Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.849
Filtrer
1.
PeerJ ; 12: e17797, 2024.
Article de Anglais | MEDLINE | ID: mdl-39221276

RÉSUMÉ

Numerous aspects of cellular signaling are regulated by the kinome-the network of over 500 protein kinases that guides and modulates information transfer throughout the cell. The key role played by both individual kinases and assemblies of kinases organized into functional subnetworks leads to kinome dysregulation driving many diseases, particularly cancer. In the case of pancreatic ductal adenocarcinoma (PDAC), a variety of kinases and associated signaling pathways have been identified for their key role in the establishment of disease as well as its progression. However, the identification of additional relevant therapeutic targets has been slow and is further confounded by interactions between the tumor and the surrounding tumor microenvironment. In this work, we attempt to link the state of the human kinome, or kinotype, with cell viability in treated, patient-derived PDAC tumor and cancer-associated fibroblast cell lines. We applied classification models to independent kinome perturbation and kinase inhibitor cell screen data, and found that the inferred kinotype of a cell has a significant and predictive relationship with cell viability. We further find that models are able to identify a set of kinases whose behavior in response to perturbation drive the majority of viability responses in these cell lines, including the understudied kinases CSNK2A1/3, CAMKK2, and PIP4K2C. We next utilized these models to predict the response of new, clinical kinase inhibitors that were not present in the initial dataset for model devlopment and conducted a validation screen that confirmed the accuracy of the models. These results suggest that characterizing the perturbed state of the human protein kinome provides significant opportunity for better understanding of signaling behavior and downstream cell phenotypes, as well as providing insight into the broader design of potential therapeutic strategies for PDAC.


Sujet(s)
Fibroblastes associés au cancer , Carcinome du canal pancréatique , Survie cellulaire , Tumeurs du pancréas , Protein kinases , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/enzymologie , Survie cellulaire/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/anatomopathologie , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/enzymologie , Lignée cellulaire tumorale , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/enzymologie , Protein kinases/métabolisme , Transduction du signal , Microenvironnement tumoral , Inhibiteurs de protéines kinases/pharmacologie
2.
J Pathol ; 263(4-5): 466-481, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38924548

RÉSUMÉ

The E3 ubiquitin ligase thyroid hormone receptor interacting protein 12 (TRIP12) has been implicated in pancreatic adenocarcinoma (PDAC) through its role in mediating the degradation of pancreas transcription factor 1a (PTF1a). PTF1a is a transcription factor essential for the acinar differentiation state that is notably diminished during the early steps of pancreatic carcinogenesis. Despite these findings, the direct involvement of TRIP12 in the onset of pancreatic cancer has yet to be established. In this study, we demonstrated that TRIP12 protein was significantly upregulated in human pancreatic preneoplastic lesions. Furthermore, we observed that TRIP12 overexpression varied within PDAC samples and PDAC-derived cell lines. We further demonstrated that TRIP12 was required for PDAC-derived cell growth and for the expression of E2F-targeted genes. Acinar-to-ductal cell metaplasia (ADM) is a reversible process that reflects the high plasticity of acinar cells. ADM becomes irreversible in the presence of oncogenic Kras mutations and leads to the formation of preneoplastic lesions. Using two genetically modified mouse models, we showed that a loss of TRIP12 prevented acini from developing ADM in response to pancreatic injury. With two additional mouse models, we further discovered that a depletion of TRIP12 prevented the formation of KrasG12D-induced preneoplastic lesions and impaired metastasis formation in the presence of mutated KrasG12D and Trp53R172H genes. In summary our study identified an overexpression of TRIP12 from the early stages of pancreatic carcinogenesis and proposed this E3 ubiquitin ligase as a novel regulator of acinar plasticity with an important dual role in initiation and metastatic steps of PDAC. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Sujet(s)
Cellules acineuses , Carcinome du canal pancréatique , Tumeurs du pancréas , Ubiquitin-protein ligases , Animaux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/enzymologie , Humains , Cellules acineuses/anatomopathologie , Cellules acineuses/métabolisme , Cellules acineuses/enzymologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/enzymologie , Métaplasie/anatomopathologie , Métaplasie/métabolisme , Plasticité cellulaire , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Souris , Lignée cellulaire tumorale , Prolifération cellulaire , Souris knockout , Régulation de l'expression des gènes tumoraux , États précancéreux/anatomopathologie , États précancéreux/génétique , États précancéreux/métabolisme , États précancéreux/enzymologie , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Transformation cellulaire néoplasique/métabolisme , Protéines de transport
3.
Nat Cell Biol ; 26(6): 975-990, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38862786

RÉSUMÉ

Identifying the adaptive mechanisms of metastatic cancer cells remains an elusive question in the treatment of metastatic disease, particularly in pancreatic cancer (pancreatic adenocarcinoma, PDA). A loss-of-function shRNA targeted screen in metastatic-derived cells identified Gstt1, a member of the glutathione S-transferase superfamily, as uniquely required for dissemination and metastasis, but dispensable for primary tumour growth. Gstt1 is expressed in latent disseminated tumour cells (DTCs), is retained within a subpopulation of slow-cycling cells within existing metastases, and its inhibition leads to complete regression of macrometastatic tumours. This distinct Gstt1high population is highly metastatic and retains slow-cycling phenotypes, epithelial-mesenchymal transition features and DTC characteristics compared to the Gstt1low population. Mechanistic studies indicate that in this subset of cancer cells, Gstt1 maintains metastases by binding and glutathione-modifying intracellular fibronectin, in turn promoting its secretion and deposition into the metastatic microenvironment. We identified Gstt1 as a mediator of metastasis, highlighting the importance of heterogeneity and its influence on the metastatic tumour microenvironment.


Sujet(s)
Glutathione transferase , Tumeurs du pancréas , Microenvironnement tumoral , Glutathione transferase/métabolisme , Glutathione transferase/génétique , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/métabolisme , Animaux , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse , Fibronectines/métabolisme , Métastase tumorale , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Adénocarcinome/métabolisme , Adénocarcinome/enzymologie , Survie cellulaire , Régulation de l'expression des gènes tumoraux , Souris , Femelle , Souris de lignée C57BL
4.
Cancer Lett ; 596: 216993, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38801884

RÉSUMÉ

Ribonucleotide Reductase (RNR) is a rate-limiting enzyme in the production of deoxyribonucleoside triphosphates (dNTPs), which are essential substrates for DNA repair after radiation damage. We explored the radiosensitization property of RNR and investigated a selective RRM2 inhibitor, 3-AP, as a radiosensitizer in the treatment of metastatic pNETs. We investigated the role of RNR subunit, RRM2, in pancreatic neuroendocrine (pNET) cells and responses to radiation in vitro. We also evaluated the selective RRM2 subunit inhibitor, 3-AP, as a radiosensitizer to treat pNET metastases in vivo. Knockdown of RNR subunits demonstrated that RRM1 and RRM2 subunits, but not p53R3, play significant roles in cell proliferation. RRM2 inhibition activated DDR pathways through phosphorylation of ATM and DNA-PK protein kinases but not ATR. RRM2 inhibition also induced Chk1 and Chk2 phosphorylation, resulting in G1/S phase cell cycle arrest. RRM2 inhibition sensitized pNET cells to radiotherapy and induced apoptosis in vitro. In vivo, we utilized pNET subcutaneous and lung metastasis models to examine the rationale for RNR-targeted therapy and 3-AP as a radiosensitizer in treating pNETs. Combination treatment significantly increased apoptosis of BON (human pNET) xenografts and significantly reduced the burden of lung metastases. Together, our results demonstrate that selective RRM2 inhibition induced radiosensitivity of metastatic pNETs both in vitro and in vivo. Therefore, treatment with the selective RRM2 inhibitor, 3-AP, is a promising radiosensitizer in the therapeutic armamentarium for metastatic pNETs.


Sujet(s)
Apoptose , Prolifération cellulaire , Souris nude , Tumeurs du pancréas , Radiotolérance , Radiosensibilisants , Ribonucleoside diphosphate reductase , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/radiothérapie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/enzymologie , Ribonucleoside diphosphate reductase/génétique , Ribonucleoside diphosphate reductase/antagonistes et inhibiteurs , Ribonucleoside diphosphate reductase/métabolisme , Animaux , Lignée cellulaire tumorale , Radiosensibilisants/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Radiotolérance/effets des médicaments et des substances chimiques , Phosphorylation , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/radiothérapie , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/enzymologie , Tumeurs neuroendocrines/métabolisme , Tumeurs du poumon/secondaire , Tumeurs du poumon/radiothérapie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/génétique , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/génétique , Souris , Checkpoint kinase 2/métabolisme , Checkpoint kinase 2/génétique , Checkpoint kinase 2/antagonistes et inhibiteurs , Femelle , Interférence par ARN , DNA-activated protein kinase
5.
Mol Oncol ; 18(8): 2020-2041, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38650175

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a limited number of known driver mutations but considerable cancer cell heterogeneity. Phosphoproteomics provides a direct read-out of aberrant signaling and the resultant clinically relevant phenotype. Mass spectrometry (MS)-based proteomics and phosphoproteomics were applied to 42 PDAC tumors. Data encompassed over 19 936 phosphoserine or phosphothreonine (pS/T; in 5412 phosphoproteins) and 1208 phosphotyrosine (pY; in 501 phosphoproteins) sites and a total of 3756 proteins. Proteome data identified three distinct subtypes with tumor intrinsic and stromal features. Subsequently, three phospho-subtypes were apparent: two tumor intrinsic (Phos1/2) and one stromal (Phos3), resembling known PDAC molecular subtypes. Kinase activity was analyzed by the Integrative iNferred Kinase Activity (INKA) scoring. Phospho-subtypes displayed differential phosphorylation signals and kinase activity, such as FGR and GSK3 activation in Phos1, SRC kinase family and EPHA2 in Phos2, and EGFR, INSR, MET, ABL1, HIPK1, JAK, and PRKCD in Phos3. Kinase activity analysis of an external PDAC cohort supported our findings and underscored the importance of PI3K/AKT and ERK pathways, among others. Interestingly, unfavorable patient prognosis correlated with higher RTK, PAK2, STK10, and CDK7 activity and high proliferation, whereas long survival was associated with MYLK and PTK6 activity, which was previously unknown. Subtype-associated activity profiles can guide therapeutic combination approaches in tumor and stroma-enriched tissues, and emphasize the critical role of parallel signaling pathways. In addition, kinase activity profiling identifies potential disease markers with prognostic significance.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/enzymologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Pronostic , Femelle , Mâle , Phosphorylation , Adulte d'âge moyen , Protéomique , Lignée cellulaire tumorale , Sujet âgé
6.
Hum Mol Genet ; 33(11): 1001-1014, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38483348

RÉSUMÉ

The CEL gene encodes carboxyl ester lipase, a pancreatic digestive enzyme. CEL is extremely polymorphic due to a variable number tandem repeat (VNTR) located in the last exon. Single-base deletions within this VNTR cause the inherited disorder MODY8, whereas little is known about VNTR single-base insertions in pancreatic disease. We therefore mapped CEL insertion variants (CEL-INS) in 200 Norwegian patients with pancreatic neoplastic disorders. Twenty-eight samples (14.0%) carried CEL-INS alleles. Most common were insertions in repeat 9 (9.5%), which always associated with a VNTR length of 13 repeats. The combined INS allele frequency (0.078) was similar to that observed in a control material of 416 subjects (0.075). We performed functional testing in HEK293T cells of a set of CEL-INS variants, in which the insertion site varied from the first to the 12th VNTR repeat. Lipase activity showed little difference among the variants. However, CEL-INS variants with insertions occurring in the most proximal repeats led to protein aggregation and endoplasmic reticulum stress, which upregulated the unfolded protein response. Moreover, by using a CEL-INS-specific antibody, we observed patchy signals in pancreatic tissue from humans without any CEL-INS variant in the germline. Similar pancreatic staining was seen in knock-in mice expressing the most common human CEL VNTR with 16 repeats. CEL-INS proteins may therefore be constantly produced from somatic events in the normal pancreatic parenchyma. This observation along with the high population frequency of CEL-INS alleles strongly suggests that these variants are benign, with a possible exception for insertions in VNTR repeats 1-4.


Sujet(s)
Répétitions minisatellites , Pancréas exocrine , Humains , Répétitions minisatellites/génétique , Animaux , Souris , Pancréas exocrine/métabolisme , Pancréas exocrine/enzymologie , Cellules HEK293 , Mutagenèse par insertion/génétique , Allèles , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/enzymologie , Fréquence d'allèle , Mâle , Femelle , Triacylglycerol lipase/génétique
7.
J Enzyme Inhib Med Chem ; 38(1): 2166039, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-36683274

RÉSUMÉ

Inhibiting a specific target in cancer cells and reducing unwanted side effects has become a promising strategy in pancreatic cancer treatment. MAP4K4 is associated with pancreatic cancer development and correlates with poor clinical outcomes. By phosphorylating MKK4, proteins associated with cell apoptosis and survival are translated. Therefore, inhibiting MAP4K4 activity in pancreatic tumours is a new therapeutic strategy. Herein, we performed a structure-based virtual screening to identify MAP4K4 inhibitors and discovered the compound F389-0746 with a potent inhibition (IC50 120.7 nM). The results of kinase profiling revealed that F389-0746 was highly selective to MAP4K4 and less likely to cause side effects. Results of in vitro experiments showed that F389-0746 significantly suppressed cancer cell growth and viability. Results of in vivo experiments showed that F389-0746 displayed comparable tumour growth inhibition with the group treated with gemcitabine. These findings suggest that F389-0746 has promising potential to be further developed as a novel pancreatic cancer treatment.


Sujet(s)
Antinéoplasiques , Tumeurs du pancréas , Inhibiteurs de protéines kinases , Protein-Serine-Threonine Kinases , Humains , Lignée cellulaire tumorale , Gemcitabine/composition chimique , Gemcitabine/pharmacologie , Protéines et peptides de signalisation intracellulaire , Tumeurs du pancréas/enzymologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Simulation numérique , Tumeurs du pancréas
8.
Pancreatology ; 22(5): 619-625, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35550115

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is notorious for high mortality due to limited options of appropriate chemotherapy drugs. Here we report that Aurora kinase-A expression is elevated in both human and mouse PDAC samples. MLN8237, an inhibitor of Aurora kinase-A, efficiently reduced the proliferation and motility of PDAC cells in vitro as well as tumor growth in orthotropic xenograft model and genetic pancreatic cancer animal models (p53/LSL/Pdx-Cre mice) in vivo. MLN8237 exhibited tumor inhibitory effect through inhibiting proliferation and migration, and inducing apoptosis and senescence. These results provide the molecular basis for a novel chemotherapy strategy for PDAC patients.


Sujet(s)
Aurora kinase A , Azépines , Carcinome du canal pancréatique , Tumeurs du pancréas , Pyrimidines , Animaux , Apoptose/effets des médicaments et des substances chimiques , Aurora kinase A/antagonistes et inhibiteurs , Aurora kinase A/génétique , Aurora kinase A/métabolisme , Azépines/pharmacologie , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/enzymologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Souris , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines/pharmacologie , Tumeurs du pancréas
9.
Oncogene ; 41(20): 2860-2872, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35422475

RÉSUMÉ

RNA N6-methyladenosine (m6A) is an emerging regulator of mRNA modifications and represents a novel player in tumorigenesis. Although it has functional significance in both pathological and physiological processes, the role of m6A modification in pancreatic ductal cancer (PDAC) remains elusive. Here, we showed that high fat mass and obesity-associated gene (FTO) expression was associated with a poor prognosis in PDAC patients and that suppression of FTO expression inhibited cell proliferation. Here, m6A sequencing (m6A-seq) was performed to screen genes targeted by FTO. The effects of FTO stimulation on the biological characteristics of pancreatic cancer cells, including proliferation and colony formation, were investigated in vitro and in vivo. The results indicate that FTO directly targets platelet-derived growth factor C (PDGFC) and stabilizes its mRNA expression in an m6A-YTHDF2-dependent manner. m6A-methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR), RNA immunoprecipitation (RIP), and luciferase reporter assays were employed to validate the specific binding of FTO to PDGFC. PDGFC upregulation led to reactivation of the Akt signaling pathway, promoting cell growth. Overall, our study reveals that FTO downregulation leads to increased m6A modifications in the 3' UTR of PDGFC and then modulates the degradation of its transcriptional level in an m6A-YTHDF2-dependent manner, highlighting a potential therapeutic target for PDAC treatment and prognostic prediction.


Sujet(s)
Alpha-ketoglutarate-dependent dioxygenase FTO , Tumeurs du pancréas , Facteur de croissance dérivé des plaquettes , Protéines de liaison à l'ARN , Adénosine/génétique , Adénosine/métabolisme , Alpha-ketoglutarate-dependent dioxygenase FTO/génétique , Alpha-ketoglutarate-dependent dioxygenase FTO/métabolisme , Humains , Lymphokines , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Facteur de croissance dérivé des plaquettes/génétique , Facteur de croissance dérivé des plaquettes/métabolisme , ARN , ARN messager/génétique , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Tumeurs du pancréas
10.
Anticancer Res ; 42(3): 1207-1215, 2022 Mar.
Article de Anglais | MEDLINE | ID: mdl-35220210

RÉSUMÉ

BACKGROUND: We aimed to clarify the role of complement C3a and its receptor C3aR in progression of pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS: We evaluated the serum levels of C3 and C3a in patients with PDAC. C3aR expression in tissue was assessed using a tissue microarray. To confirm the protumoral effects of C3a in PDAC, we conducted in vitro experiments using PDAC cell lines (Panc-1 and MiaPaca-2) that exhibit high C3aR expression. RESULTS: Serum levels of both C3 and C3a were higher in 26 patients with PDAC than in 28 nontumor-bearing controls. In the tissue microarray, we observed increased expression of C3aR in PDAC cells, especially in cases with metastatic lesions. In vitro experiments showed that C3a facilitated tumor cell proliferation, migration and invasion by activating the extracellular-regulated kinase signaling pathway and inducing epithelial-to-mesenchymal transition. Inhibition of the C3a-C3aR axis by pharmacological blockade and short-hairpin RNA-mediated knockdown of C3aR alleviated its protumoral effect. CONCLUSION: These findings provide a new approach for the development of treatments targeting the C3a-C3aR axis.


Sujet(s)
Carcinome du canal pancréatique/enzymologie , Complément C3/métabolisme , Transition épithélio-mésenchymateuse , Extracellular Signal-Regulated MAP Kinases/métabolisme , Tumeurs du pancréas/enzymologie , Récepteurs au complément/métabolisme , Sujet âgé , Sujet âgé de 80 ans ou plus , Arginine/analogues et dérivés , Arginine/pharmacologie , Composés benzhydryliques/pharmacologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Inhibiteurs du complément/pharmacologie , Activation enzymatique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Adulte d'âge moyen , Invasion tumorale , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Récepteurs au complément/antagonistes et inhibiteurs , Récepteurs au complément/génétique , Transduction du signal
11.
Mol Biol Rep ; 49(5): 3713-3720, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-35129766

RÉSUMÉ

BACKGROUND: Pancreatic ductal adenocarcinomas (PDACs) is a malignant disorder and is the most common pancreatic cancer type. The malignant cells depend on the uptake of asparagine (Asn) for growth. The synthesis of Asn occurs through the enzyme asparagine synthetase (ASNS). Interestingly, ASNS is known as is direct target of nonsense-mediated RNA decay (NMD). We have previously reported that NMD major factor UPF1 mutations in the pancreatic tumors. However, the relationship between NMD and the level of ASNS is unknown. METHOD: We constructed point mutations by site-specific mutagenesis. To evaluate NMD magnitude, we assessed the expression ratio of an exogenously expressed wild-type and mutated ß-globin mRNA with N39 allele, and five known NMD targets. Then, reverse transcription-polymerase chain reaction (RT-PCR), RT-qPCR and western bolt to determine RNA or protein levels, after knockdown of endogenous UPF1 by small RNA interference in the cells. RESULTS: An RNA editing event (c.3101 A > G) at UPF1 transcripts resulting in an Asparagine (p.1034) changed to a Serine is found in one primary PDAC patient. The edited UPF1 increases the ability of degrading of NMD provoking transcripts, such as ß-globin mRNA with N39 allele and 5 out of 5 known endogenous NMD substrate mRNAs, including ASNS. In addition, ASNS mRNA is subjected to NMD degradation by virtue of its possessing uORFs at the 5'UTR. A reduction of endogenous ASNS RNA and the increased protein expression level is found either in the PDAC patient or in the cells with edited UPF1 at c.3101 A > G relative to the controls. CONCLUSIONS: This edited UPF1 found in the PDAC results in hyperactivated NMD, which is tightly correlation to elevated expression level of ASNS. The targeting of knockdown of ASNS may improve the antitumor potency in PDACs.


Sujet(s)
Aspartate-ammonia ligase , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor , Tumeurs du pancréas , Transactivateurs , Asparagine/génétique , Asparagine/métabolisme , Aspartate-ammonia ligase/génétique , Aspartate-ammonia ligase/métabolisme , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/génétique , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/métabolisme , Carcinome du canal pancréatique/enzymologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Humains , Mutagenèse dirigée , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , RNA helicases/génétique , RNA helicases/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Transactivateurs/génétique , Transactivateurs/métabolisme , Globines bêta/métabolisme
12.
Am J Surg Pathol ; 46(1): 97-104, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34889867

RÉSUMÉ

Carboxypeptidase A1 (CPA1) is a zinc metalloprotease that is produced in pancreatic acinar cells and plays a role in cleaving C-terminal branched-chain and aromatic amino acids from dietary proteins. This study assessed the utility of immunohistochemical CPA1 staining for diagnosing pancreatic acinar cell carcinoma (ACC). A total of 12,274 tumor samples from 132 different tumor types and subtypes as well as 8 samples each of 76 different normal tissue types were interpretable by immunohistochemistry in a tissue microarray format. CPA1 was strongly expressed in acinar cells of all normal pancreas samples but not in any other normal tissues. CPA1 immunostaining was detected in 100% of 11 pancreatic ACCs and 1 mixed acinar endocrine carcinoma, but absent in 449 pancreatic ductal adenocarcinomas, 75 adenocarcinomas of the ampulla Vateri, and 11,739 other evaluable cancers from 128 different tumor entities. A weak to moderate diffuse staining of epithelial and stromal cells of cancer tissues immediately adjacent to non-neoplastic pancreatic acinar cells often occurred and was considered to be caused by the diffusion of the highly abundant CPA1 from normal acinar cells that may have suffered some autolytic cell damage. In conclusion, our data show that CPA1 is a highly sensitive and largely specific marker for normal and neoplastic pancreatic acinar cells. CPA1 immunohistochemistry greatly facilitates the otherwise often difficult diagnosis of pancreatic ACC.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Carboxypeptidases A/analyse , Carcinome à cellules acineuses/enzymologie , Immunohistochimie , Tumeurs du pancréas/enzymologie , Carcinome à cellules acineuses/anatomopathologie , Allemagne , Humains , Tumeurs du pancréas/anatomopathologie , Valeur prédictive des tests , Reproductibilité des résultats , Analyse sur puce à tissus
13.
Cancer Lett ; 526: 53-65, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-34813892

RÉSUMÉ

Carboxy-terminal domain (CTD) small phosphatase like 2 (CTDSPL2), also known as SCP4 or HSPC129, is a new member of the small CTD phosphatase (SCP) family and its role in cancers remains unclear. Here, we used a Phos-tag technique to screen a series of phosphatases and identified CTDSPL2 as a mitotic regulator. We demonstrated that CTDSPL2 was phosphorylated at T86, S104, and S134 by cyclin-dependent kinase 1 (CDK1) in mitosis. Depletion of CTDSPL2 led to mitotic defects and prolonged mitosis. Resultantly, CTDSPL2 deletion restrained proliferation, migration, and invasion in pancreatic cancer cells. We further confirmed the dominant negative effects of a phosphorylation-deficient mutant form of CTDSPL2, implying the biological significance of CTDSPL2 mitotic phosphorylation. Moreover, RT2 cell cycle array analysis revealed p21 and p27 as downstream regulators of CTDSPL2, and inhibition of p21 and/or p27 partially rescued the phenotype in CTDSPL2-deficient cell lines. Importantly, both CTDSPL2 depletion and phosphorylation-deficient mutant CTDSPL2 hindered tumor growth in xenograft models. Together, our findings for the first time highlight the novel role of CTDSPL2 in regulating cell mitosis, proliferation and motility in pancreatic cancer and point out the implications of CTDSPL2 in regulating two critical cell cycle participants (p21 and p27), providing an alternative molecular target for pancreatic cancer treatment.


Sujet(s)
Tumeurs du pancréas/enzymologie , Phosphoprotein Phosphatases/métabolisme , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire/physiologie , Cellules HEK293 , Cellules HeLa , Hétérogreffes , Humains , Mâle , Souris , Souris nude , Mitose/physiologie , Tumeurs du pancréas/anatomopathologie , Phosphorylation
14.
Diagn Pathol ; 16(1): 117, 2021 Dec 13.
Article de Anglais | MEDLINE | ID: mdl-34895278

RÉSUMÉ

BACKGROUND: Pancreatic medullary carcinoma (PMC) is a rare pancreatic tumor, usually showing the presence of microsatellite instability, mostly MLH1 silencing, and a wild-type KRAS mutation status. We report here a PMC arising from a Pancreatic Intraductal Papillary Mucinous Neoplasm (IPMN), both having KRAS and TP53 mutations. CASE PRESENTATION: We report the case of a 73-year-old woman presenting with right iliac fossa pain. MRI revealed a 16 mm diameter mass in the pancreas, leading to a pancreatic duct stricture and upstream a dilatation of the distal pancreatic duct of Wirsung. A fine needle aspiration was performed, and pathology analysis revealed malignant glandular cells. The patient underwent distal pancreatectomy. Gross examination revealed an12 mm indurated white lesion, adjacent to a cystic lesion extending into the rest of the pancreatic body. Microscopically, the cystic area represented a mixed (gastric-type and pancreatobiliary-type) IPMN, involving the main and secondary pancreatic ducts with low-grade and high-grade dysplasia. In the periphery of this IPMN, a 14mm associated invasive carcinoma was observed, characterized by focal gland formation and by poorly differentiated cells with a syncytial appearance, associated with a dense lymphoplasmocytic and neutrophilic infiltrate. Immunohistochemical analyses showed loss of MSH2 and MSH6 expression. Microsatellite instability was confirmed by molecular test. Molecular analysis was performed both on the invasive carcinoma and on the high-grade dysplasia IPMN, revealing the same mutation profile with KRAS and TP53 mutations. The proposed diagnosis was mixed IPMN with associated invasive medullary carcinoma that presented loss of MSH2 and MSH6 expression. CONCLUSIONS: The present case reports for the first time, at the best of our knowledge, the coexistence of IPMN lesions and PMC, both having the same molecular alterations. It also describes the second case of PMC with microsatellite instability, MSH2 and MSH6 silenced.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Carcinome médullaire/enzymologie , Protéines de liaison à l'ADN/analyse , Protéine-2 homologue de MutS/analyse , Tumeurs intracanalaires pancréatiques/enzymologie , Tumeurs du pancréas/enzymologie , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Carcinome médullaire/génétique , Carcinome médullaire/anatomopathologie , Carcinome médullaire/chirurgie , Régulation négative , Femelle , Humains , Instabilité des microsatellites , Mutation , Pancréatectomie , Tumeurs intracanalaires pancréatiques/génétique , Tumeurs intracanalaires pancréatiques/anatomopathologie , Tumeurs intracanalaires pancréatiques/chirurgie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/chirurgie , Protéines proto-oncogènes p21(ras)/génétique , Protéine p53 suppresseur de tumeur/génétique
15.
J Cancer Res Ther ; 17(6): 1419-1424, 2021.
Article de Anglais | MEDLINE | ID: mdl-34916372

RÉSUMÉ

BACKGROUND: Pancreatic cancer is the second type of cancer that causes the most death among the digestive system cancers. Difficulties in early diagnosis and rapidly progressing to advanced stages are most common in high mortality rate of pancreatic carcinoma. The mutation of Bcr-Abl tyrosine kinase and mitotic kinases (such as Aurora kinases), which are involved in the cell cycle, plays an important role in the progression of cancer. Enzymes belonging to Aurora kinase family (-A, -B, -C) have been reported to play a major role in cancer progression, invasion and metastasis. Therefore, the purpose of this study, investigate of the effect of danusertib, an Aurora kinase inhibitor, onto cytotoxicity, apoptosis and cell cycle in human pancreatic carcinoma CFPAC-1 cells. MATERIALS AND METHODS: For determining the IC50 value, the 20,000 cells were seeded in E-plate 16 wells in a real-time cell analyzer and various concentrations of danusertib (1-10,000 nM) were applied onto CFPAC-1 cells incubated in IMDM medium. Cell index demonstrated that the proliferation of fraction cells was measured in real time. On the other hand, cell apoptosis and cell cycle arrest test were stained with Annexin V-APC/PI and DNA-cell cycle PI staining respectively by using flow cytometry. RESULTS: The IC50 value was found to be approximately 400 nM. Danusertib at this concentration induced apoptosis in CFPAC-1 cells (%14,8 at 24 hours; %21,3 at 48 hours). Furthermore, in the cells treated with danusertib, 31.77% and 11.05% were arrested in the S and G2 phases, respectively. CONCLUSIONS: Aurora kinase inhibitor danusertib induced a significant effect of cytotoxic, apoptotic and cell cycle arrest in CFPAC-1 ductal adenocarcinoma cells. Therefore, it may be a potential alternative to the treatment of pancreatic cancers.


Sujet(s)
Aurora kinases/antagonistes et inhibiteurs , Benzamides/pharmacologie , Carcinome du canal pancréatique/anatomopathologie , Points de contrôle du cycle cellulaire , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs du pancréas/anatomopathologie , Pyrazoles/pharmacologie , Apoptose , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/enzymologie , Prolifération cellulaire , Humains , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/enzymologie , Cellules cancéreuses en culture
16.
Cell Death Dis ; 12(10): 918, 2021 10 07.
Article de Anglais | MEDLINE | ID: mdl-34620839

RÉSUMÉ

Pancreatic cancer is the third leading cause of cancer-related mortalities and is characterized by rapid disease progression. Identification of novel therapeutic targets for this devastating disease is important. Phosphoenolpyruvate carboxykinase 1 (PCK1) is the rate-limiting enzyme of gluconeogenesis. The current study tested the expression and potential functions of PCK1 in pancreatic cancer. We show that PCK1 mRNA and protein levels are significantly elevated in human pancreatic cancer tissues and cells. In established and primary pancreatic cancer cells, PCK1 silencing (by shRNA) or CRISPR/Cas9-induced PCK1 knockout potently inhibited cell growth, proliferation, migration and invasion, and induced robust apoptosis activation. Conversely, ectopic overexpression of PCK1 in pancreatic cancer cells accelerated cell proliferation and migration. RNA-seq analyzing of differentially expressed genes (DEGs) in PCK1-silenced pancreatic cancer cells implied that DEGs were enriched in the PI3K-Akt-mTOR cascade. In pancreatic cancer cells, Akt-mTOR activation was largely inhibited by PCK1 shRNA, but was augmented after ectopic PCK1 overexpression. In vivo, the growth of PCK1 shRNA-bearing PANC-1 xenografts was largely inhibited in nude mice. Akt-mTOR activation was suppressed in PCK1 shRNA-expressing PANC-1 xenograft tissues. Collectively, PCK1 is a potential therapeutic target for pancreatic cancer.


Sujet(s)
Thérapie moléculaire ciblée , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/enzymologie , Phosphoenolpyruvate carboxykinase (ATP)/métabolisme , Adulte , Sujet âgé , Animaux , Apoptose/génétique , Systèmes CRISPR-Cas/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Études de cohortes , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Extinction de l'expression des gènes , Humains , Mâle , Souris de lignée BALB C , Souris nude , Adulte d'âge moyen , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Phosphoenolpyruvate carboxykinase (ATP)/génétique , ARN messager/génétique , ARN messager/métabolisme , Transduction du signal , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Biomed Res Int ; 2021: 6673125, 2021.
Article de Anglais | MEDLINE | ID: mdl-34595239

RÉSUMÉ

BACKGROUND: Pancreatic cancer (PC) is one of the most common cancers worldwide, with high mortality. The UGT1A gene family plays important roles in pharmacology and toxicology, contributing to interindividual differences in drug disposition. However, mRNA expression and prognostic value of the UGT1A gene family in PC have not been identified. METHODS: Oncomine, GEPIA2, DAVID 6.8, Metascape, Kaplan-Meier plotter, cBioPortal, GeneMANIA, TRRUST v2, TIMER, and R software were used in our study. RESULTS: The transcriptional levels of UGT1A1/3/6/8/9/10 in PC tissues were significantly higher than those in normal tissues. These results were further validated using five pairs of PC tumor tissues and adjacent nontumor tissues. A significant correlation was found between the expression of UGT1A1/6/10 and the pathological stage of PC. PC patients with lower transcriptional levels of UGT1A1/4/5/6/10 were associated with a better prognosis. The differentially expressed UGT1A gene family functions were primarily related to the glucuronidation pathway, cytokine-cytokine receptor interactions, and the ILK signaling pathway. Our data suggest that HNF1A, AHR, and CDX2 are key transcription factors for the UGT1A gene family. Furthermore, the expression levels of UGT1A1/3/8/9/10 were positively correlated with the activities of tumor-infiltrating immune cells, especially B cells. The expression levels of UGT1A6/9 were negatively correlated with macrophage infiltration levels. CONCLUSIONS: These results suggest that the UGT1A gene family could serve as a potential prognostic biomarker and target for PC. However, future studies are required to validate our findings and promote the clinical utility of the UGT1A gene family in PC.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Glucuronosyltransferase/génétique , Famille multigénique , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Marqueurs biologiques tumoraux/métabolisme , Régulation de l'expression des gènes codant pour des enzymes , Régulation de l'expression des gènes tumoraux , Glucuronosyltransferase/métabolisme , Humains , Estimation de Kaplan-Meier , Lymphocytes TIL/immunologie , Tumeurs du pancréas/immunologie , Pronostic , Liaison aux protéines , Cartes d'interactions protéiques/génétique , ARN messager/génétique , ARN messager/métabolisme , Reproductibilité des résultats , Transcription génétique
18.
Biomed Pharmacother ; 144: 112325, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34656065

RÉSUMÉ

BACKGROUND: MK8722 is a potent and systemic pan-AMPK activator. It is an effective, direct, allosteric activator of AMPK complex in many mammals. This study tried to explore the underlying anti-cancer molecular mechanism of MK8722 in human pancreatic cancer cells (PCCs). METHODS: The anti-proliferation, invasion and migration functions of MK8722 in human pancreatic cancer analyzed by real time cellular analysis, colony formation assay, cell migration assay, transwell assay and flow cytometery analysis. Moreover, the potential targeted signaling pathway was tested via RNA-seq and pathway enrichment analysis. RESULTS: In the present study, we investigated the anti-PCCs effects of MK8722 on two different human pancreatic cancer cell lines (PANC-1 and Patu8988). The results showed that MK8722 significantly inhibited human tumor cells proliferation and migration/invasion in a dose-dependent manner. Additionally, the influence of MK8722 was examined by analyzing the expression of potential key genes and pathways, which may provide novel insights to the mechanism of MK8722. CONCLUSION: The inhibition of pancreatic cancer by MK8722 through a number of pathways that inhibit carcinoma proliferation, invasion and migration. The potential effect of MK8722 might be determined by regulating the expression of AL162151, IER2, REPIN1, KRT80 to inhibit cycle arrest and migration.


Sujet(s)
AMP-Activated Protein Kinases/métabolisme , Antinéoplasiques/pharmacologie , Benzimidazoles/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Activateurs d'enzymes/pharmacologie , Tumeurs du pancréas/traitement médicamenteux , Pyridines/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Activation enzymatique , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Souris nude , Invasion tumorale , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Transduction du signal , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
19.
J Biol Chem ; 297(5): 101335, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34688654

RÉSUMÉ

Oncogenic KRAS drives cancer growth by activating diverse signaling networks, not all of which have been fully delineated. We set out to establish a system-wide profile of the KRAS-regulated kinase signaling network (kinome) in KRAS-mutant pancreatic ductal adenocarcinoma (PDAC). We knocked down KRAS expression in a panel of six cell lines and then applied multiplexed inhibitor bead/MS to monitor changes in kinase activity and/or expression. We hypothesized that depletion of KRAS would result in downregulation of kinases required for KRAS-mediated transformation and in upregulation of other kinases that could potentially compensate for the deleterious consequences of the loss of KRAS. We identified 15 upregulated and 13 downregulated kinases in common across the panel of cell lines. In agreement with our hypothesis, all 15 of the upregulated kinases have established roles as cancer drivers (e.g., SRC, TGF-ß1, ILK), and pharmacological inhibition of one of these upregulated kinases, DDR1, suppressed PDAC growth. Interestingly, 11 of the 13 downregulated kinases have established driver roles in cell cycle progression, particularly in mitosis (e.g., WEE1, Aurora A, PLK1). Consistent with a crucial role for the downregulated kinases in promoting KRAS-driven proliferation, we found that pharmacological inhibition of WEE1 also suppressed PDAC growth. The unexpected paradoxical activation of ERK upon WEE1 inhibition led us to inhibit both WEE1 and ERK concurrently, which caused further potent growth suppression and enhanced apoptotic death compared with WEE1 inhibition alone. We conclude that system-wide delineation of the KRAS-regulated kinome can identify potential therapeutic targets for KRAS-mutant pancreatic cancer.


Sujet(s)
Carcinome du canal pancréatique , Protéines du cycle cellulaire/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mutation , Tumeurs du pancréas , Protein-tyrosine kinases/métabolisme , Protéines proto-oncogènes p21(ras) , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/enzymologie , Carcinome du canal pancréatique/génétique , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Humains , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes p21(ras)/antagonistes et inhibiteurs , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme
20.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G477-G488, 2021 11 01.
Article de Anglais | MEDLINE | ID: mdl-34468207

RÉSUMÉ

Pancreatic cancer (PC) is among the most notorious malignancies worldwide. Long noncoding RNA (lncRNA) repulsive guidance molecule bone morphogenetic protein (BMP) coreceptor b antisense RNA 1 (RGMB-AS1) was an oncogene in glioma. However, the RGMB-AS1 function in PC remains largely unknown. Herein, quantitative real-time polymerase chain reaction was performed to analyze the expression of RGMB-AS1. We determined RGMB-AS1 influence on PC cell malignant behaviors via functional assays. Besides, we applied subcellular fractionation and fluorescence in situ hybridization (FISH) assays to confirm the cellular distribution of RGMB-AS1 in PC cells. We used mechanism assays to detect the regulatory axis of RGMB-AS1 in PC cells. Briefly, the level of RGMB-AS1 expression in PC cells was abnormally high. RGMB-AS1 knockdown impeded PC cell proliferation and migration, but induced cell apoptosis, and RGMB-AS1 overexpression led the opposite consequences. RGMB-AS1 acted as a competing endogenous RNA (ceRNA) to sequester miR-574-3p and thereby regulated Pim-3 proto-oncogene, serine/threonine kinase (PIM3) expression. Conclusively, our work revealed the cancer-promoting function of RGMB-AS1 in PC and that the regulatory mechanism of the RGMB-AS1/miR-574-3p/PIM3 axis might contribute to novel biomarker development in PC treatment.NEW & NOTEWORTHY RGMB-AS1 promotes PC cell proliferation, elevates PC cell migration capacity, inhibits PC cell apoptosis, and promotes PC cell proliferation and migration but inhibits cell apoptosis via targeting miR-574-3p. PIM3 is directly targeted by miR-574-3p.


Sujet(s)
Apoptose , Mouvement cellulaire , Prolifération cellulaire , microARN/métabolisme , Tumeurs du pancréas/enzymologie , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/métabolisme , ARN long non codant/métabolisme , Animaux , Lignée cellulaire tumorale , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Souris de lignée BALB C , Souris nude , microARN/génétique , Invasion tumorale , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Protein-Serine-Threonine Kinases/génétique , Proto-oncogène Mas , Protéines proto-oncogènes/génétique , ARN long non codant/génétique , Transduction du signal , Charge tumorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE