Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46.569
Filtrar
1.
Int J Oncol ; 64(1)2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37975227

RESUMO

A variety of malignancies preferentially meet energy demands through the glycolytic pathway. Hypoxia­induced cancer cell adaptations are essential for tumor development. However, in cancerous glycolysis, the functional importance and underlying molecular mechanism of prolyl hydroxylase domain protein 2 (PHD2) have not been fully elucidated. Gain­ and loss­of­function assays were conducted to evaluate PHD2 functions in colon cancer cells. Glucose uptake, lactate production and intracellular adenosine­5'­triphosphate/adenosine diphosphate ratio were measured to determine glycolytic activities. Protein and gene expression levels were measured by western blot analysis and reverse transcription­quantitative PCR, respectively. The human colon cancer xenograft model was used to confirm the role of PHD2 in tumor progression in vivo. Functionally, the data demonstrated that PHD2 knockdown leads to increased glycolysis, while PHD2 overexpression resulted in suppressed glycolysis in colorectal cancer cells. In addition, the glycolytic activity was enhanced without PHD2 and normalized after PHD2 reconstitution. PHD2 was shown to inhibit colorectal tumor growth, suppress cancer cell proliferation and improve tumor­bearing mice survival in vivo. Mechanically, it was found that PHD2 inhibits the expression of critical glycolytic enzymes (glucose transporter 1, hexokinase 2 and phosphoinositide­dependent protein kinase 1). In addition, PHD2 inhibited Ikkß­mediated NF­κB activation in a hypoxia­inducible factor­1α­independent manner. In conclusion, the data demonstrated that PHD2/Ikkß/NF­κB signaling has critical roles in regulating glycolysis and suggests that PHD2 potentially suppresses colorectal cancer.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Glicólise , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Quinase I-kappa B/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolil Hidroxilases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166906, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37802156

RESUMO

Sphingolipids are important for the physicochemical properties of cellular membranes and deregulated in tumors. In human colon cancer tissue ceramide synthase (CerS) 4 and CerS5 are reduced which correlates with a reduced survival probability of late-stage colon cancer patients. Both enzymes are reduced after hypoxia in advanced colorectal cancer (CRC) cells (HCT-116, SW620) but not in non-metastatic CRC cells (SW480, Caco-2). Downregulation of CerS4 or CerS5 in advanced CRC cells enhanced tumor formation in nude mice and organoid growth in vitro. This was accompanied by an enhanced proliferation rate and metabolic changes leading to a shift towards the Warburg effect. In contrast, CerS4 or CerS5 depletion in Caco-2 cells reduced tumor growth in vivo. Lipidomic and proteomic analysis of membrane fractions revealed significant changes in tumor-promoting cellular pathways and cellular transporters. This study identifies CerS4 and CerS5 as prognostic markers for advanced colon cancer patients and provides a comprehensive overview about the associated cellular metabolic changes. We propose that the expression level of CerS4 and CerS5 in colon tumors could serve as a basis for decision-making for personalized treatment of advanced colon cancer patients. Trial registration: The study was accredited by the study board of the Deutsche Krebsgesellschaft (Registration No: St-D203, 2017/06/30, retrospectively registered).


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Animais , Camundongos , Humanos , Esfingolipídeos/metabolismo , Células CACO-2 , Camundongos Nus , Prognóstico , Proteômica , Neoplasias do Colo/metabolismo , Neoplasias Colorretais/patologia , Hipóxia
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166880, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37696461

RESUMO

Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the world's fourth most deadly cancer. CRC, as a genetic susceptible disease, faces significant challenges in optimizing prognosis through optimal drug treatment modalities. In recent decades, the development of innovative small-molecule drugs is expected to provide targeted interventions that accurately address the different molecular characteristics of CRC. Although the clinical application of single-target drugs is limited by the heterogeneity and high metastasis of CRC, novel small-molecule drug treatment strategies such as dual/multiple-target drugs, drug repurposing, and combination therapies can help overcome these challenges and provide new insights for improving CRC treatment. In this review, we focus on the current status of a range of small molecule drugs that are being considered for CRC therapy, including single-target drugs, dual/multiple-target drugs, drug repurposing and combination strategies, which will pave the way for targeting CRC vulnerabilities with small-molecule drugs in future personalized treatment.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Sistemas de Liberação de Medicamentos , Terapia Combinada
4.
Gene ; 891: 147828, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37748628

RESUMO

Long non-coding RNAs have been implicated in biological processes, and are dysregulated in types of cancer. Studies have shown that PCAT19 and CKMT2-AS1 lncRNAs promote tumor growth, invasion, and metastasis by regulating signaling pathways and modulating the gene expression. This study investigated the expression levels of lncRNAs PCAT19 and CKMT2-AS1 in colorectal tumors and normal tissues. First, Using GEPIA2 database, we compared the expression level of target lncRNAs between primary colon adenocarcinoma tumor and normal tissues. Then, the expression levels of lncRNAs PCAT19 and CKMT2-AS1 were detected in 35 colorectal tumors and paired adjacent tissues using qRT-PCR. A receiver operating characteristic (ROC) curve was used to evaluate the value of these lncRNAs as biomarkers. Statistical analysis based on GEPIA2 showed that both lncRNAs PCAT19 and CKMT2-AS1 were significantly decreased in colon adenocarcinoma compared to the normal group (P < 0.001). Experimental analysis showed that the expression level of lncRNA PCAT19 was decreased in colorectal tumors (p < 0.0001) compared to normal tissues. While the expression level of lncRNA CKMT2-AS1 did not change in tumor tissues, it decreased in non-metastatic tumors compared to normal tissues (p = 0.04). The significantly downregulation of lncRNA PCAT19 expression in both metastatic and non-metastatic colorectal tumors compared to normal tissue suggests that PCAT19 may play a role in the carcinogenesis and progression of colorectal cancer and may provide potential therapeutic targets for colorectal cancer. Based on the results of ROC curve analysis, lncRNA PCAT19 may also serves as a novel potential good biomarker in diagnosis colorectal cancer (AUC = 0.94, p < 0.0001) but no significant was found for lncRNA CKMT2-AS1 (p > 0.05).


Assuntos
Adenocarcinoma , Neoplasias do Colo , Neoplasias Colorretais , RNA Longo não Codificante , Humanos , Neoplasias do Colo/genética , RNA Longo não Codificante/genética , Adenocarcinoma/genética , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Biomarcadores , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética , Linhagem Celular Tumoral , Creatina Quinase Mitocondrial/genética
5.
Surg Clin North Am ; 104(1): 227-242, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37953038

RESUMO

Colorectal cancer is one of the most common malignancies worldwide. Approximately half of the patients diagnosed will develop colorectal liver metastases (CRLM). Liver resection has a 50% 5-year survival; however, only a fourth of cases are resectable. Unresectable CRLM has poor prognosis despite improved systemic and local ablative treatments. Liver transplantation (LT) has demonstrated a survival benefit in initial prospective clinical trials. Current use of LT for CRLM is limited to several randomized trials and high-performing centers. Improving patient selection criteria and perioperative management, LT will likely become an important part of the multidisciplinary approach to managing the metastatic disease.


Assuntos
Neoplasias Colorretais , Neoplasias Hepáticas , Transplante de Fígado , Humanos , Neoplasias Colorretais/patologia , Estudos Prospectivos , Neoplasias Hepáticas/cirurgia , Neoplasias Hepáticas/secundário , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Hepatectomia
6.
Int J Mol Med ; 53(1)2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37937666

RESUMO

Tumor­associated macrophages (TAMs) are pivotal components in colorectal cancer (CRC) progression, markedly influencing the tumor microenvironment through their polarization into the pro­inflammatory M1 or pro­tumorigenic M2 phenotypes. Recent studies have highlighted that the Grb2­associated binder 2 (Gab2) is a critical gene involved in the development of various types of tumor, including CRC. However, the precise role of Gab2 in mediating TAM polarization remains incompletely elucidated. In the present study, it was discovered that Gab2 was highly expressed within CRC tissue TAMs, and was associated with a poor prognosis of patients with CRC. Functionally, it was identified that the tumor­conditioned medium (TCM) induced Gab2 expression, facilitating the TAMs towards an M2­like phenotype polarization. Of note, the suppression of Gab2 expression using shRNA markedly inhibited the TCM­induced expression of M2­associated molecules, without affecting M1­type markers. Furthermore, the xenotransplantation model demonstrated that Gab2 deficiency in TAMs inhibited tumor growth in the mouse model of CRC. Mechanistically, Gab2 induced the M2 polarization of TAMs by regulating the AKT and ERK signaling pathways, promoting CRC growth and metastasis. In summary, the present study study elucidates that decreasing Gab2 expression hinders the transition of TAMs towards the M2 phenotype, thereby suppressing the growth of CRC. The exploration of the regulatory mechanisms of Gab2 in TAM polarization may enhance the current understanding of the core molecular pathways of CRC development and may thus provide a foundation for the development of novel immunotherapeutic strategies targeted against TAMs.


Assuntos
Neoplasias Colorretais , Macrófagos Associados a Tumor , Animais , Camundongos , Humanos , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/patologia , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Macrófagos/metabolismo , Transdução de Sinais , Microambiente Tumoral , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
7.
J Colloid Interface Sci ; 654(Pt A): 612-625, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37862809

RESUMO

Reprogramming immunosuppressive M2 macrophages into M1 macrophages in tumor site provides a new strategy for the immunotherapy of colorectal cancer. In this study, M1 macrophage-derived exosome nanoprobe (M1UC) with Ce6-loaded upconversion material is designed to enhance the photodynamic performance of Ce6 while reprogramming M2 macrophages at tumor site and producing NO gas for three-mode synergistic therapy. Under the excitation of near-infrared light at 808 nm, the probe can generate 660 nm up-conversion fluorescence, which enables the photosensitizer Ce6 to produce ROS efficiently. In addition, the probe leads the production of NO by nitric oxide synthase on exosomes. Confocal laser and flow cytometry results show that M1UC probe reprograms M2 macrophages into M1 macrophages with an efficiency of 95.12%. The cell experiments show that the apoptosis rate of the three-mode synergistic therapy group is 78.8%, and the therapeutic effect is significantly higher than those of the other single treatment groups. In vivo experiments results show that M1UC probes maximally gather at the tumor site after 12 h of intravenous injection in orthotopic colorectal cancer mice. After 808 nm laser irradiation, the survival rate of mice is 100% and the recurrence rate was 0 within 60 d, and the therapeutic effect is significantly higher than those of other single treatment groups, which is also confirmed by immunohistochemistry. This M1 macrophage-derived exosome nanoplatform which is based on the three modes of immunotherapy, gas therapy and photodynamic therapy, provides a new design idea for the diagnosis and treatment of deep tumors.


Assuntos
Neoplasias Colorretais , Exossomos , Fotoquimioterapia , Camundongos , Animais , Linhagem Celular Tumoral , Fotoquimioterapia/métodos , Macrófagos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia
8.
Gene ; 892: 147887, 2024 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-37813207

RESUMO

Colorectal cancer (CRC) continues to be a major contributor to cancer-related mortality. Connexin 40 (CX40) is one of the major gap junction proteins with the capacity in regulating cell-to-cell communication and angiogenesis. This study investigates its role in angiogenesis in CRC and explores the regulatory mechanism. Aberrant high CX40 expression was detected in tumor tissues, which was associated with a poor prognosis in CRC patients. Elevated CX40 expression was detected in CRC cell lines as well. Conditioned medium of SW620 and HT29 cell lines was used to induce angiogenesis of human umbilical vein endothelial cells (HUVECs). CX40 knockdown in CRC cells reduced angiogenesis and mobility of HUVECs and blocked CRC cell proliferation, mobility, and survival. Following bioinformatics predictions, we validated by chromatin immunoprecipitation and luciferase assays that nuclear receptor subfamily 3 group C member 1 (NR3C1), which was poorly expressed in CRC samples, suppressed CX40 transcription. The poor NR3C1 expression was attributive to DNA hypermethylation induced by DNA methyltransferase 1 (DNMT1). Restoration of NR3C1 suppressed the pro-angiogenic effect, proliferation and survival, and tumorigenic activity of CRC cells, which were, however, rescued by CX40 upregulation. Collectively, this study demonstrates that transcription activation of CX40 upon DNMT1-mediated NR3C1 DNA methylation potentiates angiogenesis in CRC.


Assuntos
Neoplasias Colorretais , Humanos , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Colorretais/patologia , Conexinas/genética , DNA/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Células HT29 , Células Endoteliais da Veia Umbilical Humana/metabolismo , Receptores de Glucocorticoides/genética , Ativação Transcricional
9.
Rev. esp. med. nucl. imagen mol. (Ed. impr.) ; 42(6): 359-366, nov.- dec. 2023. tab
Artigo em Espanhol | IBECS | ID: ibc-227099

RESUMO

Objetivo El objetivo de nuestro estudio fue determinar el valor de la tomografía por emisión de positrones/tomografía computarizada con 18F-fluorodesoxiglucosa (PET/TC con18F-FDG) basada en la radiómica del tumor primario y peritumoral en la predicción de depósitos tumorales (TD), crecimiento tumoral (TB) e invasión venosa extramural (EMVI) del cáncer colorrectal (CCR). Métodos Nuestro estudio retrospectivo incluyó a 77 pacientes con CCR a los que se les realizó un 18F-FDG PET/TC preoperatoria entre junio de 2020 y febrero de 2022. Se extrajeron un total de 131 características radiómicas del tumor primario y áreas peritumorales en imágenes de fusión PET/TC. Se investigó la relación entre TD, TB, EMVI y estadio T en el estudio patológico postoperatoria de los tumores y las características radiómicas. Las características con un coeficiente de correlación (CC) inferior a 0,8 se analizaron con regresión logística. El rendimiento del modelo se evaluó mediante el área bajo la curva (AUC) obtenida del análisis de las características operativas del receptor. Resultados Se desarrolló un modelo a partir de datos de radiómica peritumoral y tumor primario para predecir el estadio T (AUC 0,931), y también se construyó un modelo predictivo a partir de radiómica derivada del tumor primario para predecir EMVI (AUC 0,739). Los datos radiómicos derivados del tumor primario se obtuvieron como factor pronóstico predictivo del DT y se encontró que una característica peritumoral era un factor pronóstico en la predicción de TB. Conclusiones La radiómica intratumoral y peritumoral derivada de la PET con18F-FDG es útil para la predicción preoperatoria no invasiva de propiedades patológicas que tienen implicaciones importantes en el manejo del CCR (AU)


Objective We aimed to determine the value of 18F-fluorodeoxyglucose positron emission tomogra-phy/computed tomography (18F-FDG PET/CT) based primary tumoral and peritumoral radiomics in the prediction of tumor deposits (TDs), tumor budding (TB) and extramural venous invasion (EMVI) of colorectal cancer (CRC). Methods Our retrospective study included 77 CRC patients who had preoperative18F-FDG PET/CT between June 2020 and February 2022. A total of 131 radiomic features were extracted from primary tumors and peritumoral areas on PET/CT fusion images. The relationship between TDs, TB, EMVI and T stage in the postoperative pathology of the tumors and radiomic features was investigated. Features with a correlation coefficient (CC) less than 0.8 were analyzed by logistic regression. The area under curve (AUC) obtained from the receiver operating characteristic analysis was used to measure the model performance. Results A model was developed from primary tumoral and peritumoral radiomics data to predict T stage (AUC 0.931), and also a predictive model was constructed from primary tumor derived radiomics to predict EMVI (AUC 0.739). Radiomic data derived from the primary tumor was obtained as a predictive prognostic factor in predicting TDs and a peritumoral feature was found to be a prognostic factor in predicting TB. Conclusions Intratumoral and peritumoral radiomics derived from18F-FDG PET/CT are useful for non-invasive early prediction of pathological features that have important implications in the management of CRC (AU)


Assuntos
Humanos , Masculino , Feminino , Adolescente , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Fluordesoxiglucose F18 , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/patologia , Estadiamento de Neoplasias , Estudos Retrospectivos , Biópsia
10.
Clin. transl. oncol. (Print) ; 25(12): 3378-3394, dec. 2023.
Artigo em Inglês | IBECS | ID: ibc-227284

RESUMO

Peritoneal metastases (PM) occur when cancer cells spread inside the abdominal cavity and entail an advanced stage of colorectal cancer (CRC). Prognosis, which is poor, correlates highly with tumour burden, as measured by the peritoneal cancer index (PCI). Cytoreductive surgery (CRS) in specialized centres should be offered especially to patients with a low to moderate PCI when complete resection is expected. The presence of resectable metastatic disease in other organs is not a contraindication in well-selected patients. Although several retrospective and small prospective studies have suggested a survival benefit of adding hyperthermic intraperitoneal chemotherapy (HIPEC) to CRS, the recently published phase III studies PRODIGE-7 in CRC patients with PM, and COLOPEC and PROPHYLOCHIP in resected CRC with high-risk of PM, failed to show any survival advantage of this strategy using oxaliplatin in a 30-min perfusion. Final results from ongoing randomized phase III trials testing CRS plus HIPEC based on mitomycin C (MMC) are awaited with interest. In this article, a group of experts selected by the Spanish Group for the Treatment of Digestive Tumours (TTD) and the Spanish Group of Peritoneal Oncologic Surgery (GECOP), which is part of the Spanish Society of Surgical Oncology (SEOQ), reviewed the role of HIPEC plus CRS in CRC patients with PM. As a result, a series of recommendations to optimize the management of these patients is proposed (AU)


Assuntos
Humanos , Neoplasias Colorretais/patologia , Hipertermia Induzida/métodos , Neoplasias Peritoneais/secundário , Metástase Neoplásica , Modalidades de Fisioterapia , Estudos Retrospectivos , Estudos Prospectivos , Taxa de Sobrevida
11.
Clin. transl. oncol. (Print) ; 25(11): 3262-3276, 11 nov. 2023. graf
Artigo em Inglês | IBECS | ID: ibc-226849

RESUMO

Background Cancer stem cells (CSCs) have unique biological characteristics, including tumorigenicity, immortality, and chemoresistance. Colorectal CSCs have been identified and isolated from colorectal cancers by various methods. AKAP12, a scaffolding protein, is considered to act as a potential suppressor in colorectal cancer, but its role in CSCs remains unknown. In this study, we investigated the function of AKAP12 in Colorectal CSCs. Methods Herein, Colorectal CSCs were enriched by cell culture with a serum-free medium. CSC-associated characteristics were evaluated by Flow cytometry assay and qPCR. AKAP12 gene expression was regulated by lentiviral transfection assay. The tumorigenicity of AKAP12 in vivo by constructing a tumor xenograft model. The related pathways were explored by qPCR and Western blot. Results The depletion of AKAP12 reduced colony formation, sphere formation, and expression of stem cell markers in colorectal cancer cells, while its knockdown decreased the volume and weight of tumor xenografts in vivo. AKAP12 expression levels also affected the expression of stemness markers associated with STAT3, potentially via regulating the expression of protein kinase C. Conclusion This study suggests Colorectal CSCs overexpress AKAP12 and maintain stem cell characteristics through the AKAP12/PKC/STAT3 pathway. AKAP12 may be an important therapeutic target for blocking the development of colorectal cancer in the field of cancer stem cells (AU)


Assuntos
Humanos , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Fenótipo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Células-Tronco Neoplásicas/patologia , Fator de Transcrição STAT3
12.
Cell Death Dis ; 14(11): 750, 2023 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-37973787

RESUMO

Our previous study demonstrated that tumor-suppressor circular RNAs (circRNAs) can be specifically secreted outside of colorectal cancer (CRC) cells within exosomes to maintain tumor cell fitness. However, whether tumor-driving circRNAs can be specifically retained in cells to facilitate tumor progression remains unknown. In this study, circRNA-seq showed that circSKA3 was significantly upregulated in CRC tissues but downregulated in serum samples from CRC patients. In addition, circSKA3 promoted CRC progression in vitro and in vivo and was retained in CRC cells via a specific cellmotif element. Interestingly, the cellmotif element was also the site of interaction of circSKA3 with SLUG, which inhibited SLUG ubiquitination degradation and promoted CRC epithelial-mesenchymal transition (EMT). Moreover, FUS was identified as a key circularization regulator of circSKA3 that bound to the key element. Finally, we designed and synthesized specific antisense oligonucleotides (ASOs) targeting circularization and cellmotif elements, which repressed circSKA3 expression, abolished the SLUG-circSKA3 interaction, and further inhibited CRC EMT and metastasis in vitro and in vivo.


Assuntos
Neoplasias Colorretais , RNA Circular , Humanos , Neoplasias Colorretais/patologia , Genes Supressores de Tumor , RNA Circular/genética , RNA Circular/metabolismo , Ubiquitinação
13.
J Exp Clin Cancer Res ; 42(1): 306, 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37986103

RESUMO

BACKGROUND: Liver metastasis is one of the most important reasons for high mortality of colorectal cancer (CRC). Growing evidence illustrates that lncRNAs play a critical role in CRC liver metastasis. Here we described a novel function and mechanisms of BACE1-AS promoting CRC liver metastasis. METHODS: qRT-PCR and in situ hybridization were performed to examine the BACE1-AS level in CRC. IGF2BP2 binding to m6A motifs in BACE1-AS was determined by RIP assay and S1m-tagged immunoprecipitation. Transwell assay and liver metastasis mice model experiments were performed to examine the metastasis capabilities of BACE1-AS knockout cells. Stemness-like properties was examined by tumor sphere assay and the expression of stemness biomarkers. Microarray data were acquired to analyze the signaling pathways involved in BACE1-AS promoting CRC metastasis. RESULTS: BACE1-AS is the most up-regulated in metastatic CRC associated with unfavorable prognosis. Sequence blast revealed two m6A motifs in BACE1-AS. IGF2BP2 binding to these two m6A motifs is required for BACE1-AS boost in metastatic CRC. m6A modified BACE1-AS drives CRC cells migration and invasion and liver metastasis both in vitro and in vivo. Moreover, BACE1-AS maintains the stemness-like properties of CRC cells. Mechanically, BACE1-AS promoted TUFT1 expression by ceRNA network through miR-214-3p. CRC patients with such ceRNA network suffer poorer prognosis than ceRNA-negative patients. Depletion of TUFT1 mimics BACE1-AS loss. BACE1-AS activated Wnt signaling pathway in a TUFT1 dependent manner. BACE1-AS/miR-214-3p/TUFT1/Wnt signaling regulatory axis is essential for CRC liver metastasis. Pharmacologic inhibition of Wnt signaling pathway repressed liver metastasis and stemness-like features in BACE1-AS over-expressed CRC cells. CONCLUSION: Our study demonstrated BACE1-AS as a novel target of IGF2BP2 through m6A modification. m6A modified BACE1-AS promotes CRC liver metastasis through TUFT1 dependent activation of Wnt signaling pathway. Thus, targeting BACE1-AS and its downstream Wnt signaling pathways may provide a new opportunity for metastatic CRC intervention and treatment.


Assuntos
Secretases da Proteína Precursora do Amiloide , Neoplasias Colorretais , Proteínas do Esmalte Dentário , Neoplasias Hepáticas , RNA Antissenso , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Via de Sinalização Wnt , RNA Antissenso/metabolismo , Ácido Aspártico Endopeptidases/genética , Secretases da Proteína Precursora do Amiloide/genética , Neoplasias Hepáticas/secundário , Linhagem Celular Tumoral , Adenosina/análogos & derivados , Humanos , Proteínas de Ligação a RNA/metabolismo , Proteínas do Esmalte Dentário/metabolismo
14.
Cancer Lett ; 579: 216479, 2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-37924938

RESUMO

Therapy resistance is the primary problem in treating late-stage colorectal cancer (CRC). Claudins are frequently dysregulated in cancer, and several are being investigated as novel therapeutic targets and biomarkers. We have previously demonstrated that Claudin-1 (CLDN1) expression in CRC promotes epithelial-mesenchymal transition, metastasis, and resistance to anoikis. Here, we hypothesize that CLDN1 promotes cancer stemness and chemoresistance in CRC. We found that high CLDN1 expression in CRC is associated with cancer stemness and chemoresistance signaling pathways in patient datasets, and it promotes chemoresistance both in vitro and in vivo. Using functional stemness assays, proteomics, biophysical binding assays, and patient-derived organoids, we found that CLDN1 promotes properties of cancer stemness including CD44 expression, tumor-initiating potential, and chemoresistance through a direct interaction with ephrin type-A receptor 2 (EPHA2) tyrosine kinase. This interaction is dependent on the CLDN1 PDZ-binding motif, increases EPHA2 protein expression by inhibiting its degradation, and enhances downstream AKT signaling and CD44 expression to promote stemness and chemoresistance. These results suggest CLDN1 is a viable target for pharmacological intervention and/or biomarker development.


Assuntos
Neoplasias Colorretais , Humanos , Linhagem Celular Tumoral , Claudina-1/genética , Claudina-1/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Transdução de Sinais
15.
Eur J Cancer ; 195: 113396, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37924647

RESUMO

BACKGROUND: Upfront anti-EGFR therapy represents the standard of care for patients with left-sided, MSS/pMMR, RAS and BRAF wild-type mCRC. Molecular 'hyperselection' may optimize EGFR inhibition by detecting additional resistance alterations. MATERIALS AND METHODS: We used comprehensive genomic profiling on archival samples of elderly patients enrolled in the PANDA trial to detect: HER2 amplification/mutations; MET amplification; NTRK/ROS1/ALK/RET rearrangements; PIK3CA exon 20 mutations; PTEN alterations; AKT1 mutations; MAP2K1 mutations. We defined 'Gene Altered' (GA) patients whose tumour harboured at least one alteration, and 'Hyperselected' (HS) those without. Survival and tumour response outcomes were correlated to hyperselection status alone or combined with primary tumour sidedness or treatment arm. RESULTS: Genomic alterations were detected in 41/147 patients (27.9%). PFS, OS and ORR were inferior in GA versus HS (median PFS: 7.6 versus 12.8 months, HR = 2.08, 95% CI: 1.43-3.03, p < 0.001; median OS: 20.0 versus 29.5 months, HR = 1.82, 95% CI:1.23-2.69, p = 0.002; ORR: 51% versus 71%; OR = 0.43, 95% CI: 0.21-0.91, p = 0.02). In the multivariable models, the impact of hyperselection on PFS and OS was confirmed. Lower ORR was observed with 5-FU/LV/panitumumab in GA (40% versus 62%), but not in HS (70% versus 72%). GA was associated with worse survival and response regardless of primary tumour sidedness, whereas in the HS subgroup, right-and left sided tumours had similar outcomes. CONCLUSIONS: Molecular hyperselection and comprehensive genomic profiling have a potential usefulness in elderly patients with RAS/BRAF wild-type, pMMR/MSS mCRC, eligible for upfront EGFR inhibition.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Neoplasias Retais , Idoso , Humanos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Receptores ErbB , Fluoruracila/uso terapêutico , Panitumumabe/uso terapêutico , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas B-raf/genética
16.
Biomed Pharmacother ; 168: 115690, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37939611

RESUMO

Colorectal cancer (CRC) is the most prevalent cancer of the digestive tract. Herba Patriniae (also known as Bai Jiang Cao, HP) have been widely used to manage diarrhea, ulcerative colitis, and several cancers, including CRC. Nonetheless, the molecular mechanisms underlying the pharmacological action of HP on CRC remain unclear. This study investigated the underlying mechanisms of HP against CRC using network pharmacology analysis and in vitro and in vivo experiments. The results revealed nine bioactive compounds of HP. Furthermore, 3460 CRC-related targets of the identified active compounds were predicted from the Gene Expression Omnibus (GEO) database. Furthermore, 65 common targets were identified through the intersection of two related targets. Moreover, ten hub genes, including CDK4, CDK2, CDK1, CCND1, CCNB1, CCNA2, MYC, E2F1, CHEK1, and CDKN1A were identified through the topological analysis. Meanwhile, the GO and KEGG pathway analysis revealed that the core target genes were majorly enriched in the p53 and HIF-1 signaling pathways. Moreover, HP promoted apoptosis and suppressed cell proliferation by activating the p53 signaling pathway in a dose-dependent manner, while a similar effect was observed for Isovitexin (the primary component of HP). Overall, this study provides valuable insights into the underlying mechanisms of HP and its component Isovitexin against CRC, providing a theoretical foundation for additional experimental verification of its clinical application.


Assuntos
Neoplasias Colorretais , Medicamentos de Ervas Chinesas , Proteína Supressora de Tumor p53 , Apoptose , Pontos de Checagem do Ciclo Celular , Genes cdc , Proteína Supressora de Tumor p53/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Humanos , Medicamentos de Ervas Chinesas/farmacologia , Antineoplásicos Fitogênicos/farmacologia
17.
Proc Natl Acad Sci U S A ; 120(47): e2313137120, 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37972068

RESUMO

KRAS is the most commonly mutated oncogene. Targeted therapies have been developed against mediators of key downstream signaling pathways, predominantly components of the RAF/MEK/ERK kinase cascade. Unfortunately, single-agent efficacy of these agents is limited both by intrinsic and acquired resistance. Survival of drug-tolerant persister cells within the heterogeneous tumor population and/or acquired mutations that reactivate receptor tyrosine kinase (RTK)/RAS signaling can lead to outgrowth of tumor-initiating cells (TICs) and drive therapeutic resistance. Here, we show that targeting the key RTK/RAS pathway signaling intermediates SOS1 (Son of Sevenless 1) or KSR1 (Kinase Suppressor of RAS 1) both enhances the efficacy of, and prevents resistance to, the MEK inhibitor trametinib in KRAS-mutated lung (LUAD) and colorectal (COAD) adenocarcinoma cell lines depending on the specific mutational landscape. The SOS1 inhibitor BI-3406 enhanced the efficacy of trametinib and prevented trametinib resistance by targeting spheroid-initiating cells in KRASG12/G13-mutated LUAD and COAD cell lines that lacked PIK3CA comutations. Cell lines with KRASQ61 and/or PIK3CA mutations were insensitive to trametinib and BI-3406 combination therapy. In contrast, deletion of the RAF/MEK/ERK scaffold protein KSR1 prevented drug-induced SIC upregulation and restored trametinib sensitivity across all tested KRAS mutant cell lines in both PIK3CA-mutated and PIK3CA wild-type cancers. Our findings demonstrate that vertical inhibition of RTK/RAS signaling is an effective strategy to prevent therapeutic resistance in KRAS-mutated cancers, but therapeutic efficacy is dependent on both the specific KRAS mutant and underlying comutations. Thus, selection of optimal therapeutic combinations in KRAS-mutated cancers will require a detailed understanding of functional dependencies imposed by allele-specific KRAS mutations.


Assuntos
Neoplasias Colorretais , Fosfatidilinositol 3-Quinases , Humanos , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , MAP Quinase Quinase Quinases/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo
18.
J Exp Clin Cancer Res ; 42(1): 322, 2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38012650

RESUMO

BACKGROUND: Lines of evidence indicated that, immune checkpoints (ICs) inhibitors enhanced T cell immune response to exert anti-tumor effects. However, T cell exhaustion has been so far a major obstacle to antitumor immunotherapy in colorectal cancer patients. Our previous studies showed that ginseng-derived nanoparticles (GDNPs) inhibited the growth of various tumors by reprograming tumor-associated macrophages (TAMs) and downregulated the ICs expression on T cells in tumor microenvironment (TME), but the underlying effector mechanisms remained unclear. METHODS: The correlation between arginase-1 (ARG1) and T cells was computed based on the colorectal cancer patients in TCGA database. In vitro, we observed that GDNPs reprogrammed TAMs inhibited ARG1 release and ultimately ameliorated T cell exhaustion according to several techniques including WB, PCR, ELISA and flow cytometry. We also used an in vivo MC38 tumor-bearing model and administered GDNPs to assess their anti-tumor effects through multiple indices. The mechanism that GDNPs improved T cell exhaustion was further clarified using the bioinformatics tools and flow cytometry. RESULTS: GDNPs reprogramed TAMs via reducing ARG1 production. Moreover, normalized arginine metabolism ameliorated T cell exhaustion through mTOR-T-bet axis, resulting in reduced ICs expression and enhanced CD8+ T cells expansion. CONCLUSIONS: By regulating the mTOR-T-bet axis, GDNPs reprogramed macrophages to regulate ARG1 release, which further ameliorated T cell exhaustion in TME. These findings provided new insights into comprehending the mechanisms underlying the mitigation of T cell exhaustion, which may facilitate the development of innovative therapeutic strategies in the field of cancer treatment.


Assuntos
Neoplasias Colorretais , Nanopartículas , Panax , Humanos , Linfócitos T CD8-Positivos/metabolismo , Arginase/metabolismo , Arginase/farmacologia , Microambiente Tumoral , Exaustão das Células T , Macrófagos/metabolismo , Neoplasias Colorretais/patologia , Serina-Treonina Quinases TOR/metabolismo
19.
Pathol Res Pract ; 252: 154958, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37988793

RESUMO

As one of the most common cancers worldwide, the incidence of colorectal cancer (CRC) continues to increase. Metastasis is the leading cause of death for this malignant disease. Regulator of telomere elongation helicase 1 (RTEL1) is a key factor that maintains telomere stability and contributes to DNA repair. Recent advances have suggested that RTEL1 exerts other functions through various mechanisms. However, little is known about its role in human cancers, including CRC. In this study, we revealed that the copy number of RTEL1 was significantly higher in CRC tissues, especially in metastatic CRC tissues, than in paired normal tissues, which was associated with increased expression. Increased RTEL1 expression was significantly correlated with CRC progression and poor survival. Functionally, RTEL1 knockdown suppressed the proliferation and metastasis of CRC both in vitro and in vivo. In addition, multiple signalling pathways, including the mitotic cell cycle, DNA repair, and EMT, were potentially regulated by RTEL1. Notably, GPR17 appeared to be a candidate downstream target gene that partially mediated the tumor-promoting effects of RTEL1 in CRC. Altogether, our results indicate that RTEL1 plays a crucial role in CRC progression and appears to be a promising therapeutic target and prognosis for CRC.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/patologia , Prognóstico , Reparo do DNA , Transdução de Sinais , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Movimento Celular/genética , Receptores Acoplados a Proteínas G/genética , DNA Helicases/genética , DNA Helicases/metabolismo
20.
Int J Mol Sci ; 24(22)2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-38003292

RESUMO

S-adenosylhomocysteine hydrolase (AHCY) deficiency results mainly in hypermethioninemia, developmental delay, and is potentially fatal. In order to shed new light on molecular aspects of AHCY deficiency, in particular any changes at transcriptome level, we enabled knockdown of AHCY expression in the colon cancer cell line SW480 to simulate the environment occurring in AHCY deficient individuals. The SW480 cell line is well known for elevated AHCY expression, and thereby represents a suitable model system, in particular as AHCY expression is regulated by MYC, which, on the other hand, is involved in Wnt signaling and the regulation of Wnt-related genes, such as the ß-catenin co-transcription factor LEF1 (lymphoid enhancer-binding factor 1). We selected LEF1 as a potential target to investigate its association with S-adenosylhomocysteine hydrolase deficiency. This decision was prompted by our analysis of RNA-Seq data, which revealed significant changes in the expression of genes related to the Wnt signaling pathway and genes involved in processes responsible for epithelial-mesenchymal transition (EMT) and cell proliferation. Notably, LEF1 emerged as a common factor in these processes, showing increased expression both on mRNA and protein levels. Additionally, we show alterations in interconnected signaling pathways linked to LEF1, causing gene expression changes with broad effects on cell cycle regulation, tumor microenvironment, and implications to cell invasion and metastasis. In summary, we provide a new link between AHCY deficiency and LEF1 serving as a mediator of changes to the Wnt signaling pathway, thereby indicating potential connections of AHCY expression and cancer cell phenotype, as Wnt signaling is frequently associated with cancer development, including colorectal cancer (CRC).


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Humanos , Adenosil-Homocisteinase/genética , Adenosil-Homocisteinase/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias Colorretais/patologia , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral , Via de Sinalização Wnt/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...