic effect of DMF on an experimental model of CAC. Male NMRI mice were given two subcutaneous injections of 1,2 Dimethylhydrazine (DMH), followed by three cycles of dextran sulfate sodium (DSS). Low-dose (DMF30) and high-dose of DMF (DMF100) or oxaliplatin (OXA) were administered from the 8th to 12th week of the experiment, and then the colon tissues were analysed histologically and biochemically. DMH/DSS induced dysplastic aberrant crypt foci (ACF), oxidative stress, and severe colonic inflammation, with a predominance of pro-inflammatory M1 macrophages. As OXA, DMF30 reduced ACF multiplicity and crypt dysplasia, but further restored redox status, and reduced colitis severity by shifting macrophages towards the anti-inflammatory M2 phenotype. Surprisingly, DMF100 exacerbated ACF multiplicity, oxidative stress, and colon inflammation, likely through NRF2 and p53 overexpression in colonic inflammatory cells. DMF had a dual effect on CAC. At low dose, DMF is chemotherapeutic and acts as an antioxidant and immunomodulator, whereas at high dose, DMF is pro-oxidant and exacerbates colitis-associated cancer.


Asunto(s)
Neoplasias Asociadas a Colitis , Sulfato de Dextran , Dimetilfumarato , Macrófagos , Estrés Oxidativo , Animales , Dimetilfumarato/farmacología , Estrés Oxidativo/efectos de los fármacos , Masculino , Ratones , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/prevención & control , Sulfato de Dextran/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Antioxidantes/farmacología , Colon/efectos de los fármacos , Colon/patología , Colon/metabolismo , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/patología , Modelos Animales de Enfermedad , Antiinflamatorios/farmacología , Focos de Criptas Aberrantes/patología , Relación Dosis-Respuesta a Droga , Antineoplásicos/farmacología , Antineoplásicos/toxicidad
15.
Theranostics ; 14(7): 2719-2735, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38773969

RESUMEN

Aim: To elucidate dynamics and functions in colonic macrophage subsets, and their regulation by Bifidobacterium breve (B. breve) and its associated metabolites in the initiation of colitis-associated colorectal cancer (CAC). Methods: Azoxymethane (AOM) and dextran sodium sulfate (DSS) were used to create a CAC model. The tumor-suppressive effect of B. breve and variations of macrophage subsets were evaluated. Intestinal macrophages were ablated to determine their role in the protective effects of B. breve. Efficacious molecules produced by B. breve were identified by non-targeted and targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. The molecular mechanism was further verified in murine bone marrow-derived macrophages (BMDMs), macrophages derived from human peripheral blood mononuclear cells (hPBMCs), and demonstrated in CAC mice. Results: B. breve alleviated colitis symptoms, delayed colonic tumorigenesis, and promoted phenotypic differentiation of immature inflammatory macrophages into mature homeostatic macrophages. On the contrary, the ablation of intestinal macrophages largely annulled the protective effects of B. breve. Microbial analysis of colonic contents revealed the enrichment of probiotics and the depletion of potential pathogens following B. breve supplementation. Moreover, indole-3-lactic acid (ILA) was positively correlated with B. breve in CAC mice and highly enriched in the culture supernatant of B. breve. Also, the addition of ILA directly promoted AKT phosphorylation and restricted the pro-inflammatory response of murine BMDMs and macrophages derived from hPBMCs in vitro. The effects of ILA in murine BMDMs and macrophages derived from hPBMCs were abolished by the aryl hydrocarbon receptor (AhR) antagonist CH-223191 or the AKT inhibitor MK-2206. Furthermore, ILA could protect against tumorigenesis by regulating macrophage differentiation in CAC mice; the AhR antagonist largely abrogated the effects of B. breve and ILA in relieving colitis and tumorigenesis. Conclusion: B. breve-mediated tryptophan metabolism ameliorates the precancerous inflammatory intestinal milieu to inhibit tumorigenesis by directing the differentiation of immature colonic macrophages.


Asunto(s)
Bifidobacterium breve , Diferenciación Celular , Colitis , Indoles , Macrófagos , Probióticos , Animales , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Bifidobacterium breve/metabolismo , Indoles/farmacología , Indoles/metabolismo , Humanos , Colitis/inducido químicamente , Colitis/microbiología , Colitis/complicaciones , Diferenciación Celular/efectos de los fármacos , Probióticos/farmacología , Probióticos/administración & dosificación , Modelos Animales de Enfermedad , Carcinogénesis/efectos de los fármacos , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/metabolismo , Ratones Endogámicos C57BL , Colon/microbiología , Colon/patología , Colon/metabolismo , Sulfato de Dextran , Masculino , Microbioma Gastrointestinal , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Azoximetano
16.
Biomed Pharmacother ; 175: 116580, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38723513

RESUMEN

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.


Asunto(s)
Azoximetano , Neoplasias Asociadas a Colitis , Sulfato de Dextran , Modelos Animales de Enfermedad , Clorhidrato de Erlotinib , Animales , Clorhidrato de Erlotinib/farmacología , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Ratones , Azoximetano/toxicidad , Receptores ErbB/metabolismo , Receptores ErbB/genética , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Ratones Endogámicos C57BL , Masculino , Transducción de Señal/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/patología
17.
Int Immunopharmacol ; 135: 112262, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38805906

RESUMEN

BACKGROUND AND AIM: Huangqin decoction (HQD) is a Chinese medicine used to treat colitis and colorectal cancer (CRC). However, the specific compounds and mechanisms of HQD remain unclear despite its good curative clinical results. Through bioinformatics, network pharmacology, and experiments, this study aims to explore the progressive mechanisms of colitis-associated colorectal cancer (CAC) from ulcerative colitis (UC) while examining the protective effects of HQD and its compounds against this. METHODS: Bioinformatics was utilized to identify the hub genes between UC and CRC, and their clinical predictive significance, function, and expression were validated. Employing network pharmacology in combination with hub genes, key targets of HQD for preventing the development of UC into CAC were identified. Molecular docking and molecular dynamics (MD) were utilized to procure compounds that effectively bind to these targets and their transcription factors (TFs). Finally, the expression and mechanism of key targets were demonstrated in mice with UC or CAC. RESULTS: (1) Joint analysis of UC and CRC gene sets resulted in 14 hub genes, mainly related to extracellular matrix receptor binding, biological processes in the extracellular matrix, focal adhesion and neutrophil migration; (2) Network pharmacology results show HQD has 133 core targets for treating UC and CRC, acting on extracellular matrix, inflammatory bowel disease, chemical carcinogen receptor activation and other pathways; (3) The intersection of hub genes and core targets yielded two key targets, MMP1 and MMP3; (4) STAT3 is a shared TF of MMP1 and MMP3. (5) Molecular docking and MD verified that the dockings between Glabridin and STAT3/MMP1/MMP3 are stable and reliable; (6) In murine vivo experiments verified that Glabridin reduces inflammation, extracellular matrix degradation, and the occurrence of epithelial-mesenchymal transition to prevent UC transforming into CAC by inhibiting the phosphorylation of STAT3 and regulating the activity of MMP1/3.


Asunto(s)
Colitis Ulcerosa , Medicamentos Herbarios Chinos , Isoflavonas , Metaloproteinasa 1 de la Matriz , Metaloproteinasa 3 de la Matriz , Simulación del Acoplamiento Molecular , Fenoles , Factor de Transcripción STAT3 , Animales , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Humanos , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Fenoles/uso terapéutico , Fenoles/farmacología , Ratones , Masculino , Factor de Transcripción STAT3/metabolismo , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/prevención & control , Ratones Endogámicos C57BL , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/prevención & control , Modelos Animales de Enfermedad , Sulfato de Dextran
18.
Cancer Lett ; 593: 216940, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38729554

RESUMEN

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.


Asunto(s)
Ácido 3-Hidroxibutírico , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neovascularización Patológica , Ácido 3-Hidroxibutírico/farmacología , Ácido 3-Hidroxibutírico/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Animales , Ratones , Humanos , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Línea Celular Tumoral , Carcinogénesis/efectos de los fármacos , Masculino , Azoximetano/toxicidad , Colitis/complicaciones , Colitis/metabolismo , Colitis/patología , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Angiogénesis
19.
J Cancer Res Clin Oncol ; 150(5): 243, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38717677

RESUMEN

Colitis-associated colorectal cancer has been a hot topic in public health issues worldwide. Numerous studies have demonstrated the significance of myeloid-derived suppressor cells (MDSCs) in the progression of this ailment, but the specific mechanism of their role in the transformation of inflammation to cancer is unclear, and potential therapies targeting MDSC are also unclear. This paper outlines the possible involvement of MDSC to the development of colitis-associated colorectal cancer. It also explores the immune and other relevant roles played by MDSC, and collates relevant targeted therapies against MDSC. In addition, current targeted therapies for colorectal cancer are analyzed and summarized.


Asunto(s)
Neoplasias Asociadas a Colitis , Neoplasias Colorrectales , Células Supresoras de Origen Mieloide , Humanos , Células Supresoras de Origen Mieloide/inmunología , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/etiología , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Animales , Colitis/complicaciones , Colitis/inmunología
20.
Front Endocrinol (Lausanne) ; 15: 1363748, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38616821

RESUMEN

Corticotropin-releasing factor family peptides (CRF peptides) comprise corticotropin releasing hormone (CRH), urocortin (UCN1), UCN2 and UCN3. CRH is first isolated in the brain and later with UCNs found in many peripheral cells/tissues including the colon. CRH and UCNs function via the two types of receptors, CRF1 and CRF2, with CRH mainly acting on CRF1, UCN1 on both CRF1 &CRF2 and UCN2-3 on CRF2. Compiling evidence shows that CRH participates in inflammation and cancers via both indirect central effects related to stress response and direct peripheral influence. CRH, as a stress-response mediator, plays a significant central role in promoting the development of colitis involving colon motility, immunity and gut flora, while a few anti-colitis results of central CRH are also reported. Moreover, CRH is found to directly influence the motility and immune/inflammatory cells in the colon. Likewise, CRH is believed to be greatly related to tumorigenesis of many kinds of cancers including colon cancer via the central action during chronic stress while the peripheral effects on colitis-associated-colon cancer (CAC) are also proved. We and others observe that CRH/CRF1 plays a significant peripheral role in the development of colitis and CAC in that CRF1 deficiency dramatically suppresses the colon inflammation and CAC. However, up to date, there still exist not many relevant experimental data on this topic, and there seems to be no absolute clearcut between the central and direct peripheral effects of CRH in colitis and colon cancer. Taken together, CRH, as a critical factor in stress and immunity, may participate in colitis and CAC as a centrally active molecule; meanwhile, CRH has direct peripheral effects regulating the development of colitis and CAC, both of which will be summarized in this review.


Asunto(s)
Neoplasias Asociadas a Colitis , Colitis , Neoplasias del Colon , Humanos , Hormona Liberadora de Corticotropina , Colitis/complicaciones , Neoplasias del Colon/etiología , Inflamación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA