Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Carcinog ; 62(6): 771-785, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36988339

RESUMEN

Replication factor C 5 (RFC5) is involved in a variety of biological functions of cancer. However, the expression pattern of RFC5 and the underlying mechanisms in colorectal cancer (CRC) remain elusive. Here, we show that RFC5 is significantly upregulated in CRC tissues and cells. Patients with CRC and increased RFC5 levels have an unfavorable prognosis. RFC5 can promote the proliferation, migration, and invasion of CRC cells and inhibit the apoptosis of CRC cells. Additionally, upstream of RFC5, we constructed the competing endogenous RNA network and confirmed that RFC5 in this network was inhibited by miR-3614-5p by directly targeting its 3'-untranslated regions. We verified that circ_0038985, which is positively correlated with RFC5, directly targeted miR-3614-5p. Overexpression of circ_0038985 promoted CRC cell migration and invasion, and these effects were partially reversed by the reintroduction of miR-3614-5p. Moreover, we found that RFC5 may promote the vascular endothelial growth factor A (VEGFa)/vascular endothelial growth factor receptor 2 (VEGFR2)/extracellular signal-regulated protein kinase (ERK) pathway. The knockdown of RFC5 reduced CRC tumorigenesis in vivo. Collectively, these data demonstrate that the circ_0038985/miR-3614-5p/RFC5 axis plays a critical role in the progression of CRC, and RFC5 may promote CRC progression by affecting the VEGFa/VEGFR2/ERK pathway.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína de Replicación C/genética , Proteína de Replicación C/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Oncogenes
3.
Mol Cell Biochem ; 426(1-2): 17-26, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27878499

RESUMEN

A disintegrin and metalloproteinase 17 (ADAM17) is highly expressed in various tumours and affects tumour progression. In this study, ADAM17 expression in 60 gastric cancer and 20 normal gastric mucosal tissues was assessed using immunohistochemistry. ADAM17 expression was higher in gastric cancer tissues than in normal gastric mucosal tissues (P < 0.0005). A significant relationship was identified between ADAM17 expression and the depth of tumour invasion, metastasis, and carcinoma stage. Furthermore, the effects of ADAM17 knockdown on the proliferation, cell invasion, and apoptosis of human gastric carcinoma cells (SGC-7901) were determined. SGC-7901 cells were transfected with ADAM17-shRNA, and cell proliferation and migration were assessed using CCK-8 and transwell assays, respectively, to evaluate the role of ADAM17 in tumour proliferation and invasion. Furthermore, the EGFR signalling pathway, the cell membrane receptor-bound TNF-α level, and apoptosis were evaluated by western blotting and flow cytometry. The inhibition of cell proliferation and invasion was observed in the ADAM17 knockdown cells, which was associated with modulation of the EGFR signalling pathway. Apoptosis was increased, and TNF-α signalling was attenuated in the ADAM17 knockdown cells. Our study demonstrated that ADAM17 over-expression in gastric cancer tissues was closely associated with tumour proliferation, invasion, and apoptosis.


Asunto(s)
Proteína ADAM17/metabolismo , Receptores ErbB/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína ADAM17/genética , Línea Celular Tumoral , Proliferación Celular/genética , Receptores ErbB/genética , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , ARN Interferente Pequeño/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Factor de Necrosis Tumoral alfa/genética
4.
J Cancer ; 15(4): 916-925, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38230226

RESUMEN

Objective: To establish a nomogram prediction model (based on clinicopathological and radiological features) for the development of metachronous liver metastasis (MLM) in patients with colorectal cancer (CRC). Methods: This retrospective study included patients with CRC who underwent surgery at Changshu No.1 People's Hospital and the Second Affiliated Hospital of Soochow University between January 2016 and December 2018. The clinical, pathological, and radiological features of each patient were investigated. Risk factors for MLM were identified by univariable and multivariable analyses. The predictive nomogram for MLM development was constructed. The predictive performance of the nomogram was estimated by the receiver operating characteristics curve, calibration curve, and decision curve analysis. Results: This study included 161 patients with CRC [median age: 66 (range, 33-87) years]. Fifty-nine developed MLM after a median of 12 (range, 2-52) months after surgery. The multivariable logistic regression analysis showed that age >66 years (OR=3.471, 95% CI: 1.272-9.473, P=0.015), N2 stage (OR=6.534, 95% CI: 1.456-29.317, P=0.014), positive vascular invasion (OR=2.995, 95% CI: 1.132-7.926, P=0.027), positive tumor deposit (OR=4.451, 95% CI: 1.153-17.179, P=0.030), and linear (OR=6.774, 95% CI: 1.306-35.135, P=0.023) and nodal pericolic fat infiltration patterns (OR=8.762, 95% CI: 1.521-50.457, P=0.015) were independently associated with MLM. These five factors were used to create a nomogram. The area under the receiver operating characteristics curve of the nomogram was 0.866 (95% CI: 0.803-0.914), indicating favorable prediction performance. The calibration curve of the nomogram showed a satisfactory agreement between the predicted and actual probabilities. Conclusions: A nomogram prediction model based on five clinicopathological and radiological features might have favorable prediction performance for MLM in patients who underwent surgery for CRC. Hence, the present study proposes a nomogram that can easily be used to predict MLM after CRC surgery based on readily available features.

5.
Crit Rev Oncol Hematol ; 197: 104353, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38615869

RESUMEN

Bortezomib is the first-line standard and most effective chemotherapeutic for multiple myeloma; however, bortezomib-induced peripheral neuropathy (BIPN) severely affects the chemotherapy regimen and has long-term impact on patients under maintenance therapy. The pathogenesis of BIPN is poorly understood, and basic research and development of BIPN management drugs are in early stages. Besides chemotherapy dose reduction and regimen modification, no recommended prevention and treatment approaches are available for BIPN apart from the International Myeloma Working Group guidelines for peripheral neuropathy in myeloma. An in-depth exploration of the pathogenesis of BIPN, development of additional therapeutic approaches, and identification of risk factors are needed. Optimizing effective and standardized BIPN treatment plans and providing more decision-making evidence for clinical diagnosis and treatment of BIPN are necessary. This article reviews the recent advances in BIPN research; provides an overview of clinical features, underlying molecular mechanisms, and therapeutic approaches; and highlights areas for future studies.


Asunto(s)
Antineoplásicos , Bortezomib , Mieloma Múltiple , Enfermedades del Sistema Nervioso Periférico , Humanos , Bortezomib/efectos adversos , Bortezomib/uso terapéutico , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Enfermedades del Sistema Nervioso Periférico/terapia , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico
6.
Sci Rep ; 14(1): 12415, 2024 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816560

RESUMEN

Gastrointestinal stromal tumors (GISTs) are a rare type of tumor that can develop liver metastasis (LIM), significantly impacting the patient's prognosis. This study aimed to predict LIM in GIST patients by constructing machine learning (ML) algorithms to assist clinicians in the decision-making process for treatment. Retrospective analysis was performed using the Surveillance, Epidemiology, and End Results (SEER) database, and cases from 2010 to 2015 were assigned to the developing sets, while cases from 2016 to 2017 were assigned to the testing set. Missing values were addressed using the multiple imputation technique. Four algorithms were utilized to construct the models, comprising traditional logistic regression (LR) and automated machine learning (AutoML) analysis such as gradient boost machine (GBM), deep neural net (DL), and generalized linear model (GLM). We evaluated the models' performance using LR-based metrics, including the area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA), as well as AutoML-based metrics, such as feature importance, SHapley Additive exPlanation (SHAP) Plots, and Local Interpretable Model Agnostic Explanation (LIME). A total of 6207 patients were included in this study, with 2683, 1780, and 1744 patients allocated to the training, validation, and test sets, respectively. Among the different models evaluated, the GBM model demonstrated the highest performance in the training, validation, and test cohorts, with respective AUC values of 0.805, 0.780, and 0.795. Furthermore, the GBM model outperformed other AutoML models in terms of accuracy, achieving 0.747, 0.700, and 0.706 in the training, validation, and test cohorts, respectively. Additionally, the study revealed that tumor size and tumor location were the most significant predictors influencing the AutoML model's ability to accurately predict LIM. The AutoML model utilizing the GBM algorithm for GIST patients can effectively predict the risk of LIM and provide clinicians with a reference for developing individualized treatment plans.


Asunto(s)
Tumores del Estroma Gastrointestinal , Neoplasias Hepáticas , Aprendizaje Automático , Programa de VERF , Humanos , Tumores del Estroma Gastrointestinal/patología , Neoplasias Hepáticas/secundario , Masculino , Femenino , Persona de Mediana Edad , Estudios Retrospectivos , Anciano , Pronóstico , Adulto , Algoritmos , Curva ROC , Neoplasias Gastrointestinales/patología
7.
J Exp Clin Cancer Res ; 43(1): 59, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38413999

RESUMEN

BACKGROUND: Hematological metastasis has been recognized as a crucial factor contributing to the high rates of metastasis and mortality observed in colorectal cancer (CRC). Notably, exosomes derived from cancer cells participate in the formation of CRC pre-metastatic niches; however, the mechanisms underlying their effects are largely unknown. While our preliminary research revealed the role of exosome-derived disintegrin and metalloproteinase 17 (ADAM17) in the early stages of CRC metastasis, the role of exosomal ADAM17 in CRC hematogenous metastasis remains unclear. METHODS: In the present study, we isolated and purified exosomes using ultracentrifugation and identified exosomal proteins through quantitative mass spectrometry. In vitro, co-culture assays were conducted to evaluate the impact of exosomal ADAM17 on the permeability of the blood vessel endothelium. Vascular endothelial cell resistance, the cell index, membrane protein separation, flow cytometry, and immunofluorescence were employed to investigate the mechanisms underlying exosomal ADAM17-induced vascular permeability. Additionally, a mouse model was established to elucidate the role of exosomal ADAM17 in the modulation of blood vessel permeability and pre-metastatic niche formation in vivo. RESULTS: Our clinical data indicated that ADAM17 derived from the circulating exosomes of patients with CRC could serve as a blood-based biomarker for predicting metastasis. The CRC-derived exosomal ADAM17 targeted vascular endothelial cells, thus enhancing vascular permeability by influencing vascular endothelial cadherin cell membrane localization. Moreover, exosomal ADAM17 mediated the formation of a pre-metastatic niche in nude mice by inducing vascular leakage, thereby promoting CRC metastasis. Nonetheless, ADAM17 selective inhibitors effectively reduced CRC metastasis in vivo. CONCLUSIONS: Our results suggest that exosomal ADAM17 plays a pivotal role in the hematogenous metastasis of CRC. Thus, this protein may serve as a valuable blood-based biomarker and potential drug target for CRC metastasis intervention.


Asunto(s)
Neoplasias Colorrectales , Exosomas , MicroARNs , Animales , Ratones , Humanos , MicroARNs/metabolismo , Células Endoteliales/metabolismo , Permeabilidad Capilar , Ratones Desnudos , Biomarcadores/metabolismo , Neoplasias Colorrectales/patología , Exosomas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteína ADAM17/metabolismo
8.
Front Immunol ; 15: 1344637, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38962013

RESUMEN

Disulfidptosis, a regulated form of cell death, has been recently reported in cancers characterized by high SLC7A11 expression, including invasive breast carcinoma, lung adenocarcinoma, and hepatocellular carcinoma. However, its role in colon adenocarcinoma (COAD) has been infrequently discussed. In this study, we developed and validated a prognostic model based on 20 disulfidptosis-related genes (DRGs) using LASSO and Cox regression analyses. The robustness and practicality of this model were assessed via a nomogram. Subsequent correlation and enrichment analysis revealed a relationship between the risk score, several critical cancer-related biological processes, immune cell infiltration, and the expression of oncogenes and cell senescence-related genes. POU4F1, a significant component of our model, might function as an oncogene due to its upregulation in COAD tumors and its positive correlation with oncogene expression. In vitro assays demonstrated that POU4F1 knockdown noticeably decreased cell proliferation and migration but increased cell senescence in COAD cells. We further investigated the regulatory role of the DRG in disulfidptosis by culturing cells in a glucose-deprived medium. In summary, our research revealed and confirmed a DRG-based risk prediction model for COAD patients and verified the role of POU4F1 in promoting cell proliferation, migration, and disulfidptosis.


Asunto(s)
Adenocarcinoma , Biomarcadores de Tumor , Neoplasias Colorrectales , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/diagnóstico , Pronóstico , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Biomarcadores de Tumor/genética , Femenino , Línea Celular Tumoral , Masculino , Proliferación Celular/genética , Perfilación de la Expresión Génica , Transcriptoma , Nomogramas , Factor 3 de Transcripción de Unión a Octámeros/genética , Movimiento Celular/genética
9.
J Clin Med ; 12(6)2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36983320

RESUMEN

BACKGROUND: Immune function is an important indicator for assessing postoperative recovery and long-term survival in patients with malignancy, and laparoscopic surgery is thought to have a less suppressive effect on the immune response than open surgery. This study aimed to investigate this effect in a retrospective clinical study. METHODS: In this retrospective clinical study, we enrolled 63 patients with colorectal cancer in the Department of General Surgery of the First Affiliated Hospital of Soochow University and assessed the changes in their postoperative immune function by measuring CD3+T, CD4+T, CD8+T lymphocytes, and CD4+/CD8+ ratio. RESULTS: Compared with open surgery, laparoscopic colorectal surgery was effective in improving the postoperative decline in immune function. We determined that the number of CD4+, CD8+T lymphocytes, and the CD4+/CD8+ ratio was not significantly reduced in the laparoscopic group. CONCLUSION: Laparoscopic-assisted colorectal resection can reduce the inhibition of immune functions compared with conventional open surgery.

10.
J Gastrointest Oncol ; 13(5): 2458-2471, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36388670

RESUMEN

Background: In recent years, reports regarding stimulator of interferon genes (STING) and the progression of colorectal cancer (CRC) have emerged rapidly, yet their association remains controversial. This research was aimed to provide an insight into the prognostic biomarker and therapeutic target significance of STING in CRC. Methods: CRC Cell lines of HCT116 and SW480, as well as 32 paired CRC specimens were chosen for this study. STING expressions were examined by immunohistochemistry to evaluate the correlation with clinicopathological factors. Data analysis of STING expressions in colon cancer and rectal cancer were performed using The Cancer Genome Atlas (TCGA) database. siRNA was transfected into cell lines for knocking down the expression of STING. Transwell assay was employed to evaluate cell migration and invasiveness. CCK-8 assay was used for assessing the change of cell proliferation. Drug sensitive test was involved to evaluate drug resistance of cell lines. Gene Set Enrichment Analysis (GSEA) was applied for exploring potential downstream mechanism of STING in CRC progression and Western blotting is used for mechanism validation. Results: In the thirty-two paired CRC and adjacent normal tissues, we found a significant up-regulated in STING expression with immunohistochemical staining in cancer tissues compared with adjacent normal tissues (P<0.01), which was correlated with the tumor-node-metastasis (TNM) stage of patients (P=0.028). Meanwhile, GESA enrichment analysis indicated a remarkable change in mTOR signaling following STING regulation. In HCT116 and SW480 cell lines of CRC, When STING was down-regulated, its biological behavior of cell viability, cell invasion and drug sensitivity to 5-fluorouracil were significantly reduced (P<0.05), we also observed the up-regulation of P-AMPK (P<0.05) and down-regulation of p-mTOR (P<0.05). Conclusions: STING expressions was significantly up-regulated in CRC tissues. Expression of STING was correlated with the TNM stage of patients. STING is found to promote cell proliferation, invasion ability and drug resistance mediating AMPK-mTOR signaling in CRC. STING could be a promising target for the sensitization of chemotherapy and inhibits CRC progression.

11.
Comput Math Methods Med ; 2021: 9976440, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34567237

RESUMEN

Texture analysis (TA) techniques derived from T2-weighted imaging (T2WI) and apparent diffusion coefficient (ADC) maps of rectal cancer can both achieve good diagnosis performance. This study was to compare TA from T2WI and ADC maps between different pathological T and N stages to confirm which TA analysis is better in diagnosis performance. 146 patients were enrolled in this study. Tumor TA was performed on every patient's T2WI and ADC maps, respectively; then, skewness, kurtosis, uniformity, entropy, energy, inertia, and correlation were calculated. Our results demonstrated that those significant different parameters derived from T2WI had better diagnostic performance than those from ADC maps in differentiating pT3b-4 and pN1-2 stage tumors. In particular, the energy derived from T2WI was an optimal parameter for diagnostic efficiency. High-resolution T2WI plays a key point in the local stage of rectal cancer; thus, TA derived from T2WI may be a more useful tool to aid radiologists and surgeons in selecting treatment.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/estadística & datos numéricos , Interpretación de Imagen Asistida por Computador/estadística & datos numéricos , Imagen por Resonancia Magnética/estadística & datos numéricos , Neoplasias del Recto/diagnóstico por imagen , Adulto , Anciano , Anciano de 80 o más Años , China , Biología Computacional , Femenino , Humanos , Masculino , Persona de Mediana Edad , Curva ROC , Neoplasias del Recto/patología , Estudios Retrospectivos
12.
Comput Math Methods Med ; 2021: 5095940, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367318

RESUMEN

This study was aimed to determine the diagnostic performance of perfusion-related parameters derived from intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) by comparing them with quantitative parameters from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) based on differentiation grades of rectal cancer. We retrospectively analyzed 98 patients with rectal cancer. Perfusion-related IVIM parameters (D ∗, f, and f·D ∗) and quantitative DCE parameters (K trans, K ep, V e , and V p ) were obtained by plotting the volume-of-interest on in-house software. Furthermore, we compared the difference and diagnostic performance of all well-moderately and poorly differentiated rectal cancer parameters. Finally, we analyzed the correlation between those DCE and IVIM parameters and pathological differentiation grade. The values of f, K trans, and K ep significantly differentiated poor and well-moderate rectal cancers. K trans achieved the highest area under the curve (AUC) value compared to perfusion-related IVIM and DCE parameters. Furthermore, K trans showed a better correlation with pathological differentiation grade than f. The diagnostic efficiency of DCE-MRI was greater than perfusion-related IVIM parameters. The f value derived from perfusion-related IVIM offered a diagnostic performance similar to DCE-MRI for patients with renal insufficiency.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/métodos , Neoplasias del Recto/diagnóstico por imagen , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Biología Computacional , Medios de Contraste , Imagen de Difusión por Resonancia Magnética/estadística & datos numéricos , Femenino , Humanos , Interpretación de Imagen Asistida por Computador/métodos , Interpretación de Imagen Asistida por Computador/estadística & datos numéricos , Imagen por Resonancia Magnética/estadística & datos numéricos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Neoplasias del Recto/irrigación sanguínea , Neoplasias del Recto/patología , Estudios Retrospectivos
13.
J Exp Clin Cancer Res ; 40(1): 331, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686205

RESUMEN

Oxaliplatin (OHP)-induced peripheral neurotoxicity (OIPN) is a severe clinical problem and potentially permanent side effect of cancer treatment. For the management of OIPN, accurate diagnosis and understanding of significant risk factors including genetic vulnerability are essential to improve knowledge regarding the prevalence and incidence of OIPN as well as enhance strategies for the prevention and treatment of OIPN. The molecular mechanisms underlying OIPN are complex, with multi-targets and various cells causing neuropathy. Furthermore, mechanisms of OIPN can reinforce each other, and combination therapies may be required for effective management. However, despite intense investigation in preclinical and clinical studies, no preventive therapies have shown significant clinical efficacy, and the established treatment for painful OIPN is limited. Duloxetine is the only agent currently recommended by the American Society of Clinical Oncology. The present article summarizes the most recent advances in the field of studies on OIPN, the overview of the clinical syndrome, molecular basis, therapy development, and outlook of future drug candidates. Importantly, closer links between clinical pain management teams and oncology will advance the effectiveness of OIPN treatment, and the continued close collaboration between preclinical and clinical research will facilitate the development of novel prevention and treatments for OIPN.


Asunto(s)
Antineoplásicos/efectos adversos , Oxaliplatino/efectos adversos , Enfermedades del Sistema Nervioso Periférico/etiología , Enfermedades del Sistema Nervioso Periférico/terapia , Antineoplásicos/uso terapéutico , Biomarcadores , Terapia Combinada , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Desarrollo de Medicamentos , Predisposición Genética a la Enfermedad , Humanos , Incidencia , Terapia Molecular Dirigida , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Oxaliplatino/uso terapéutico , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Enfermedades del Sistema Nervioso Periférico/epidemiología , Fenotipo , Polimorfismo Genético , Prevalencia , Factores de Riesgo , Síndrome
14.
Front Pharmacol ; 12: 674807, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33959032

RESUMEN

Colorectal cancer is a multifactorial disease involving genetic, environmental, and lifestyle risk factors. Intestinal microbiota plays an important role in the occurrence and development of colorectal cancer. Studies have shown that the behavior of intestinal microbiota can lead to pathological changes in the host intestine, which can be divided into epigenetic changes and carcinogenic changes at the gene level, and ultimately promote the formation and development of colorectal cancer. Intestinal microbiota is mainly distributed in the intestinal epithelium, which is composed of a large number of microorganisms interacting with the host intestinal cells. It can affect the immune-inflammation and metabolism of the gastrointestinal tract, and may be used as a biomarker for disease diagnosis. Regulation of gut microbiota is a promising strategy for the prevention and treatment of colorectal cancer. This article reviews the role of intestinal microbiota in the development of colorectal cancer, including the related mechanisms of intestinal microbiota promoting colorectal cancer, the use of intestinal microbiota in the diagnosis of colorectal cancer, and the regulation of intestinal microbiota in the prevention or treatment of colorectal cancer.

15.
Front Pharmacol ; 12: 734351, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34650435

RESUMEN

Exosomes derived from cancer cells are deemed important drivers of pre-metastatic niche formation at distant organs, but the underlying mechanisms of their effects remain largely unknow. Although the role of ADAM17 in cancer cells has been well studied, the secreted ADAM17 effects transported via exosomes are less understood. Herein, we show that the level of exosome-derived ADAM17 is elevated in the serum of patients with metastatic colorectal cancer as well as in metastatic colorectal cancer cells. Furthermore, exosomal ADAM17 was shown to promote the migratory ability of colorectal cancer cells by cleaving the E-cadherin junction. Moreover, exosomal ADAM17 overexpression as well as RNA interference results highlighted its function as a tumor metastasis-promoting factor in colorectal cancer in vitro and in vivo. Taken together, our current work suggests that exosomal ADAM17 is involved in pre-metastatic niche formation and may be utilized as a blood-based biomarker of colorectal cancer metastasis.

16.
Redox Biol ; 36: 101677, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32823168

RESUMEN

Management of oxaliplatin-induced peripheral neuropathy (OIPN) has proven challenging owing to the concern that any OIPN-preventing agents may also decrease the efficacy of the chemotherapeutic agent and fail to reverse established neuronal damage. Nevertheless, targeting redox signaling pathways constitutes a promising therapy in OIPN and we have previously demonstrated the protective role of nuclear factor erythroid-2 related factor 2 (NRF2) in this disorder. Here, we investigated the protective properties of formononetin (FN), a clinical preparation extract, in OIPN. RNA interference experiments revealed that FN protects against OIPN directly through activation of the NRF2 pathway. Further expression profile sequencing showed that FN exerts its protective effect via the NRF2 downstream-oxaliplatin metabolism enzyme, GSTP1. We also demonstrated that FN does not influence the chemotherapeutic function of oxaliplatin, as NRF2 exhibits a different drug metabolic enzyme activation state downstream in colorectal cell lines than that in neurons. Following synthesis of Bio-FN to screen the target binding proteins, we found that FN selectively binds to His129 and Lys131 in the BTB domain of KEAP1. In vivo experiments revealed that FN-induced activation of the NRF2 signaling pathway alleviated the nociceptive sensations in mice. Our findings highlight a new binding mechanism between KEAP1 and isoflavones for activation of the NRF2 system and suggest that pharmacological or therapeutic activation of the NRF2-GSTP1 axis may serve as an effective strategy to prevent or attenuate the progression of OIPN.


Asunto(s)
Isoflavonas , Enfermedades del Sistema Nervioso Periférico , Animales , Isoflavonas/farmacología , Proteína 1 Asociada A ECH Tipo Kelch/genética , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Oxaliplatino/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/genética
17.
Redox Biol ; 30: 101416, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31927409

RESUMEN

GSTpi is a Phase II metabolic enzyme which is originally considered as an important facilitator of cellular detoxification. Here, we found that GSTpi stabilized VE-cadherin in endothelial cell membrane through inhibiting VE-cadherin phosphorylation and VE-cadherin/catenin complex dissociation, and consequently maintained endothelial barrier function. Our findings demonstrated a novel mechanism that GSTpi inhibited VE-cadherin phosphorylation through suppressing the activation of Src/VE-cadherin pathway. Mass spectrometry analysis and molecular docking showed that GSTpi enhanced Src S-glutathionylation at Cys185, Cys245, and Cys400 of Src. More important, we found that GSTpi promoted S-glutathionylation of Src was essential for GSTpi to inhibit Src phosphorylation and activation. Furthermore, in vivo experiments indicated that AAV-GSTpi exerted the protective effect on pulmonary vessel permeability in the animal model of acute lung injury. This study revealed a novel regulatory effect of GSTpi on vascular endothelial barrier function and the importance of S-glutathionylation of Src induced by GSTpi in the activation of Src/VE-cadherin pathway.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Antígenos CD/química , Cadherinas/química , Gutatión-S-Transferasa pi/metabolismo , Familia-src Quinasas/metabolismo , Lesión Pulmonar Aguda/etiología , Animales , Permeabilidad Capilar , Línea Celular , Modelos Animales de Enfermedad , Gutatión-S-Transferasa pi/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Espectrometría de Masas , Ratones , Simulación del Acoplamiento Molecular , Fosforilación , Estabilidad Proteica , Familia-src Quinasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA