Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.438
Filtrer
1.
J Ethnopharmacol ; 336: 118754, 2025 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-39208999

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Tubeimoside-I (TBM) promotes various cancer cell death by increasing the reactive oxygen species (ROS) production. However, the specific molecular mechanisms of TBM and its impact on oxaliplatin-mediated anti-CRC activity are not yet fully understood. AIM OF THE STUDY: To elucidate the therapeutic effect and underlying molecular mechanism of TBM on oxaliplatin-mediated anti-CRC activity. MATERIALS AND METHODS: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation, wound healing assays and flow cytometry were conducted to investigate the changes in cell phenotypes and ROS generation. Real-time quantitative PCR (qRT-PCR) and western blotting were performed to detect the expressions of related mRNA and proteins. Finally, mouse xenograft models demonstrated that synergistic anti-tumor effects of combined treatment with TBM and oxaliplatin. RESULTS: The synergistic enhancement of the anti-tumor effects of oxaliplatin in colon cancer cells by TBM involved in the regulation of ROS-mediated endoplasmic reticulum (ER) stress, C-jun-amino-terminal kinase (JNK), and p38 MAPK signaling pathways. Mechanistically, TBM increased ROS generation in colon cancer cells by inhibiting heat shock protein 60 (HSPD1) expression. Knocking down HSPD1 increased TBM-induced antitumor activity and ROS generation in colon cancer cells. The mouse xenograft tumor models further validated that the combination therapy exhibited stronger anti-tumor effects than monotherapy alone. CONCLUSIONS: Combined therapy with TBM and oxaliplatin might be an effective therapeutic strategy for some CRC patients.


Sujet(s)
Tumeurs colorectales , Synergie des médicaments , Stress du réticulum endoplasmique , Oxaliplatine , Espèces réactives de l'oxygène , Saponines , Triterpènes , Animaux , Humains , Mâle , Souris , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Lignée cellulaire tumorale , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Cellules HCT116 , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Souris nude , Oxaliplatine/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Saponines/pharmacologie , Triterpènes/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Sci Rep ; 14(1): 21827, 2024 09 18.
Article de Anglais | MEDLINE | ID: mdl-39294234

RÉSUMÉ

Zinc finger protein 263 (ZNF263) is frequently upregulated in various tumor types; however, its function and regulatory mechanism in colorectal cancer (CRC) have not yet been elucidated. In this study, the expression of ZNF263 was systematically examined using data from The Cancer Genome Atlas database and samples from patients with CRC. The results indicated that high expression of ZNF263 in CRC tissues is significantly associated with tumor grade, lymph node metastasis and disant metastasis. Additionally, overexpression of ZNF263 significantly promoted the proliferation, invasion, migration, and epithelial-mesenchymal transition of CRC cells, while also increasing signal transducer and activator of transcription 3 (STAT3) expression and mRNA stability. Conversely, knockdown of ZNF263 inhibited the malignant behavior of CRC cells and decreased STAT3 expression and mRNA stability. Further mechanism studies using chromatin immunoprecipitation (CHIP) and luciferase assays verified that ZNF263 directly binds to the STAT3 promoter. Rescue experiments demonstrated that the knockdown or overexpression of STAT3 could significantly reverse the effects of ZNF263 on CRC cells. Additionally, our study found that overexpression of ZNF263 enhanced the resistance of CRC cells to the chemoradiotherapy. In summary, this study not only elucidated the significant role of ZNF263 in CRC but also proposed novel approaches and methodologies for the diagnosis and treatment of this malignancy.


Sujet(s)
Prolifération cellulaire , Tumeurs colorectales , Protéines de liaison à l'ADN , Régulation de l'expression des gènes tumoraux , Facteur de transcription STAT-3 , Femelle , Humains , Mâle , Adulte d'âge moyen , Lignée cellulaire tumorale , Mouvement cellulaire , Chimioradiothérapie/méthodes , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Évolution de la maladie , Résistance aux médicaments antinéoplasiques/génétique , Transition épithélio-mésenchymateuse/génétique , Facteur de transcription STAT-3/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme
3.
Sci Rep ; 14(1): 21852, 2024 09 19.
Article de Anglais | MEDLINE | ID: mdl-39300121

RÉSUMÉ

5-Fluorouracil (5-FU) is used as a standard first-line drug for colorectal cancer malignancy (CRC), but it brings a series of side effects such as severe diarrhea and intestinal damage. Our previous study found that a large number of senescent cells increased while 5-Fu induced intestinal damage, and anti-senescence drugs can alleviate its side effects of inflammatory damage. Oleanolic acid (OA) is a common pentacyclic triterpenoid mainly derived from food fungi and medicinal plants, and studies have shown that it mainly possesses hepatoprotective, enzyme-lowering, anti-inflammatory, and anti-tumor effects. But its role in senescence is still unclear. In the present study, we demonstrated for the first time that OA ameliorated 5-Fu-induced human umbilical vein endothelial cells (HUVECs) and human normal intestinal epithelial cells (NCM460) in a 5-Fu-induced cellular senescence model by decreasing the activity of SA-ß-gal-positive cells, and the expression of senescence-associated proteins (p16), senescence-associated genes (p53 and p21), and senescence-associated secretory phenotypes (SASPs: IL-1ß, IL-6, IL-8, IFN-γ and TNF-α). Meanwhile, in this study, in a BALB/c mouse model, we demonstrated that 5-FU induced intestinal inflammatory response and injury, which was also found to be closely related to the increase of senescent cells, and that OA treatment was effective in ameliorating these adverse phenomena. Furthermore, our in vivo and in vitro studies showed that OA could alleviate senescence by inhibiting mTOR. In colon cancer cell models, OA also enhanced the ability of 5-FU to kill HCT116 cells and SW480 cells. Overall, this study demonstrates for the first time the potential role of OA in counteracting the side effects of 5-FU chemotherapy, providing a new option for the treatment of colorectal cancer to progressively achieve the goal of high efficacy and low toxicity of chemotherapy.


Sujet(s)
Vieillissement de la cellule , Fluorouracil , Cellules endothéliales de la veine ombilicale humaine , Inflammation , Acide oléanolique , Acide oléanolique/pharmacologie , Fluorouracil/effets indésirables , Fluorouracil/pharmacologie , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Animaux , Souris , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Souris de lignée BALB C , Intestins/effets des médicaments et des substances chimiques , Intestins/anatomopathologie , Mâle , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie
4.
BMC Cancer ; 24(1): 1162, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39300378

RÉSUMÉ

BACKGROUND: Despite a high incidence of colorectal carcinoma, data regarding genetic aberrations in colorectal carcinoma (CRC) patients in Pakistan is scarce. This study aimed to determine the frequency of BRAFV600E mutations in colorectal carcinoma tissue in the Pakistani population and to associate BRAFV600E expression with CD133, a marker of colorectal stem cells, and CDX2 marker of differentiation. METHODS: Sanger Sequencing of exon 15 (426 bp) including the hotspot V600E was performed on formalin-fixed-paraffin-embedded (FFPE) CRC tissue samples of 115 patients. The samples were subjected to immunohistochemistry (IHC) to assess the expression of BRAFV600E, CDX2, and CD133. Additionally, homology modelling and docking were performed to investigate novel deletions revealed in sequencing. RESULTS: Twenty-four (20.8%) BRAF variants were identified in the coding region, with V600E mutations detected in 14 (12.2% )cases (GenBank: PP003258.1; Pop Set: 2678087296). Moreover, a wide spectrum of novel non-V600E mutations (8.6%) were identified, including deletions and missense variations. In-silico analysis revealed that due to large deletions in the coding region of three samples, the affinity of the anti-BRAF drugs (Encorafenib and Vemurafenib) for the active site decreased in comparison to the wild type. The IHC analysis showed that BRAFV600E expression was significantly associated with CD133 expression (χ2(1, n=115) = 26.351; p = < 0.001) and with CDX2 expression (χ2(1, n=115) = 14.88; p = 0.001). Multivariate analysis using binary logistic regression revealed association of BRAFV600E mutations with age (OR = 1.123; CI = 1.024-1.232; p = 0.014), gender (OR = 0.071; CI = 0.006-0.831; p = 0.035), grade (0.007; CI = 0-0.644) and CD133 expression (OR = 65.649; CI = 2.153-2001.556; p = 0.016). CONCLUSION: The present study demonstrates a notably high V600E frequency (12.2%) in comparison to global reported data, which ranges from 0.4 to 18%. This finding reflects the importance of upfront BRAF testing of the genetically distinct population of Pakistan. Previously unreported mutations identified in the sample may be of clinical significance and warrant further investigation. The concomitant high expression and significant association between CD133 and BRAFV600E represent vital actionable genes that may be targeted together to improve CRC patient management.


Sujet(s)
Antigène AC133 , Facteurs de transcription CDX2 , Tumeurs colorectales , Mutation , Protéines proto-oncogènes B-raf , Humains , Protéines proto-oncogènes B-raf/génétique , Facteurs de transcription CDX2/génétique , Facteurs de transcription CDX2/métabolisme , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Antigène AC133/génétique , Antigène AC133/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Adulte , Pakistan/épidémiologie , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Études de cohortes , Immunohistochimie , Sujet âgé de 80 ans ou plus
5.
Mol Med Rep ; 30(5)2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39301629

RÉSUMÉ

The advancement of tumor cell metastasis is significantly influenced by epithelial­to­mesenchymal transition (EMT), and metastasis is a prominent contributor to the mortality of patients diagnosed with colorectal cancer (CRC). AT­rich interactive domain­containing protein 1A (ARID1A), which acts as a tumor suppressor, frequently exhibits a loss­of­function mutation in metastatic CRC tissues. However, the underlying molecular mechanisms of ARID1A relating to EMT remain poorly understood. The present study aimed to clarify the association between ARID1A and EMT regulation in human CRC cells. The investigation into the loss of ARID1A expression in tissues from patients with CRC was performed using immunohistochemistry. Furthermore, ARID1A­overexpressing SW48 cells were established using lentiviruses carrying human full­length ARID1A. The results revealed that overexpression of ARID1A induced cellular morphological changes by promoting the tight junction molecule zonula occludens 1 (ZO­1) and the adherens junction molecule E­cadherin, whereas it decreased the intermediate filament protein vimentin. The results of reverse transcription­quantitative PCR also confirmed that ARID1A overexpression upregulated the mRNA expression levels of TJP1/ZO­1 and CDH1/E­cadherin, and downregulated VIM/vimentin and zinc finger E­box binding homeobox 1 expression, which are considered epithelial and mesenchymal markers, respectively. In addition, the overexpression of ARID1A in CRC cells resulted in a suppression of cell motility and migratory capabilities. The present study also demonstrated that the tumor suppressor ARID1A was commonly absent in CRC tissues. Notably, ARID1A overexpression could reverse the EMT­like phenotype and inhibit cell migration through alterations in EMT­related markers, leading to the inhibition of malignant progression. In conclusion, ARID1A may serve as a biomarker and therapeutic target in the clinical management of metastatic CRC.


Sujet(s)
Mouvement cellulaire , Tumeurs colorectales , Protéines de liaison à l'ADN , Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Facteurs de transcription , Humains , Transition épithélio-mésenchymateuse/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Mouvement cellulaire/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Lignée cellulaire tumorale , Mâle , Adulte d'âge moyen , Femelle , Sujet âgé , Adulte
6.
Cell Death Dis ; 15(9): 675, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39277583

RÉSUMÉ

Rap2b, a proto-oncogene upregulated in colorectal cancer (CRC), undergoes protein S-palmitoylation at specific C-terminus sites (C176/C177). These palmitoylation sites are crucial for Rap2b localization on the plasma membrane (PM), as mutation of C176 or C177 results in cytosolic relocation of Rap2b. Our study demonstrates that Rap2b influences cell migration and invasion in CRC cells, independent of proliferation, and this activity relies on its palmitoylation. We identify ABHD17a as the depalmitoylating enzyme for Rap2b, altering PM localization and inhibiting cell migration and invasion. EGFR/PI3K signaling regulates Rap2b palmitoylation, with PI3K phosphorylating ABHD17a to modulate its activity. These findings highlight the potential of targeting Rap2b palmitoylation as an intervention strategy. Blocking the C176/C177 sites using an interacting peptide attenuates Rap2b palmitoylation, disrupting PM localization, and suppressing CRC metastasis. This study offers insights into therapeutic approaches targeting Rap2b palmitoylation for the treatment of metastatic CRC, presenting opportunities to improve patient outcomes.


Sujet(s)
Membrane cellulaire , Tumeurs colorectales , Lipoylation , Protéines G rap , Animaux , Humains , Souris , Lignée cellulaire tumorale , Membrane cellulaire/métabolisme , Mouvement cellulaire , Prolifération cellulaire , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Récepteurs ErbB/métabolisme , Souris nude , Métastase tumorale , Phosphatidylinositol 3-kinases/métabolisme , Proto-oncogène Mas , Protéines G rap/métabolisme , Protéines G rap/génétique , Transduction du signal
7.
Biomed Pharmacother ; 179: 117428, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39255737

RÉSUMÉ

Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Serotonin (5-HT) is a biogenic monoamine that acts as a neurotransmitter in the central nervous system and as a paracrine, exocrine, or endocrine messenger in peripheral tissues. In this study, we hypothesized that inhibition of serotonin signaling using 5-HT receptor 2B (HTR2B) inhibitors could potentially impede the progression of CRC. We treated CT26 and COLO-205 cells with SB204741, an inhibitor of HTR2B, and evaluated CRC cell proliferation and migration. We then evaluated the effects of HTR2B inhibition in a xenograft mouse model of human colorectal cancer. We also evaluated the role of a novel inhibitor, GM-60186, using both in vitro and in vivo models. RNA sequencing analysis was performed to elucidate the underlying mechanism of the anti-tumor effects of pharmacological inhibition of HTR2B on CRC. In both CRC cell lines and xenograft mouse models, we show that pharmacological inhibition of HTR2B with SB204741 and GM-60186 significantly inhibits CRC cell proliferation and migration. HTR2B inhibition leads to the suppression of extracellular signal-regulated kinase (ERK) signaling, a critical pathway in CRC pathogenesis. Notably, transcriptomic analysis reveals distinct gene expression changes associated with HTR2B inhibition, providing insight into its therapeutic potential. In this study, we found that pharmacological inhibition of HTR2B suppressed CRC proliferation via ERK signaling. In addition, we proposed a novel HTR2B inhibitor for the treatment of CRC. This study highlights the potential role of HTR2B signaling in CRC. These inhibitors may contribute to new therapeutics for CRC treatment.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Tumeurs colorectales , Système de signalisation des MAP kinases , Récepteur de la sérotonine de type 5-HT2B , Sérotonine , Animaux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteur de la sérotonine de type 5-HT2B/métabolisme , Lignée cellulaire tumorale , Sérotonine/métabolisme , Sérotonine/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Souris nude , Souris de lignée BALB C , Antagonistes des récepteurs 5-HT2 de la sérotonine/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
8.
Epigenomics ; 16(15-16): 1097-1115, 2024.
Article de Anglais | MEDLINE | ID: mdl-39268727

RÉSUMÉ

Aim: This study investigated the role of lncRNA LINC01232 in ferroptosis of colorectal cancer (CRC).Materials & methods: Real time quantitative polymerase chain reaction or western blot experiments were performed to examine relevant mRNAs and proteins expression. The kit assays evaluated malondialdehyde, iron, Fe2+ and glutathione levels. ROS levels were verified by flow cytometry. Chromatin immunoprecipitation and RNA immunoprecipitation analysis monitored the correlation among LINC01232, H3K27ac, p300 and ARNTL2.Results: LINC01232 or ARNTL2 knockdown facilitated erastin-induced ferroptosis. The interaction between LINC01232 and p300 resulted in the enhancement of H3K27ac levels at ARNTL2 promoter to promote ARNTL2 transcriptional activity. ARNTL2 overexpression reversed the promoting effect of LINC01232 knockdown on ferroptosis.Conclusion: LINC01232 inhibited the ferroptosis in CRC by epigenetically upregulating the transcriptional activity of ARNTL2.


Colorectal cancer (CRC) is a malignant disease of the digestive tract that occurs worldwide, which has high morbidity and mortality but has not effective targeted therapy. Ferroptosis has emerged as a new target for treating CRC since its proposed in 2012. Long noncoding RNAs are noncoding RNAs with a length greater than 200 nucleotides and their role in ferroptosis of cancer cells has attracted more and more attention in recent years. Herein, our study explored the effect of long noncoding RNA LINC01232 on CRC progression. This research exhibited the relationship between LINC01232 and the ferroptosis at the occurrence and development of CRC, which is expected to provide a potential therapeutic target for the treatment of CRC.


Sujet(s)
Facteurs de transcription ARNTL , Tumeurs colorectales , Ferroptose , ARN long non codant , Activation de la transcription , Humains , Ferroptose/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , ARN long non codant/génétique , Facteurs de transcription ARNTL/génétique , Facteurs de transcription ARNTL/métabolisme , Histone/métabolisme , Protéine p300-E1A/métabolisme , Protéine p300-E1A/génétique , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale
9.
Int J Biol Sci ; 20(12): 4618-4634, 2024.
Article de Anglais | MEDLINE | ID: mdl-39309424

RÉSUMÉ

Some colorectal cancer patients have experienced normal epithelial transformation into inflammatory and tumor states, but the molecular basis still needs to be further determined. The expression levels of SIX4 are gradually increased in dextran sodium sulfate (DSS) and azoxymethane (AOM)/DSS-induced colonic epithelial inflammation and tumors, respectively, in mice. Targeting SIX4 alleviates intestinal inflammation occurrence and reduces adenoma formation in mice. Clinical sample assays indicated that SIX4 is upregulated in inflammatory bowel disease (IBD) and colorectal cancer (CRC) tissues compared to normal colorectal tissues. In a subsequent study, we found that SIX4, transcriptionally activated by the proinflammatory IL-6/STAT3 signal, binds to c-Jun to transcribe IL-6, thus forming a positive IL-6/STAT3/SIX4/c-Jun feedback loop, which further induces intestinal inflammation occurrence. In addition, elevated SIX4 also induces the expression of DeltaNp63, rather than wild-type p63, by binding to its promoter and thus facilitates the activation of tumor stemness signals, which ultimately leads to the formation of colorectal cancer. Our study first observes that activated SIX4 in inflammation induction drives the transformation of colorectal epithelium into inflammation and tumor, which demonstrates SIX4 as a significant therapeutic target in IBD and colitis-associated colorectal cancer (CAC) and CRC pathogenesis.


Sujet(s)
Tumeurs colorectales , Inflammation , Transduction du signal , Animaux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Souris , Inflammation/métabolisme , Humains , Interleukine-6/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Sulfate dextran , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/induit chimiquement , Souris de lignée C57BL , Muqueuse intestinale/métabolisme , Oxyde de diméthyl-diazène
10.
Int J Biol Sci ; 20(12): 4999-5026, 2024.
Article de Anglais | MEDLINE | ID: mdl-39309442

RÉSUMÉ

Background: Colorectal cancer (CRC) remains a significant global health challenge, often characterized by late-stage metastasis and poor prognosis. The Runt-related transcription factor 1 (RUNX1) plays a dual role as both an oncogene and a tumor suppressor in various cancers, including CRC. However, the specific regulatory mechanisms of RUNX1 in CRC, particularly its direct roles, are not fully understood. Objective: This study aimed to investigate the role of RUNX1 in CRC progression and its interaction with Mucin 13 (MUC13) as a potential regulatory target. Methods: RUNX1 expression was analyzed in CRC tissues and cell lines compared to controls. In vitro and in vivo assays were conducted to assess the effects of RUNX1 overexpression and knockdown on cell behavior. ChIP-seq and RNA-seq analyses were performed to identify RUNX1 targets, with a focus on MUC13. Results: RUNX1 expression was significantly upregulated in CRC tissues and cells, correlating with advanced pathological characteristics and poor patient outcomes. RUNX1 overexpression enhanced CRC cell proliferation, migration, invasion, and G2/M phase arrest, while its knockdown had the opposite effects. MUC13 was identified as a direct transcriptional target of RUNX1, with its expression contributing to the activation of the Wnt/ß-catenin signaling pathway. Disruption of MUC13 partially reversed the malignant phenotypes induced by RUNX1. Conclusion: RUNX1 promotes CRC progression by upregulating MUC13 and activating the Wnt/ß-catenin pathway. This RUNX1-MUC13 axis represents a potential therapeutic target for managing CRC.


Sujet(s)
Tumeurs colorectales , Sous-unité alpha 2 du facteur CBF , Voie de signalisation Wnt , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Humains , Sous-unité alpha 2 du facteur CBF/métabolisme , Sous-unité alpha 2 du facteur CBF/génétique , Lignée cellulaire tumorale , Mucines/métabolisme , Mucines/génétique , Animaux , Prolifération cellulaire/génétique , Souris , Mâle , Femelle , Souris nude , Régulation de l'expression des gènes tumoraux , Mouvement cellulaire/génétique , bêta-Caténine/métabolisme , Métastase tumorale , Adulte d'âge moyen , Souris de lignée BALB C
11.
Theranostics ; 14(14): 5443-5460, 2024.
Article de Anglais | MEDLINE | ID: mdl-39310106

RÉSUMÉ

Rationale : the proto-oncogene KRAS is frequently mutated in colorectal cancer (CRC), leading to inherent resistance against monoclonal antibodies targeting the epidermal growth factor receptor (EGFR), such as cetuximab. Therefore, addressing the primary resistance and expanding the indications for target therapy have become critical challenges. Methods : the screening of a natural product library against KRAS mutant CRC cells was conducted, leading to the discovery of a small molecule compound that sensitive to the KRASG13D mutation site. The anti-tumor activity of this small molecule compound in combination with cetuximab was evaluated using the KRASG13D mutant CRC models both in vivo and in vitro. This evaluation includes an examination of its effects on cell proliferation, viability, apoptosis, cell cycle progression, and tumor growth. Furthermore, RNA sequencing, western blot analysis, immunofluorescence, real-time quantitative PCR, and pull-down assays were employed to explore the molecular mechanisms underlying the synergistic anti-tumor effect of this small molecule compound in combination with cetuximab. Results : our study screened 882 compounds in KRAS mutant CRC cells and identified honokiol, a small molecule compound that exhibits specific sensitivity to KRASG13D mutant CRC cells. Furthermore, we revealed that the synergistic augmentation of cetuximab's sensitivity in vivo and in vitro models of KRASG13D mutant CRC in combination with honokiol. Mechanistically, honokiol suppresses SNX3-retromer mediated trafficking, thereby impeding lysosomal proteolytic capacity and inhibiting autophagy and macropinocytosis fluxes. Moreover, honokiol inhibits the conversion of RAS GDP to RAS GTP, heightening the susceptibility of KRASG13D CRC mutant cells to cetuximab. Conclusions : honokiol enhances the sensitivity of cetuximab by destroying SNX3 retromer in KRASG13D mutant CRC preclinical model. These findings present a promising strategy for expanding the indications of target therapy in KRAS mutant colorectal cancer patients.


Sujet(s)
Apoptose , Dérivés du biphényle , Prolifération cellulaire , Cétuximab , Tumeurs colorectales , Lignanes , Mutation , Protéines proto-oncogènes p21(ras) , Cétuximab/pharmacologie , Cétuximab/usage thérapeutique , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Humains , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Animaux , Lignanes/pharmacologie , Lignanes/usage thérapeutique , Lignée cellulaire tumorale , Dérivés du biphényle/pharmacologie , Dérivés du biphényle/usage thérapeutique , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Proto-oncogène Mas , Synergie des médicaments , Souris nude , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Souris de lignée BALB C , Composés allyliques , Phénols
12.
BMC Cancer ; 24(1): 1141, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39267014

RÉSUMÉ

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


Sujet(s)
Tumeurs colorectales , Curcumine , Microbiome gastro-intestinal , Souris de lignée C57BL , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Curcumine/pharmacologie , Tumeurs colorectales/microbiologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/induit chimiquement , Souris , Mâle , Modèles animaux de maladie humaine , Oxyde de diméthyl-diazène/toxicité , Carcinogenèse/effets des médicaments et des substances chimiques , Sulfate dextran/toxicité , ARN ribosomique 16S/génétique , Métabolomique/méthodes , Humains
13.
J Exp Clin Cancer Res ; 43(1): 261, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39267084

RÉSUMÉ

BACKGROUND: Long-term accumulation of misfolded proteins leads to endoplasmic reticulum (ER) stress in colorectal cancer (CRC). However, the precise pathways controlling the decision between survival and apoptosis in CRC are unclear. Therefore, in this study, we investigated the function and molecular mechanism of glucosidase I (GCS1) in regulating ER stress in CRC. METHODS: A public database was used to confirm the expression level of GCS1 in CRC and normal tissues. Clinical samples from our center were used to confirm the mRNA and protein expression levels of GCS1. Cell proliferation, migration, invasion, and apoptosis assays revealed the biological role of GCS1. Immunohistochemical techniques were used to evaluate the expression of key proteins in subcutaneous implanted tumors in nude mice, which provided further evidence for the biological function of GCS1 in promoting cancer in vivo. The results of coimmunoprecipitation-mass spectrometry analysis and immunofluorescence colocalization analysis the interaction between GCS1 and GRP78. In addition, the mechanism of action of USP10, GRP78, and GCS1 at the post- translational level was investigated. Finally, a tissue microarray was used to examine the connection between GCS1 and GRP78 expression and intracellular localization of these proteins using immunohistochemistry and immunofluorescence. RESULTS: The experimental results revealed that GCS1 was substantially expressed in CRC, with higher expression indicating a worse prognosis. Thus, GCS1 can enhance the proliferation and metastasis while inhibiting the apoptosis of CRC cells both in vivo and in vitro. Mechanistically, GCS1 binds to GRP78, recruits USP10 for deubiquitination of GRP78 to promote its degradation, and decreases ER stress-mediated apoptosis, increasing CRC cell proliferation and metastasis. CONCLUSIONS: In summary, GCS1 stimulates CRC growth and migration and reduces ER stress-mediated apoptosis via USP10-mediated deubiquitination of GRP78. Our findings identify a possible therapeutic target for CRC.


Sujet(s)
Tumeurs colorectales , Évolution de la maladie , Chaperonne BiP du réticulum endoplasmique , Stress du réticulum endoplasmique , Protéines du choc thermique , Ubiquitin thiolesterase , Ubiquitination , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Chaperonne BiP du réticulum endoplasmique/métabolisme , Animaux , Souris , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Protéines du choc thermique/métabolisme , Protéines du choc thermique/génétique , Souris nude , Prolifération cellulaire , Mâle , Lignée cellulaire tumorale , Apoptose , Femelle , Mouvement cellulaire
14.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39273090

RÉSUMÉ

Colorectal cancer (CRC) represents a significant challenge in oncology, with increasing incidence and mortality rates worldwide, particularly among younger adults. Despite advancements in treatment modalities, the urgent need for more effective therapies persists. Immunotherapy has emerged as a beacon of hope, offering the potential for improved outcomes and quality of life. This review delves into the critical interplay between ferroptosis, an iron-dependent form of regulated cell death, and immunotherapy within the CRC context. Ferroptosis's influence extends beyond tumor cell fate, reshaping the tumor microenvironment (TME) to enhance immunotherapy's efficacy. Investigations into Ferroptosis-related Genes (OFRGs) reveal their pivotal role in modulating immune cell infiltration and TME composition, closely correlating with tumor responsiveness to immunotherapy. The integration of ferroptosis inducers with immunotherapeutic strategies, particularly through novel approaches like ferrotherapy and targeted co-delivery systems, showcases promising avenues for augmenting treatment efficacy. Furthermore, the expression patterns of OFRGs offer novel prognostic tools, potentially guiding personalized and precision therapy in CRC. This review underscores the emerging paradigm of leveraging ferroptosis to bolster immunotherapy's impact, highlighting the need for further research to translate these insights into clinical advancements. Through a deeper understanding of the ferroptosis-immunotherapy nexus, new therapeutic strategies can be developed, promising enhanced efficacy and broader applicability in CRC treatment, ultimately improving patient outcomes and quality of life in the face of this formidable disease.


Sujet(s)
Tumeurs colorectales , Ferroptose , Immunothérapie , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Tumeurs colorectales/thérapie , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Immunothérapie/méthodes , Animaux
15.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273409

RÉSUMÉ

Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.


Sujet(s)
Tumeurs colorectales , Humains , Tumeurs colorectales/thérapie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/étiologie , Tumeurs colorectales/génétique , Carcinogenèse/génétique , Animaux , Facteurs de risque
16.
Eur J Med Chem ; 278: 116801, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-39241481

RÉSUMÉ

The rational installation of pharmacophores targeting HSP90 and LSD1 axes has achieved significant anti-cancer capacity in prostate and colorectal cancer. Among the series of hybrids, inhibitor 6 exhibited remarkable anti-proliferative activity against prostate cancer cell lines PC-3 and DU145, with GI50 values of 0.24 and 0.30 µM, respectively. It demonstrated notable efficacy in combinatorial attack and cell death initiation towards apoptosis. The cell death process was mediated by PARP induction and γH2AX signaling, and was also characterized as caspase-dependent and Bcl-xL/Bax-independent. Notably, no difference in eye size or morphology was observed in the zebrafish treated with compound 6 compared to the reference group (AUY922). The profound treatment response in docetaxel-resistant PC-3 cells highlighted the dual inhibitory ability in improving docetaxel sensitivity. Additionally, at a minimum concentration of 1.25 µM, compound 6 effectively inhibited the growth of patient-derived colorectal cancer (CRC) organoids for up to 10 days in vitro. Together, the designed HSP90/LSD1 inhibitors present a novel route and significant clinical value for anti-cancer drug therapy.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Tumeurs colorectales , Tests de criblage d'agents antitumoraux , Protéines du choc thermique HSP90 , Histone Demethylases , Organoïdes , Tumeurs de la prostate , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/métabolisme , Humains , Mâle , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Animaux , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/anatomopathologie , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Relation structure-activité , Structure moléculaire , Relation dose-effet des médicaments , Danio zébré , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale
17.
Cancer Med ; 13(17): e70169, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39225546

RÉSUMÉ

BACKGROUND: Colorectal cancer (CRC) is a prevalent and lethal tumor, with metastasis being the leading cause of mortality. Previous research has indicated that the long non-coding RNA (lncRNA) CCAT2 is involved in the regulation of various tumor progression mechanisms. However, the precise role of CCAT2 in CRC proliferation and metastasis remains ambiguous. This study seeks to elucidate the mechanisms through which CCAT2 influences CRC. METHODS: High-throughput sequencing and RT-qPCR were used to detect CCAT2 expression in CRC. Functional analyses including CCK8, colony formation, wound healing migration, transwell chamber, and Muse® Cell Analyzer assays were performed to study the effects of CCAT2 gene deletion on CRC cells. RNA-pulldown and protein mass spectrometry were employed to identify the interaction between CCAT2 and GNB2 protein. RESULTS: Increased CCAT2 expression was found in CRC, especially in metastatic CRC. Deletion of CCAT2 gene inhibited CRC cell proliferation, migration, and invasion while promoting apoptosis. The interaction between CCAT2 and GNB2 protein was shown to modulate GNB2 protein alterations and affect the ERK and Wnt signaling pathways, thereby promoting CRC proliferation and metastasis. CONCLUSION: CCAT2 plays a crucial role in CRC progression by modulating the ERK and Wnt signaling pathways through its interaction with GNB2. These findings highlight the importance of CCAT2 as a key regulatory element in the mechanisms underlying CRC proliferation and metastasis.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , ARN long non codant , Voie de signalisation Wnt , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Apoptose , Lignée cellulaire tumorale , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Système de signalisation des MAP kinases , Métastase tumorale , ARN long non codant/génétique , ARN long non codant/métabolisme
18.
Cell Death Dis ; 15(9): 654, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39231945

RÉSUMÉ

Transforming growth factor (TGF)-ß signaling is critical for epithelial-mesenchymal transition (EMT) and colorectal cancer (CRC) metastasis. Disruption of Smad-depednent TGF-ß signaling has been shown in CRC cells. However, TGF-ß receptor remains expressed on CRC cells. Here, we investigated whether the cooperation between tumor-associated N-glycosylation and a glycan-binding protein modulated the TGF-ß-driven signaling and metastasis of CRC. We showed that galectin-8, a galactose-binding lectin, hampered TGF-ß-induced EMT by interacting with the type II TGF-ß receptor and competing with TGF-ß binding. Depletion of galectin-8 promoted the migration of CRC cells by increasing TGF-ß-receptor-mediated RAS and Src signaling, which was attenuated after recombinant galectin-8 treatment. Treatment with recombinant galectin-8 also induces JNK-dependent apoptosis in CRC cells. The anti-migratory effect of galectin-8 depended on ß4-galactosyltransferase-I (B4GALT1), an enzyme involved in N-glycan synthesis. Increased B4GALT1 expression was observed in clinical CRC samples. Depletion of B4GALT1 reduced the metastatic potential of CRC cells. Furthermore, inducible expression of galectin-8 attenuated tumor development and metastasis of CRC cells in an intra-splenic injection model. Our results thus demonstrate that galectin-8 alters non-canonical TGF-ß response in CRC cells and suppresses CRC progression.


Sujet(s)
Mouvement cellulaire , Tumeurs colorectales , Transition épithélio-mésenchymateuse , Galactosyltransferases , Galectines , Métastase tumorale , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Galectines/métabolisme , Galectines/génétique , Galactosyltransferases/métabolisme , Galactosyltransferases/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Évolution de la maladie , Lignée cellulaire tumorale , Transduction du signal , Souris , Récepteurs TGF-bêta/métabolisme , Souris nude , Liaison aux protéines , Apoptose/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/pharmacologie , Souris de lignée BALB C
19.
Sci Rep ; 14(1): 21570, 2024 09 16.
Article de Anglais | MEDLINE | ID: mdl-39284825

RÉSUMÉ

Heterogeneous nuclear ribonucleoproteins (hnRNPs), a group of proteins that control gene expression, have been implicated in many post-transcriptional processes. SYNCRIP (also known as hnRNP Q), a subtype of hnRNPs, has been reported to be involved in mRNA splicing and translation. In addition, the deregulation of SYNCRIP was found in colorectal cancer (CRC). However, the role of SYNCRIP in regulating CRC growth remains largely unknown. Here, we found that SYNCRIP was highly expressed in colorectal cancer by analyzing TCGA and GEPIA database. Furthermore, we confirmed the expression of SYNCRIP expression in CRC tumor and CRC cell lines. Functionally, SYNCRIP depletion using shRNA in CRC cell lines (SW480 and HCT 116) resulted in increased caspase3/7 activity and decreased cell proliferation, as well as migration. Meanwhile, overexpression of SYNCRIP showed opposite results. Mechanistically, SYNCRIP regulated the expression of DNA methyltransferases (DNMT) 3A, but not DNMT1 or DNMT3B, which affected the expression of tumor suppressor, p16. More importantly, our in vivo experiments showed that SYNCRIP depletion significantly inhibited colorectal tumor growth. Taken all together, our results suggest SYNCRIP as a potent therapeutic target in colorectal cancer.


Sujet(s)
Carcinogenèse , Prolifération cellulaire , Tumeurs colorectales , DNA (cytosine-5-)-methyltransferase , DNA methyltransferase 3A , Régulation de l'expression des gènes tumoraux , Régulation positive , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , DNA (cytosine-5-)-methyltransferase/métabolisme , DNA (cytosine-5-)-methyltransferase/génétique , Prolifération cellulaire/génétique , DNA methyltransferase 3A/métabolisme , Animaux , Carcinogenèse/génétique , Lignée cellulaire tumorale , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/génétique , Souris , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Mouvement cellulaire/génétique , Cellules HCT116 , Souris nude
20.
Nat Commun ; 15(1): 8208, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39294166

RÉSUMÉ

Germline and somatic TP53 variants play a crucial role during tumorigenesis. However, genetic variations that solely affect the alternatively spliced p53 isoforms, p53ß and p53γ, are not fully considered in the molecular diagnosis of Li-Fraumeni syndrome and cancer. In our search for additional cancer predisposing variants, we identify a heterozygous stop-lost variant affecting the p53ß isoforms (p.*342Serext*17) in four families suspected of an autosomal dominant cancer syndrome with colorectal, breast and papillary thyroid cancers. The stop-lost variant leads to the 17 amino-acid extension of the p53ß isoforms, which increases oligomerization to canonical p53α and dysregulates the expression of p53's transcriptional targets. Our study reveals the capacity of p53ß mutants to influence p53 signalling and contribute to the susceptibility of different cancer types. These findings underscore the significance of p53 isoforms and the necessity of comprehensive investigation into the entire TP53 gene in understanding cancer predisposition.


Sujet(s)
Prédisposition génétique à une maladie , Mutation germinale , Syndrome de Li-Fraumeni , Pedigree , Isoformes de protéines , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Femelle , Mâle , Syndrome de Li-Fraumeni/génétique , Adulte , Adulte d'âge moyen , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/anatomopathologie , Cancer papillaire de la thyroïde/métabolisme , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Épissage alternatif/génétique , Tumeurs/génétique , Tumeurs/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE