Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 13.552
Filtrer
1.
Medicine (Baltimore) ; 103(31): e39057, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093763

RÉSUMÉ

BACKGROUND: The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, poses a huge threat to human health. Pancreatic cancer (PC) is a malignant tumor with high mortality. Research suggests that infection with SARS-CoV-2 may increase disease severity and risk of death in patients with pancreatic cancer, while pancreatic cancer may also increase the likelihood of contracting SARS-CoV-2, but the link is unclear. METHODS: This study investigated the transcriptional profiles of COVID-19 and PC patients, along with their respective healthy controls, using bioinformatics and systems biology approaches to uncover the molecular mechanisms linking the 2 diseases. Specifically, gene expression data for COVID-19 and PC patients were obtained from the Gene Expression Omnibus datasets, and common differentially expressed genes (DEGs) were identified. Gene ontology and pathway enrichment analyses were performed on the common DEGs to elucidate the regulatory relationships between the diseases. Additionally, hub genes were identified by constructing a protein-protein interaction network from the shared DEGs. Using these hub genes, we conducted regulatory network analyses of microRNA/transcription factors-genes relationships, and predicted potential drugs for treating COVID-19 and PC. RESULTS: A total of 1722 and 2979 DEGs were identified from the transcriptome data of PC (GSE119794) and COVID-19 (GSE196822), respectively. Among these, 236 common DEGs were found between COVID-19 and PC based on protein-protein interaction analysis. Functional enrichment analysis indicated that these shared DEGs were involved in pathways related to viral genome replication and tumorigenesis. Additionally, 10 hub genes, including extra spindle pole bodies like 1, holliday junction recognition protein, marker of proliferation Ki-67, kinesin family member 4A, cyclin-dependent kinase 1, topoisomerase II alpha, cyclin B2, ubiquitin-conjugating enzyme E2 C, aurora kinase B, and targeting protein for Xklp2, were identified. Regulatory network analysis revealed 42 transcription factors and 23 microRNAs as transcriptional regulatory signals. Importantly, lucanthone, etoposide, troglitazone, resveratrol, calcitriol, ciclopirox, dasatinib, enterolactone, methotrexate, and irinotecan emerged as potential therapeutic agents against both COVID-19 and PC. CONCLUSION: This study unveils potential shared pathogenic mechanisms between PC and COVID-19, offering novel insights for future research and therapeutic strategies for the treatment of PC and SARS-CoV-2 infection.


Sujet(s)
COVID-19 , Biologie informatique , Tumeurs du pancréas , Cartes d'interactions protéiques , SARS-CoV-2 , Biologie des systèmes , Humains , COVID-19/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/virologie , Biologie informatique/méthodes , Biologie des systèmes/méthodes , SARS-CoV-2/génétique , Cartes d'interactions protéiques/génétique , Réseaux de régulation génique , microARN/génétique , microARN/métabolisme , Analyse de profil d'expression de gènes/méthodes
2.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 148-154, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097882

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal and aggressive tumor that affects the digestive tract, leading to high mortality and poor survival rates. The purpose of the present study was to evaluate the expression levels of DNA damage-inducible transcript 3 (DDIT3) in pancreatic cancer and to investigate its effects in in vitro and in vivo experiments. Bioinformatics analysis indicated that DDIT3 expression was higher in pancreatic cancer tumor tissues and associated with a poor prognosis. Positive or strong positive DDIT3 expression was observed in PDAC, and no or weak expression was observed in normal pancreatic tissues. It was also highly expressed in PDAC cells, while being expressed at lower levels in normal pancreatic ductal epithelial cells. Transfection of short hairpin RNA targeting the DDIT3 gene reduced the proliferation, migration and invasion of PANC-1 cells. In vivo, in an in situ implantation tumor model with Pan02 cells, the size and weight of the tumors were reduced in the DDIT3 knockdown Pan02 cell-implanted group. These data suggested that DDIT3 represents a novel predictive biomarker for the potential treatment of patients presenting with PDAC.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas , Facteur de transcription CHOP , Humains , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Lignée cellulaire tumorale , Animaux , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Mâle , Femelle , Souris , Pronostic , Souris nude , Adulte d'âge moyen
3.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 100-105, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097890

RÉSUMÉ

Goosecoid (GSC), translated from a homeobox gene, is a protein that participates in metastasis of various cancers. Pancreatic adenocarcinoma (PAAD) is one of the deadliest malignancies associated with a poor diagnosis and prognosis. To develop new treatment target or biomarker for PAAD, this study intended to assess the effects and the molecular mechanism of GSC on PAAD metastasis. The expressive discrepancy of GSC in PAAD and normal tissues/cells was compared by both the quantitative PCR and western blot. The effects of GSC silencing and GSC over-expression on PAAD cells and TGF-ß signaling were proved by wound-healing assay, cell counting kit-8, Transwell assay and western blot. From the results, GSC mRNA and protein levels were enriched in PAAD cancer tissues and cells. GSC silencing prohibited metastasis of PAAD cells including the ability to invade, migrate and epithelial-mesenchymal transition (EMT), whereas GSC upregulation stimulated these cells behaviors above. GSC silencing reversed the effects on cellular processes induced by activation of the TGF-ß pathway. Furthermore, silencing of GSC postponed tumor growth in xenograft model. In summary, GSC was abundantly expressed in PAAD, which activated the TGF-ß pathway to enhance cell metastasis and tumor development.


Sujet(s)
Adénocarcinome , Transition épithélio-mésenchymateuse , Métastase tumorale , Tumeurs du pancréas , Transduction du signal , Facteur de croissance transformant bêta , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Humains , Facteur de croissance transformant bêta/métabolisme , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Adénocarcinome/génétique , Animaux , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/génétique , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Souris nude , Souris , Mâle , Femelle , Souris de lignée BALB C , Extinction de l'expression des gènes , Prolifération cellulaire , Protéines et peptides de signalisation intercellulaire
4.
Cell Death Dis ; 15(8): 553, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090116

RÉSUMÉ

Pancreatic cancer is an aggressive cancer with a poor prognosis. Metabolic abnormalities are one of the hallmarks of pancreatic cancer, and pancreatic cancer cells can adapt to biosynthesis, energy intake, and redox needs through metabolic reprogramming to tolerate nutrient deficiency and hypoxic microenvironments. Pancreatic cancer cells can use glucose, amino acids, and lipids as energy to maintain malignant growth. Moreover, they also metabolically interact with cells in the tumour microenvironment to change cell fate, promote tumour progression, and even affect immune responses. Importantly, metabolic changes at the body level deserve more attention. Basic research and clinical trials based on targeted metabolic therapy or in combination with other treatments are in full swing. A more comprehensive and in-depth understanding of the metabolic regulation of pancreatic cancer cells will not only enrich the understanding of the mechanisms of disease progression but also provide inspiration for new diagnostic and therapeutic approaches.


Sujet(s)
Tumeurs du pancréas , Microenvironnement tumoral , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Animaux , Métabolisme énergétique
5.
Medicine (Baltimore) ; 103(28): e38843, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996145

RÉSUMÉ

Early detection of pancreatic adenocarcinoma (PAAD) remains a pressing clinical problem. Information on the clinical prognostic value of mitochondrial fusion-related genes in PAAD remains limited. In this study, we investigated mitochondrial fusion-related genes of PAAD to establish an optimal signature plate for the early diagnosis and prognosis of PAAD. The cancer genome atlas database was used to integrate the Fragments Per Kilobase Million data and related clinical data for patients with PAAD. Least absolute shrinkage and selection operator regression, cox regression, operating characteristic curves, and cBioPortal database was used to evaluate model performance, assess the prognostic ability and sensitivity. The levels of immune infiltration were compared by CIBERSORT, QUANTISEQ, and EPIC. Chemotherapy sensitivity between the different risk groups was compared by the Genomics of Drug Sensitivity in Cancer database and the "pRRophetic" R package. At last, a total of 4 genes were enrolled in multivariate Cox regression analysis. The risk-predictive signature was constructed as: (0.5438 × BAK1) + (-1.0259 × MIGA2) + (1.1140 × PARL) + (-0.4300 × PLD6). The area under curve of these 4 genes was 0.89. Cox regression analyses indicates the signature was an independent prognostic indicator (P < .001, hazard ratio [HR] = 1.870, 95% CI = 1.568-2.232). Different levels of immune cell infiltration in the 2 risk groups were observed using the 3 algorithms, with tumor mutation load (P = .0063), tumor microenvironment score (P = .01), and Tumor Immune Dysfunction and Exclusion score (P = .0012). The chemotherapeutic sensitivity analysis also revealed that the half-maximal inhibitory concentration of 5-fluorouracil (P = .0127), cisplatin (P = .0099), docetaxel (P < .0001), gemcitabine (P = .0047), and pacilataxel (P < .0001) were lower in the high-risk groups, indicating that the high-risk group patients had a greater sensitivity to chemotherapy. In conclude, we established a gene signature plate comprised of 4 mitochondrial fusion related genes to facilitate early diagnosis and prognostic prediction of PAAD.


Sujet(s)
Adénocarcinome , Marqueurs biologiques tumoraux , Tumeurs du pancréas , Humains , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Pronostic , Adénocarcinome/génétique , Adénocarcinome/traitement médicamenteux , Adénocarcinome/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Mâle , Femelle , Adulte d'âge moyen , Dynamique mitochondriale/génétique , Sujet âgé , Modèles des risques proportionnels , Dépistage précoce du cancer/méthodes
6.
Methods Mol Biol ; 2823: 77-93, 2024.
Article de Anglais | MEDLINE | ID: mdl-39052215

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a lethal solid malignancy with many patients succumbing to the disease within 6 months of diagnosis. The mechanisms that underlie PDAC initiation and progression are poorly understood. Current treatment options are primarily limited to chemotherapy, which is often provided with palliative intent. Unfortunately, there are no robust biomarkers to guide treatment selection or monitor treatment response. This is concerning given the increasing incidence of this cancer. We and others have generated organoid models to explore the biology underlying PDAC with the goal of identifying new therapeutic targets. Here we provide protocols to generate a preclinical PDAC organoid model and methods to use these to define the proteomic landscape of this cancer.


Sujet(s)
Carcinome du canal pancréatique , Organoïdes , Tumeurs du pancréas , Protéomique , Organoïdes/métabolisme , Protéomique/méthodes , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Animaux , Protéome , Marqueurs biologiques tumoraux/métabolisme , Souris
7.
Aging (Albany NY) ; 16(13): 11103-11116, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38990159

RÉSUMÉ

Homeobox C4 (HOXC4) is a member of homeobox family and acts as a transcription factor in regulating morphological development. The current study aimed to determine its role in pancreatic cancer (PC). Bioinformatics analysis was employed to assess the expression and clinical significance of HOXC4 in PC, while the expression of HOXC4 was further confirmed in PC tissues through quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). The impact of HOXC4 on PC cell proliferation was evaluated using various assays including Cell Counting Kit-8, colony formation, apoptosis detection, cell cycle analysis, and subcutaneous tumorigenesis. Extracellular acidification rate, glucose uptake, and lactate production measurements were detected to examine the impact of HOXC4 on glycolysis. The relationship between HOXC4 and lactate dehydrogenase A (LDHA) was investigated using CHIP assay, luciferase reporter assay, and western blot. Notably, there was a substantial increase in HOXC4 expression in PC, and patients with elevated HOXC4 levels exhibited shorter survival durations. HOXC4 knockdown resulted in significantly reduced proliferation and colony formation in PC cells, accompanied by increased apoptosis and G1 phase arrest. The overexpression of HOXC4 resulted in contrasting effects. In vivo, the proliferation of PC cells was diminished upon the knockdown of HOXC4. HOXC4 exhibited an increase in LDHA expression by binding to its promoter. The suppressive effects of HOXC4 knockdown on PC cells were counteracted upon the restoration of LDHA. In conclusion, HOXC4 promoted the proliferation of PC cells by increasing LDHA-mediated glycolysis. HOXC4 can act as a target for PC therapy.


Sujet(s)
Prolifération cellulaire , Glycolyse , Protéines à homéodomaine , Tumeurs du pancréas , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Prolifération cellulaire/génétique , Glycolyse/génétique , Lignée cellulaire tumorale , Animaux , Souris , Régulation de l'expression des gènes tumoraux , L-Lactate dehydrogenase/métabolisme , L-Lactate dehydrogenase/génétique , Apoptose/génétique , Mâle , Femelle , Souris nude
8.
BMC Cancer ; 24(1): 809, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38973003

RÉSUMÉ

BACKGROUND: Pancreatic Ductal Adenocarcinoma (PDAC) is an aggressive cancer characterized by an immunosuppressive microenvironment. Patients from specific ethnicities and population groups have poorer prognoses than others. Therefore, a better understanding of the immune landscape in such groups is necessary for disease elucidation, predicting patient outcomes and therapeutic targeting. This study investigated the expression of circulating key immune cell markers in South African PDAC patients of African ancestry. METHODS: Blood samples were obtained from a total of 6 healthy volunteers (HC), 6 Chronic Pancreatitis (CP) and 34 PDAC patients consisting of 22 resectable (RPC), 8 locally advanced (LAPC) and 4 metastatic (MPC). Real-time Quantitative Polymerase Chain reactions (RT-qPCR), Metabolomics, Enzyme-Linked Immunosorbent Assay (ELISA), Reactive Oxygen Species (ROS), and Immunophenotyping assays were conducted. Statistical analysis was conducted in R (v 4.3.2). Additional analysis of single-cell RNA data from 20 patients (16 PDAC and 4 controls) was conducted to interrogate the distribution of T-cell and Natural Killer cell populations. RESULTS: Granulocyte and neutrophil levels were significantly elevated while lymphocytes decreased with PDAC severity. The total percentages of CD3 T-cell subpopulations (helper and double negative T-cells) decreased when compared to HC. Although both NK (p = 0.014) and NKT (p < 0.001) cell levels increased as the disease progressed, their subsets: NK CD56dimCD16- (p = 0.024) and NKTs CD56+ (p = 0.008) cell levels reduced significantly. Of note is the negative association of NK CD56dimCD16- (p < 0.001) cell levels with survival time. The gene expression analyses showed no statistically significant correlation when comparing the PDAC groups with the controls. The inflammatory status of PDAC was assessed by ROS levels of serum which were elevated in CP (p = 0.025), (RPC (p = 0.003) and LAPC (p = 0.008)) while no significant change was observed in MPC, compared to the HC group. ROS was shown to be positively correlated with GlycA (R = 0.45, p = 0.0096). Single-cell analyses showed a significant difference in the ratio of NKT cells per total cell counts in LAPC (p < 0.001) and MPC (p < 0.001) groups compared with HC, confirming observations in our sample group. CONCLUSION: The expression of these immune cell markers observed in this pilot study provides insight into their potential roles in tumour progression in the patient group and suggests their potential utility in the development of immunotherapeutic strategies.


Sujet(s)
Carcinome du canal pancréatique , Évolution de la maladie , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , République d'Afrique du Sud , Sujet âgé , Adulte , Marqueurs biologiques tumoraux/génétique , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Pancréatite chronique/immunologie , Pancréatite chronique/génétique , Pancréatite chronique/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Immunophénotypage
9.
Pancreas ; 53(7): e603-e610, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38986080

RÉSUMÉ

OBJECTIVES: Pancreatic cancer (PC) is one of the most aggressive malignancies due to the high rate of metastasis. The mechanisms underlying metastasis need to be elucidated. Small extracellular vesicles (sEVs) mediate cell-to-cell communication, and cancer-derived sEVs contribute to the formation of premetastatic niches. The present study examined changes in adhesiveness by the internalization of PC-derived sEVs into vascular endothelial cells, and investigated the molecular mechanisms underlying metastasis. MATERIALS AND METHODS: Pancreatic cancer-derived sEVs were internalized into vascular endothelial cells, and changes in adhesiveness were evaluated. We evaluated the effects of sEVs on the formation of liver metastasis in vivo. We also assessed molecular changes in vascular endothelial cells by the internalization of PC-derived sEVs. RESULTS: The internalization of PC-derived sEVs into vascular endothelial cells promoted the adhesiveness of vascular endothelial cells and PC cells. Pancreatic cancer-derived sEVs contained high levels of transforming growth factor ß1 mRNA and acted as its transporter. Once PC-derived sEVs were internalized into vascular endothelial cells, the expression of fibronectin 1 increased on the cell surface, and the adhesiveness of vascular endothelial cells was enhanced. CONCLUSIONS: We investigated association between PC-derived sEVs and adhesiveness. Regulation of PC-derived sEVs has potential as a therapeutic modality to suppress the metastasis of PC.


Sujet(s)
Adhérence cellulaire , Cellules endothéliales , Vésicules extracellulaires , Fibronectines , Tumeurs du pancréas , Facteur de croissance transformant bêta-1 , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Vésicules extracellulaires/métabolisme , Humains , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Animaux , Fibronectines/métabolisme , Lignée cellulaire tumorale , Facteur de croissance transformant bêta-1/métabolisme , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/secondaire , Souris nude , Communication cellulaire , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Mâle
10.
Nat Commun ; 15(1): 5763, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982051

RÉSUMÉ

While high circulating tumor DNA (ctDNA) levels are associated with poor survival for multiple cancers, variant-specific differences in the association of ctDNA levels and survival have not been examined. Here we investigate KRAS ctDNA (ctKRAS) variant-specific associations with overall and progression-free survival (OS/PFS) in first-line metastatic pancreatic ductal adenocarcinoma (mPDAC) for patients receiving chemoimmunotherapy ("PRINCE", NCT03214250), and an independent cohort receiving standard of care (SOC) chemotherapy. For PRINCE, higher baseline plasma levels are associated with worse OS for ctKRAS G12D (log-rank p = 0.0010) but not G12V (p = 0.7101), even with adjustment for clinical covariates. Early, on-therapy clearance of G12D (p = 0.0002), but not G12V (p = 0.4058), strongly associates with OS for PRINCE. Similar results are obtained for the SOC cohort, and for PFS in both cohorts. These results suggest ctKRAS G12D but not G12V as a promising prognostic biomarker for mPDAC and that G12D clearance could also serve as an early biomarker of response.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome du canal pancréatique , ADN tumoral circulant , Tumeurs du pancréas , Protéines proto-oncogènes p21(ras) , Humains , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/mortalité , Carcinome du canal pancréatique/sang , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/traitement médicamenteux , Protéines proto-oncogènes p21(ras)/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/sang , Tumeurs du pancréas/mortalité , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Femelle , Mâle , ADN tumoral circulant/sang , ADN tumoral circulant/génétique , Adulte d'âge moyen , Sujet âgé , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/génétique , Pronostic , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Mutation , Survie sans progression , Métastase tumorale
11.
Dtsch Med Wochenschr ; 149(15): 887-893, 2024 Aug.
Article de Allemand | MEDLINE | ID: mdl-39013409

RÉSUMÉ

Neuroendocrine neoplasms are classified according to the WHO classification based on morphological criteria into neuroendocrine tumors, neuroendocrine carcinomas, and mixed neuroendocrine-non-neuroendocrine neoplasms. Neuroendocrine tumors are well differentiated neoplasms and show characteristic site-specific histological and molecular features, which is important for their clinical management. In cases dealing with metastasis, pathology often can help to identify the primary tumors using a small immunohistochemical marker panel. Neuroendocrine carcinomas are poorly differentiated neoplasms. They are subdivided into neuroendocrine carcinomas of small cell and large cell type. The molecular profile of neuroendocrine carcinomas and mixed neuroendocrine-non-neuroendocrine neoplasms shows a close relationship to conventional adenocarcinomas with site-specific features. Molecular analysis of neuroendocrine carcinomas and neuroendocrine-non-neuroendocrine neoplasms are not yet fully integrated in daily diagnostics and are mainly performed in the context of precision oncology.


Sujet(s)
Tumeurs neuroendocrines , Tumeurs du pancréas , Humains , Marqueurs biologiques tumoraux/génétique , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/génétique , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie
12.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000545

RÉSUMÉ

Chemotherapy treatment against pancreatic ductal adenocarcinoma (PDAC) is thwarted by tumoral activation of multiple therapy resistance pathways. The growth hormone (GH)-GH receptor (GHR) pair is a covert driver of multimodal therapy resistance in cancer and is overexpressed in PDAC tumors, yet the therapeutic potential of targeting the same has not been explored. Here, we report that GHR expression is a negative prognostic factor in patients with PDAC. Combinations of gemcitabine with different GHR antagonists (GHRAs) markedly improve therapeutic outcomes in nude mice xenografts. Employing cultured cells, mouse xenografts, and analyses of the human PDAC transcriptome, we identified that attenuation of the multidrug transporter and epithelial-to-mesenchymal transition programs in the tumors underlie the observed augmentation of chemotherapy efficacy by GHRAs. Moreover, in human PDAC patients, GHR expression strongly correlates with a gene signature of tumor promotion and immune evasion, which corroborate with that in syngeneic tumors in wild-type vs. GH transgenic mice. Overall, we found that GH action in PDAC promoted a therapy-refractory gene signature in vivo, which can be effectively attenuated by GHR antagonism. Our results collectively present a proof of concept toward considering GHR antagonists to improve chemotherapeutic outcomes in the highly chemoresistant PDAC.


Sujet(s)
Carcinome du canal pancréatique , Désoxycytidine , , Tumeurs du pancréas , Récepteur STH , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Humains , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Souris , Récepteur STH/métabolisme , Récepteur STH/antagonistes et inhibiteurs , Récepteur STH/génétique , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Lignée cellulaire tumorale , Souris nude , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Femelle
13.
Cancer Res ; 84(14): 2297-2312, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39005053

RÉSUMÉ

Metabolic reprogramming is a hallmark of cancer and is crucial for cancer progression, making it an attractive therapeutic target. Understanding the role of metabolic reprogramming in cancer initiation could help identify prevention strategies. To address this, we investigated metabolism during acinar-to-ductal metaplasia (ADM), the first step of pancreatic carcinogenesis. Glycolytic markers were elevated in ADM lesions compared with normal tissue from human samples. Comprehensive metabolic assessment in three mouse models with pancreas-specific activation of KRAS, PI3K, or MEK1 using Seahorse measurements, nuclear magnetic resonance metabolome analysis, mass spectrometry, isotope tracing, and RNA sequencing analysis revealed a switch from oxidative phosphorylation to glycolysis in ADM. Blocking the metabolic switch attenuated ADM formation. Furthermore, mitochondrial metabolism was required for de novo synthesis of serine and glutathione (GSH) but not for ATP production. MYC mediated the increase in GSH intermediates in ADM, and inhibition of GSH synthesis suppressed ADM development. This study thus identifies metabolic changes and vulnerabilities in the early stages of pancreatic carcinogenesis. Significance: Metabolic reprogramming from oxidative phosphorylation to glycolysis mediated by MYC plays a crucial role in the development of pancreatic cancer, revealing a mechanism driving tumorigenesis and potential therapeutic targets. See related commentary by Storz, p. 2225.


Sujet(s)
Métaplasie , Tumeurs du pancréas , Animaux , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Souris , Métaplasie/métabolisme , Métaplasie/anatomopathologie , Glycolyse , Carcinogenèse/métabolisme , Cellules acineuses/métabolisme , Cellules acineuses/anatomopathologie , Phosphorylation oxydative , Glutathion/métabolisme , Reprogrammation cellulaire , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Mâle , Mitochondries/métabolisme , Mitochondries/anatomopathologie ,
14.
Theranostics ; 14(10): 4090-4106, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994016

RÉSUMÉ

Purpose: Due to intrinsic defensive response, ferroptosis-activating targeted therapy fails to achieve satisfactory clinical benefits. Though p62-Keap1-Nrf2 axis is activated to form a negative feedback loop during ferroptosis induction, how p62 is activated remains largely unknown. Methods: MTS assay was applied to measure cell growth. Lipid ROS was detected with C11-BODIPY reagent by flow cytometer. Quantitative real-time PCR (qPCR) and western blotting were performed to determine mRNA and protein level. Immunofluorescence (IF) was performed to examine the distribution of proteins. Fluorescence recovery after photobleaching (FRAP) was adopted to evaluate p62 phase separation. Immunoprecipitation (IP), co-IP and Proximal ligation assay (PLA) were performed to detected protein posttranslational modifications and protein-protein interactions. Tumor xenograft model was employed to inspect in vivo growth of pancreatic cancer cells. Results: Upon ferroptosis induction, Nuclear Factor E2 Related Factor 2 (Nrf2) protein and its downstream genes such as HMOX1 and NQO1 were upregulated. Knockdown of p62 significantly reversed Nrf2 upregulation and Keap1 decrease after ferroptosis induction. Knockdown of either p62 or Nrf2 remarkably sensitized ferroptosis induction. Due to augmented p62 phase separation, formation of p62 bodies were increased to recruit Keap1 after ferroptosis induction. Protein arginine methyltransferase 6 (PRMT6) mediated asymmetric dimethylarginine (ADMA) of p62 to increase its oligomerization, promoting p62 phase separation and p62 body formation. Knockdown of p62 or PRMT6 notably sensitized pancreatic cancer cells to ferroptosis both in vitro and in vivo through suppressing Nrf2 signaling. Conclusion: During ferroptosis induction, PRMT6 mediated p62 ADMA to promote its phase separation, sequestering Keap1 to activate Nrf2 signaling and inhibit ferroptosis. Therefore, targeting PRMT6-mediated p62 ADMA could be a new option to sensitize ferroptosis for cancer treatment.


Sujet(s)
Arginine , Ferroptose , Protéine-1 de type kelch associée à ECH , Facteur-2 apparenté à NF-E2 , Protein-arginine N-methyltransferases , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Animaux , Arginine/métabolisme , Arginine/analogues et dérivés , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Souris , Lignée cellulaire tumorale , Protéine-1 de type kelch associée à ECH/métabolisme , Protéine-1 de type kelch associée à ECH/génétique , Rétrocontrôle physiologique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Séquestosome-1/métabolisme , Séquestosome-1/génétique , Souris nude , Transduction du signal , , Protéines de liaison à l'ARN
16.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000128

RÉSUMÉ

Epigenetic changes are common in cancer and include aberrant DNA methylation and histone modifications, including both acetylation or methylation. DNA methylation in the promoter regions and histone deacetylation are usually accompanied by gene silencing, and may lead to the suppression of tumor suppressors in cancer cells. An interaction between epigenetic pathways has been reported that could be exploited to more efficiently target aggressive cancer cells, particularly those against which current treatments usually fail, such as pancreatic cancer. In this study, we explored the possibility to combine the DNA demethylating agent 5-AZA with HDAC inhibitor SAHA to treat pancreatic cancer cell lines, focusing on the acetylation of mutp53 and the consequences on its stability, as well as on the interaction of this protein with c-myc and BRCA-1, key molecules in cancer survival. The results obtained suggest that SAHA/5-AZA combination was more effective than single treatments to promote the degradation of mutp53, to upregulate p21 and downregulate c-Myc and BRCA-1, thus increasing DNA damage and cytotoxicity in pancreatic cancer cells.


Sujet(s)
Protéine BRCA1 , Inhibiteur p21 de kinase cycline-dépendante , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas , Protéines proto-oncogènes c-myc , Protéine p53 suppresseur de tumeur , Vorinostat , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Acétylation/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Vorinostat/pharmacologie , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Azacitidine/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie
17.
Mol Cancer ; 23(1): 140, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982491

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis and limited therapeutic options. Research on the tumor microenvironment (TME) of PDAC has propelled the development of immunotherapeutic and targeted therapeutic strategies with a promising future. The emergence of single-cell sequencing and mass spectrometry technologies, coupled with spatial omics, has collectively revealed the heterogeneity of the TME from a multiomics perspective, outlined the development trajectories of cell lineages, and revealed important functions of previously underrated myeloid cells and tumor stroma cells. Concurrently, these findings necessitated more refined annotations of biological functions at the cell cluster or single-cell level. Precise identification of all cell clusters is urgently needed to determine whether they have been investigated adequately and to identify target cell clusters with antitumor potential, design compatible treatment strategies, and determine treatment resistance. Here, we summarize recent research on the PDAC TME at the single-cell multiomics level, with an unbiased focus on the functions and potential classification bases of every cellular component within the TME, and look forward to the prospects of integrating single-cell multiomics data and retrospectively reusing bulk sequencing data, hoping to provide new insights into the PDAC TME.


Sujet(s)
Tumeurs du pancréas , Analyse sur cellule unique , Microenvironnement tumoral , Humains , Microenvironnement tumoral/génétique , Analyse sur cellule unique/méthodes , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Animaux , Marqueurs biologiques tumoraux , Génomique/méthodes , Régulation de l'expression des gènes tumoraux , Multi-omique
18.
J Exp Clin Cancer Res ; 43(1): 189, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978141

RÉSUMÉ

BACKGROUND: Distinguishing benign from malignant pancreaticobiliary disease is challenging because of the absence of reliable biomarkers. Circulating extracellular vesicles (EVs) have emerged as functional mediators between cells. Their cargos, including microRNAs (miRNAs), are increasingly acknowledged as an important source of potential biomarkers. This multicentric, prospective study aimed to establish a diagnostic plasma EV-derived miRNA signature to discriminate pancreatic ductal adenocarcinoma (PDAC) from benign pancreaticobiliary disease. METHODS: Plasma EVs were isolated using size exclusion chromatography (SEC) and characterised using nanoparticle tracking analysis, electron microscopy and Western blotting. EV-RNAs underwent small RNA sequencing to discover differentially expressed markers for PDAC (n = 10 benign vs. 10 PDAC). Candidate EV-miRNAs were then validated in a cohort of 61 patients (n = 31 benign vs. 30 PDAC) by RT-qPCR. Logistic regression and optimal thresholds (Youden Index) were used to develop an EV-miR-200 family model to detect cancer. This model was tested in an independent cohort of 95 patients (n = 30 benign, 33 PDAC, and 32 cholangiocarcinoma). RESULTS: Small RNA sequencing and RT-qPCR showed that EV-miR-200 family members were significantly overexpressed in PDAC vs. benign disease. Combined expression of the EV-miR-200 family showed an AUC of 0.823. In an independent validation cohort, application of this model showed a sensitivity, specificity and AUC of 100%, 88%, and 0.97, respectively, for diagnosing PDAC. CONCLUSIONS: This is the first study to validate plasma EV-miR-200 members as a clinically-useful diagnostic biomarker for PDAC. Further validation in larger cohorts and clinical trials is essential. These findings also suggest the potential utility in monitoring response and/or recurrence.


Sujet(s)
Carcinome du canal pancréatique , Vésicules extracellulaires , microARN , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/sang , Carcinome du canal pancréatique/diagnostic , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/génétique , microARN/sang , microARN/génétique , Femelle , Mâle , Adulte d'âge moyen , Tumeurs du pancréas/sang , Tumeurs du pancréas/diagnostic , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Sujet âgé , Marqueurs biologiques tumoraux/sang , Études prospectives
19.
Nat Commun ; 15(1): 6043, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025845

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a devastating cancer with dismal prognosis due to distant metastasis, even in the early stage. Using RNA sequencing and multiplex immunofluorescence, here we find elevated expression of mixed lineage kinase domain-like pseudo-kinase (MLKL) and enhanced necroptosis pathway in PDAC from early liver metastasis T-stage (T1M1) patients comparing with non-metastatic (T1M0) patients. Mechanistically, MLKL-driven necroptosis recruits macrophages, enhances the tumor CD47 'don't eat me' signal, and induces macrophage extracellular traps (MET) formation for CXCL8 activation. CXCL8 further initiates epithelial-mesenchymal transition (EMT) and upregulates ICAM-1 expression to promote endothelial adhesion. METs also degrades extracellular matrix, that eventually supports PDAC liver metastasis. Meanwhile, targeting necroptosis and CD47 reduces liver metastasis in vivo. Our study thus reveals that necroptosis facilitates PDAC metastasis by evading immune surveillance, and also suggest that CD47 blockade, combined with MLKL inhibitor GW806742X, may be a promising neoadjuvant immunotherapy for overcoming the T1M1 dilemma and reviving the opportunity for radical surgery.


Sujet(s)
Antigènes CD47 , Carcinome du canal pancréatique , Transition épithélio-mésenchymateuse , Pièges extracellulaires , Tumeurs du foie , Macrophages , Nécroptose , Tumeurs du pancréas , Protein kinases , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du foie/secondaire , Tumeurs du foie/métabolisme , Animaux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/génétique , Souris , Macrophages/métabolisme , Macrophages/immunologie , Lignée cellulaire tumorale , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Protein kinases/métabolisme , Pièges extracellulaires/métabolisme , Molécule-1 d'adhérence intercellulaire/métabolisme , Molécule-1 d'adhérence intercellulaire/génétique , Mâle , Transduction du signal , Femelle , Acrylamides , Sulfonamides
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE