Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 64
Filtrer
1.
Cell Mol Gastroenterol Hepatol ; : 101387, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39111635

RÉSUMÉ

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) can develop from precursor lesions, including pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN). Previous studies indicated that loss of Acvr1b accelerates the Kras-mediated development of papillary IPMN in the mouse pancreas, however, the cell type predominantly affected by these genetic changes remains unclear. METHODS: We investigated the contribution of cellular origin by inducing IPMN associated mutations- KRASG12D expression and Acvr1b loss - specifically in acinar (Ptf1aCreER;KrasLSL-G12D;Acvr1bfl/fl mice) or ductal (Sox9CreER;KrasLSL-G12D;Acvr1bfl/fl mice) cells in mice. We then performed MRI imaging and a thorough histopathological analysis of their pancreatic tissues. RESULTS: The loss of Acvr1b increased the development of PanIN and IPMN-like lesions when either acinar and ductal cells expressed a Kras mutation. MRI, immunohistochemistry and histology revealed large IPMN-like lesions in these mice that exhibited features of flat, gastric epithelium. In addition, cyst formation in both mouse models was accompanied by chronic pancreatitis. Experimental acute pancreatitis accelerated the development of large mucinous cysts and PanIN when acinar, but not ductal, cells expressed mutant Kras and lost Acvr1b. CONCLUSION: These findings indicate that loss of Acvr1b in the presence of the Kras oncogene promotes the development of large and small precancerous lesions from both ductal and acinar cells. However, the IPMN-like phenotype was not equivalent to that observed when these mutations were made in all pancreatic cells during development. Our study underscores the significance of the cellular context in the initiation and progression of precursor lesions from exocrine cells.

2.
Biomedicines ; 11(2)2023 Jan 18.
Article de Anglais | MEDLINE | ID: mdl-36830797

RÉSUMÉ

Any gene therapy for cancer will be predicated upon its selectivity against cancer cells and non-toxicity to normal cells. Therefore, safeguards are needed to prevent its activation in normal cells. We designed a minimal p14ARF promoter with upstream Ap1 and E2F enhancer elements and a downstream MDR1 inhibitory element, TATA box, and a transcription initiation site (hereafter p14ARFmin). The modified p14ARFmin promoter was linked to bicistronic P14 and truncated BID (tBID) genes, which led to synergistic apoptosis via the intrinsic and extrinsic pathways of apoptosis when expressed. The promoter was designed to be preferentially activated by mutant Ras and completely inhibited by wild-type p53 so that only cells with both mutant Ras and mutant p53 would activate the construct. In comparison to most p53 gene therapies, this construct has selective advantages: (1) p53-based gene therapies with a constitutive CMV promoter cannot differentiate between normal cells and cancer cells, and can be toxic to normal cells; (2) our construct does not induce p21WAF/CIPI in contrast to other p53-based gene therapies, which can induce cell cycle arrest leading to increased chemotherapy resistance; (3) the modified construct (p14ARFmin-p14-tBID) demonstrates bidirectional control of its promoter, which is completely repressed by wild-type p53 and activated only in cells with both RAS and P53 mutations; and (4) a novel combination of genes (p14 and tBID) can synergistically induce potent intrinsic and extrinsic apoptosis in cancer cells.

3.
J Hepatobiliary Pancreat Sci ; 30(1): 133-143, 2023 Jan.
Article de Anglais | MEDLINE | ID: mdl-33811460

RÉSUMÉ

BACKGROUND: Although we previously proposed a nomogram to predict malignancy in intraductal papillary mucinous neoplasms (IPMN) and validated it in an external cohort, its application is challenging without data on tumor markers. Moreover, existing nomograms have not been compared. This study aimed to develop a nomogram based on radiologic findings and to compare its performance with previously proposed American and Korean/Japanese nomograms. METHODS: We recruited 3708 patients who underwent surgical resection at 31 tertiary institutions in eight countries, and patients with main pancreatic duct >10 mm were excluded. To construct the nomogram, 2606 patients were randomly allocated 1:1 into training and internal validation sets, and area under the receiver operating characteristics curve (AUC) was calculated using 10-fold cross validation by exhaustive search. This nomogram was then validated and compared to the American and Korean/Japanese nomograms using 1102 patients. RESULTS: Among the 2606 patients, 90 had main-duct type, 900 had branch-duct type, and 1616 had mixed-type IPMN. Pathologic results revealed 1628 low-grade dysplasia, 476 high-grade dysplasia, and 502 invasive carcinoma. Location, cyst size, duct dilatation, and mural nodule were selected to construct the nomogram. AUC of this nomogram was higher than the American nomogram (0.691 vs 0.664, P = .014) and comparable with the Korean/Japanese nomogram (0.659 vs 0.653, P = .255). CONCLUSIONS: A novel nomogram based on radiologic findings of IPMN is competitive for predicting risk of malignancy. This nomogram would be clinically helpful in circumstances where tumor markers are not available. The nomogram is freely available at http://statgen.snu.ac.kr/software/nomogramIPMN.


Sujet(s)
Adénocarcinome mucineux , Carcinome du canal pancréatique , Carcinome papillaire , Tumeurs intracanalaires pancréatiques , Tumeurs du pancréas , Humains , Nomogrammes , Tumeurs intracanalaires pancréatiques/imagerie diagnostique , Carcinome du canal pancréatique/imagerie diagnostique , Carcinome du canal pancréatique/chirurgie , Carcinome du canal pancréatique/anatomopathologie , Adénocarcinome mucineux/imagerie diagnostique , Adénocarcinome mucineux/chirurgie , Adénocarcinome mucineux/anatomopathologie , Carcinome papillaire/anatomopathologie , Tumeurs du pancréas/imagerie diagnostique , Tumeurs du pancréas/chirurgie , Tumeurs du pancréas/anatomopathologie , Marqueurs biologiques tumoraux , Hyperplasie , Études rétrospectives
4.
Genes Dis ; 9(3): 820-825, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-35782981

RÉSUMÉ

In order to assess Stathmin as an immunohistochemical (IHC) indicator of phosphatidylinositol 3-kinase (PI3K) pathway activity in HPV-negative head & neck squamous cell carcinoma (HNSCC), we compared Stathmin IHC to expression of other pathway components. We also evaluated the relationship between Stathmin IHC and the mutational status of four key pathway genes. Finally, we ascertained whether Stathmin IHC correlates with tumor grade or primary site. Correlation exists between high Stathmin expression and high pAKT1 expression, indicating a role for Stathmin IHC as a marker of pathway activity. Our analysis did not show correlation between Stathmin IHC and mutation of the four genes evaluated. We also observed an association between high Stathmin expression and oropharyngeal primary site. Our results suggest utility of Stathmin IHC as an indicator of PI3K pathway activity, and thereby demonstrate potential relevance of Stathmin IHC in the context of HNSCC.

5.
Cancer Res Commun ; 2(12): 1601-1616, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36970723

RÉSUMÉ

Expression of the Notch family of receptors is often upregulated in pancreatic ductal adenocarcinoma (PDAC). In this study, we focused on Notch4, which had not been investigated in PDAC. We generated KC (LSL-KrasG12D;p48-Cre), N4 - / - KC (Notch4- / -;LSL-KrasG12D;p48-Cre), PKC (p16fl/fl;LSL-KrasG12D;p48-Cre), and N4 - / - PKC (Notch4-/ -; p16fl/f l;LSL-KrasG12D;p48-Cre) genetically engineered mouse models (GEMM). We performed caerulein treatment in both KC and N4 - / - KC mice, and the development of acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN) lesions were significantly diminished in the N4 - / - KC than in the KC GEMM (P = 0.01). This in vivo result was validated by in vitro ADM induction of the explant cultures of pancreatic acinar cells from the N4 - / - KC and KC mice (P < 0.001), confirming that Notch4 is an important contributor to early pancreatic tumorigenesis. To evaluate the role of Notch4 in the later stage of pancreatic tumorigenesis, we compared the PKC and N4 - / - PKC mice. The N4 - / - PKC mice had better overall survival (P = 0.012) and significantly reduced tumor burden (PanIN: P = 0.018 at 2 months, PDAC: P = 0.039 at 5 months) compared with the PKC GEMM. RNA-sequencing analysis of pancreatic tumor cell lines derived from the PKC and N4 - / - PKC GEMMs revealed that 408 genes were differentially expressed (FDR < 0.05) and Pcsk5 is a potential downstream effector of the Notch4 signaling pathway (P < 0.001). Low expression of Pcsk5 positively correlates with good survival in patients with PDAC (P = 0.028). We have identified a novel role for Notch4 signaling with tumor-promoting function in pancreatic tumorigenesis. Our study also uncovered a novel association between Pcsk5 and Notch4 signaling in PDAC. Significance: We demonstrated that global inactivation of Notch4 significantly improved the survival of an aggressive mouse model for PDAC and provided preclinical evidence that Notch4 and Pcsk5 are novel targets for PDAC therapies.


Sujet(s)
Épithélioma in situ , Carcinome du canal pancréatique , Tumeurs du pancréas , Souris , Animaux , Protéines proto-oncogènes p21(ras)/métabolisme , Tumeurs du pancréas/génétique , Transformation cellulaire néoplasique/génétique , Carcinogenèse/génétique , Carcinome du canal pancréatique/génétique , Épithélioma in situ/génétique , Tumeurs du pancréas
6.
Oncogene ; 40(50): 6759-6771, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34663879

RÉSUMÉ

Human pancreatic ductal adenocarcinoma (PDAC) harboring one KRAS mutant allele often displays increasing genomic loss of the remaining wild-type (WT) allele (known as LOH at KRAS) as tumors progress to metastasis, yet the molecular ramification of this WT allelic loss is unknown. In this study, we showed that the restoration of WT KRAS expression in human PDAC cell lines with LOH at KRAS significantly attenuated the malignancy of PDAC cells both in vitro and in vivo, demonstrating a tumor-suppressive role of the WT KRAS allele. Through RNA-Seq, we identified the HIPPO signaling pathway to be positively regulated by WT KRAS in PDAC cells. In accordance with this observation, PDAC cells with LOH at KRAS exhibited increased nuclear localization and activation of transcriptional co-activator YAP1. Mechanistically, we discovered that WT KRAS expression sequestered YAP1 from the nucleus, through enhanced 14-3-3zeta interaction with phosphorylated YAP1 at S127. Consistently, expression of a constitutively-active YAP1 mutant in PDAC cells bypassed the growth inhibitory effects of WT KRAS. In patient samples, we found that the YAP1-activation genes were significantly upregulated in tumors with LOH at KRAS, and YAP1 nuclear localization predicted poor survival for PDAC patients. Collectively, our results reveal that the WT allelic loss leads to functional activation of YAP1 and enhanced tumor malignancy, which explains the selection advantage of the tumor cells with LOH at KRAS during pancreatic cancer clonal evolution and progression to metastasis, and should be taken into consideration in future therapeutic strategies targeting KRAS.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Régulation de l'expression des gènes tumoraux , Perte d'hétérozygotie , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes p21(ras)/génétique , Protéines de signalisation YAP/métabolisme , Animaux , Apoptose , Marqueurs biologiques tumoraux/génétique , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Prolifération cellulaire , Femelle , Facteurs de transcription Forkhead/physiologie , Humains , Souris , Souris nude , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Pronostic , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines de signalisation YAP/génétique
7.
Biomedicines ; 9(7)2021 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-34356885

RÉSUMÉ

Activin, a member of the TGF-ß superfamily, is involved in many physiological processes, such as embryonic development and follicle development, as well as in multiple human diseases including cancer. Genetic mutations in the activin signaling pathway have been reported in many cancer types, indicating that activin signaling plays a critical role in tumorigenesis. Recent evidence reveals that activin signaling may function as a tumor-suppressor in tumor initiation, and a promoter in the later progression and metastasis of tumors. This article reviews many aspects of activin, including the signaling cascade of activin, activin-related proteins, and its role in tumorigenesis, particularly in pancreatic cancer development. The mechanisms regulating its dual roles in tumorigenesis remain to be elucidated. Further understanding of the activin signaling pathway may identify potential therapeutic targets for human cancers and other diseases.

8.
Sci Rep ; 10(1): 20140, 2020 11 18.
Article de Anglais | MEDLINE | ID: mdl-33208887

RÉSUMÉ

Most models for predicting malignant pancreatic intraductal papillary mucinous neoplasms were developed based on logistic regression (LR) analysis. Our study aimed to develop risk prediction models using machine learning (ML) and LR techniques and compare their performances. This was a multinational, multi-institutional, retrospective study. Clinical variables including age, sex, main duct diameter, cyst size, mural nodule, and tumour location were factors considered for model development (MD). After the division into a MD set and a test set (2:1), the best ML and LR models were developed by training with the MD set using a tenfold cross validation. The test area under the receiver operating curves (AUCs) of the two models were calculated using an independent test set. A total of 3,708 patients were included. The stacked ensemble algorithm in the ML model and variable combinations containing all variables in the LR model were the most chosen during 200 repetitions. After 200 repetitions, the mean AUCs of the ML and LR models were comparable (0.725 vs. 0.725). The performances of the ML and LR models were comparable. The LR model was more practical than ML counterpart, because of its convenience in clinical use and simple interpretability.


Sujet(s)
Modèles logistiques , Apprentissage machine , Tumeurs intracanalaires pancréatiques/anatomopathologie , Sujet âgé , Algorithmes , Diagnostic assisté par ordinateur/méthodes , Femelle , Humains , Mâle , Adulte d'âge moyen , Kyste du pancréas/anatomopathologie , Tumeurs intracanalaires pancréatiques/imagerie diagnostique , Pronostic , Études rétrospectives , Facteurs de risque
9.
Mol Cancer Res ; 18(6): 822-834, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32152233

RÉSUMÉ

The PI3K signaling pathway is frequently mutated in head and neck squamous cell carcinoma (HNSCC), often via gain-of-function (GOF) mutations in the PIK3CA gene. Here, we present novel genetically engineered mouse models (GEMM) carrying a GOF allele Loxp-STOP-Loxp(LSL)-PIK3CAH1047R (E20) alone or in combination with heterozygous LSL-p53+/R172H (p53) mutation with tissue-specific expression to interrogate the role of oncogenic PIK3CA in transformation of upper aerodigestive track epithelium. We demonstrated that the GOF PIK3CA mutation promoted progression of 4-nitroquinoline 1-oxide-induced oral squamous cell carcinoma (OSCC) in both E20 single mutant and E20/p53 double mutant mice, with frequent distal metastasis detected only in E20/p53 GEMM. Similar to in human OSCC, loss of p16 was associated with progression of OSCC in these mice. RNA-seq analyses revealed that among the common genes differentially expressed in primary OSCC cell lines derived from E20, p53, and E20/p53 GEMMs compared with those from the wild-type mice, genes associated with proliferation and cell cycle were predominantly represented, which is consistent with the progressive loss of p16 detected in these GEMMs. Importantly, all of these OSCC primary cell lines exhibited enhanced sensitivity to BYL719 and cisplatin combination treatment in comparison with cisplatin alone in vitro and in vivo, regardless of p53 and/or p16 status. Given the prevalence of mutations in p53 and the PI3K pathways in HNSCC in conjunction with loss of p16 genetically or epigenetically, this universal increased sensitivity to cisplatin and BYL719 combination therapy in cancer cells with PIK3CA mutation represents an opportunity to a subset of patients with HNSCC. IMPLICATIONS: Our results suggest that combination therapy of cisplatin and PI3K inhibitor may be worthy of consideration in patients with HNSCC with PIK3CA mutation.


Sujet(s)
4-Nitro-quinoléine-1-oxyde/toxicité , Phosphatidylinositol 3-kinases de classe I/génétique , Tumeurs de la tête et du cou/anatomopathologie , Mutation , Carcinome épidermoïde de la tête et du cou/secondaire , Protéine p53 suppresseur de tumeur/génétique , Animaux , Cancérogènes/toxicité , Évolution de la maladie , Tumeurs de la tête et du cou/induit chimiquement , Tumeurs de la tête et du cou/génétique , Souris , Souris nude , Carcinome épidermoïde de la tête et du cou/induit chimiquement , Carcinome épidermoïde de la tête et du cou/génétique
11.
Mol Cancer Res ; 17(10): 2029-2041, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31383722

RÉSUMÉ

Mutation or promoter hypermethylation of CDKN2A is found in over 90% of pancreatic ductal adenocarcinomas (PDAC) and leads to loss of function of cell-cycle inhibitors p16 (INK4A) and p14 (ARF) resulting in unchecked proliferation. The CDK4/6 inhibitor, abemaciclib, has nanomolar IC50s in PDAC cell lines and decreases growth through inhibition of phospho-Rb (pRb), G1 cell-cycle arrest, apoptosis, and the senescent phenotype detected with ß-galactosidase staining and relevant mRNA elevations. Daily abemaciclib treatments in mouse PDAC xenograft studies were safe and demonstrated a 3.2-fold decrease in tumor volume compared with no treatment (P < 0.0001) accompanying a decrease in both pRb and Ki67. We determined that inhibitors of HuR (ELAVL1), a prosurvival mRNA stability factor that regulates cyclin D1, and an inhibitor of Yes-Associated Protein 1 (YAP1), a pro-oncogenic, transcriptional coactivator important for CDK6 and cyclin D1, were both synergistic with abemaciclib. Accordingly, siRNA oligonucleotides targeted against HuR, YAP1, and their common target cyclin D1, validated the synergy studies. In addition, we have seen increased sensitivity to abemaciclib in a PDAC cell line that harbors a loss of the ELAVL1 gene via CRISP-Cas9 technology. As an in vitro model for resistance, we investigated the effects of long-term abemaciclib exposure. PDAC cells chronically cultured with abemaciclib displayed a reduction in cellular growth rates (GR) and coresistance to gemcitabine and 5-fluorouracil (5-FU), but not to HuR or YAP1 inhibitors as compared with no treatment controls. We believe that our data provide compelling preclinical evidence for an abemaciclib combination-based clinical trial in patients with PDAC. IMPLICATIONS: Our data suggest that abemaciclib may be therapeutically relevant for the treatment in PDAC, especially as part of a combination regimen inhibiting YAP1 or HuR.


Sujet(s)
Protéines adaptatrices de la transduction du signal/génétique , Aminopyridines/usage thérapeutique , Benzimidazoles/usage thérapeutique , Protéine-1 similaire à ELAV/génétique , Tumeurs du pancréas/traitement médicamenteux , Facteurs de transcription/génétique , Aminopyridines/pharmacologie , Animaux , Benzimidazoles/pharmacologie , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Souris nude , Tumeurs du pancréas/anatomopathologie , Transfection , Protéines de signalisation YAP
12.
Cancer Biol Ther ; 20(6): 855-865, 2019.
Article de Anglais | MEDLINE | ID: mdl-30866697

RÉSUMÉ

SRC and its activated form, phospho-SRC (pSRC), are aberrantly activated in pancreatic cancer and SRC represents a potential target for pancreatic cancer therapy. In this paper, we examined the inhibitory effect of dasatinib, a potent SRC inhibitor in combination with paclitaxel or gemcitabine on human and murine pancreatic cancer cell lines. The results showed that p-SRC can be highly expressed in most human and mouse pancreatic cancer cell lines compared with normal human cell lines and can be induced by paclitaxel or gemcitabine in HPAC cells. Dasatinib can enhance the efficacy of paclitaxel or gemcitabine by reducing the cell viability and inhibiting the cell proliferation. Dasatinib with paclitaxel combination exhibits statistically greater inhibition of the cell migration ability than single agent alone, paclitaxel with gemcitabine or FOLFIRINOX (combination of fluorouracil, leucovorin, irinotecan, and oxaliplatin) in HAPC, PANC-1, and BXPC-3 human pancreatic cancer cell lines as well as 8-285 APR and 8-365 APR mouse pancreatic cancer cell lines. In addition, dasatinib with gemcitabine combination also showed statistically greater inhibition of cell migration than single agent alone, paclitaxel with gemcitabine, or FOLFIRINOX in HAPC, PANC-1 and 8-285 APR cells. The combination of dasatinib with paclitaxel or gemcitabine also showed greater inhibition of the colony formation ability of pancreatic cancer cells compared with single-agent monotherapy or FOLFIRINOX. Dasatinib with paclitaxel or gemcitabine combination also inhibits p-SRC, p-STAT3, p-AKT, and/or p-ERK in these pancreatic cancer cells. Therefore, our results support that combined dasatinib and paclitaxel or gemcitabine therapy may be a viable therapeutic approach for human pancreatic cancer.


Sujet(s)
Antinéoplasiques/pharmacologie , Dasatinib/pharmacologie , Désoxycytidine/analogues et dérivés , Paclitaxel/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Désoxycytidine/pharmacologie , Synergie des médicaments , Humains , Tumeurs du pancréas , Facteur de transcription STAT-3/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques ,
13.
Curr Cancer Drug Targets ; 19(5): 417-427, 2019.
Article de Anglais | MEDLINE | ID: mdl-29714141

RÉSUMÉ

BACKGROUND: Elevated production of the pro-inflammatory cytokine interleukin-6 (IL-6) and dysfunction of IL-6 signaling promotes tumorigenesis and are associated with poor survival outcomes in multiple cancer types. Recent studies showed that the IL-6/GP130/STAT3 signaling pathway plays a pivotal role in pancreatic cancer development and maintenance. OBJECTIVE: We aim to develop effective treatments through inhibition of IL-6/GP130 signaling in pancreatic cancer. METHODS: The effects on cell viability and cell proliferation were measured by MTT and BrdU assays, respectively. The effects on glycolysis was determined by cell-based assays to measure lactate levels. Protein expression changes were evaluated by western blotting and immunoprecipitation. siRNA transfection was used to knock down estrogen receptor α gene expression. Colony forming ability was determined by colony forming cell assay. RESULTS: We demonstrated that IL-6 can induce pancreatic cancer cell viability/proliferation and glycolysis. We also showed that a repurposing FDA-approved drug bazedoxifene could inhibit the IL-6/IL-6R/GP130 complexes. Bazedoxifene also inhibited JAK1 binding to IL-6/IL-6R/GP130 complexes and STAT3 phosphorylation. In addition, bazedoxifene impeded IL-6 mediated cell viability/ proliferation and glycolysis in pancreatic cancer cells. Consistently, other IL-6/GP130 inhibitors SC144 and evista showed similar inhibition of IL-6 stimulated cell viability, cell proliferation and glycolysis. Furthermore, all three IL-6/GP130 inhibitors reduced the colony forming ability in pancreatic cancer cells. CONCLUSION: Our findings demonstrated that IL-6 stimulates pancreatic cancer cell proliferation, survival and glycolysis, and supported persistent IL-6 signaling is a viable therapeutic target for pancreatic cancer using IL-6/GP130 inhibitors.


Sujet(s)
Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteur gp130 de cytokines/antagonistes et inhibiteurs , Glycolyse/effets des médicaments et des substances chimiques , Indoles/pharmacologie , Interleukine-6/antagonistes et inhibiteurs , Tumeurs du pancréas/traitement médicamenteux , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Animaux , Apoptose , Test clonogénique , Humains , Souris , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Cellules cancéreuses en culture
14.
Methods Mol Biol ; 1882: 229-237, 2019.
Article de Anglais | MEDLINE | ID: mdl-30378059

RÉSUMÉ

Extracellular vesicles (EVs) are a diverse category of cellular export products that are present in a variety of biofluids and cell culture media. EVs contain a wide variety of macromolecules that represent a sampling of the cytoplasmic or endosomal compartments and function in cell-to-cell paracrine and endocrine signaling; it has been demonstrated that pathological states such as oxidative stress, transformation, apoptosis, and various cell injuries induce cells to increase their EV release rate, simultaneously altering their composition to reflect the altered state of the cellular origin. Specifically, in patients with solid tumors, EVs are released from cancerous cells at a higher rate than from healthy cells and are enriched in tumor signature molecules. Because of their stability, increased concentration, and unique signatures in cancer patients, EVs have become the subject of investigation for diagnostic and prognostic purposes. Moreover, understanding EVs' biogenesis and biological role could lead to novel insights toward cellular cross talk and complex biological pathways in cancer research. To make use of EVs for diagnostic and mechanistic cancer research, standardized well-characterized methods are required. This chapter provides an overview of two EV isolation techniques and provides detailed instructions on the isolation of EVs by ultracentrifugation, the labor-intensive gold standard, and concentrated polymer precipitation, a faster, higher-yield technique that can be utilized in cancer research.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Centrifugation en gradient de densité/méthodes , Vésicules extracellulaires/composition chimique , Tumeurs/diagnostic , Marqueurs biologiques tumoraux/métabolisme , Techniques de culture cellulaire/instrumentation , Techniques de culture cellulaire/méthodes , Lignée cellulaire tumorale , Centrifugation en gradient de densité/instrumentation , Précipitation chimique , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/ultrastructure , Cytométrie en flux/méthodes , Humains , Microscopie électronique à transmission/méthodes , Tumeurs/sang , Tumeurs/anatomopathologie , Tumeurs/urine , Polymères/composition chimique
15.
Cancer Lett ; 442: 333-340, 2019 02 01.
Article de Anglais | MEDLINE | ID: mdl-30447255

RÉSUMÉ

HHLA2 is a newly identified member of the B7 immune checkpoint family, but its function and crosstalk with immune cells is not fully understood. To gain insights into the HHLA2 expression profile and to determine the clinical significance and function of HHLA2 in pancreatic cancer, we performed immunohistochemistry (IHC) analyses on tissue microarrays (TMAs) of pancreatic ductal adenocarcinoma (PDAC, n = 92) with matched peritumoral tissues as well as in cohorts of precancerous lesions: pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN). We found that HHLA2 was rarely detected in normal acinar, islet, and ductal cells but widely expressed from early pancreatic precancerous lesions to invasive PDAC. The overall HHLA2 positivity was 95% (19/20) in low grade PanIN and 70.73% (29/41) in IPMN. HHLA2 expression was detected in 77.17% (71/92) of the PDAC cases and was significantly associated with better prognosis in this cohort. Our findings suggest that HHLA2 may behave as a costimulatory ligand in pancreatic cancer, which differs from other B7 family members that are largely characterized as checkpoint inhibitors. Further investigation of the HHLA2 signaling pathway and its receptors is warranted by our data and may lead to novel therapeutic interventions.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Épithélioma in situ/immunologie , Carcinome du canal pancréatique/immunologie , Immunoglobulines/analyse , Tumeurs intracanalaires pancréatiques/immunologie , Tumeurs du pancréas/immunologie , Épithélioma in situ/mortalité , Épithélioma in situ/anatomopathologie , Épithélioma in situ/chirurgie , Carcinome du canal pancréatique/mortalité , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/chirurgie , Femelle , Humains , Immunohistochimie , Mâle , Adulte d'âge moyen , Pancréatectomie , Tumeurs intracanalaires pancréatiques/mortalité , Tumeurs intracanalaires pancréatiques/anatomopathologie , Tumeurs intracanalaires pancréatiques/chirurgie , Tumeurs du pancréas/mortalité , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/chirurgie , Études rétrospectives , Facteurs temps , Analyse sur puce à tissus , Résultat thérapeutique , Régulation positive
16.
Cancer Lett ; 423: 71-79, 2018 06 01.
Article de Anglais | MEDLINE | ID: mdl-29526803

RÉSUMÉ

PanINs and IPMNs are the two most common precursor lesions that can progress to invasive pancreatic ductal adenocarcinoma (PDA). DCLK1 has been identified as a biomarker of progenitor cells in PDA progressed from PanINs. To explore the potential role of DCLK1-expressing cells in the genesis of IPMNs, we compared the incidence of DCLK1-positive cells in pancreatic tissue samples from genetically-engineered mouse models (GEMMs) for IPMNs, PanINs, and acinar to ductal metaplasia by immunohistochemistry and immunofluorescence. Mouse lineage tracing experiments in the IPMN GEMM showed that DCLK1+ cells originated from a cell lineage distinct from PDX1+ progenitors. The DCLK1+ cells shared the features of tuft cells but were devoid of IPMN tumor biomarkers. The DCLK1+ cells were detected in the earliest proliferative acinar clusters prior to the formation of metaplastic ductal cells, and were enriched in the "IPMN niches". In summary, DCLK1 labels a unique pancreatic cellular lineage in the IPMN GEMM. The clustering of DCLK1+ cells is an early event in Kras-induced pancreatic tumorigenesis and may contribute to IPMN initiation.


Sujet(s)
Protéines à homéodomaine/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Tumeurs intracanalaires pancréatiques/génétique , Tumeurs du pancréas/génétique , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes p21(ras)/génétique , Transactivateurs/métabolisme , Animaux , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Lignage cellulaire , Prolifération cellulaire , Kinases de type doublecortine , Femelle , Génie génétique , Protéines à homéodomaine/génétique , Humains , Facteur-4 de type Kruppel , Mâle , Souris , Invasion tumorale , Transplantation tumorale , Tumeurs intracanalaires pancréatiques/métabolisme , Tumeurs du pancréas/métabolisme , Transactivateurs/génétique
17.
Acta Neuropathol ; 135(6): 887-906, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29397421

RÉSUMÉ

The most prevalent neurological disorders of myelin include perinatal brain injury leading to cerebral palsy in infants and multiple sclerosis in adults. Although these disorders have distinct etiologies, they share a common neuropathological feature of failed progenitor differentiation into myelin-producing oligodendrocytes and lack of myelin, for which there is an unmet clinical need. Here, we reveal that a molecular pathology common to both disorders is dysregulation of activin receptors and that activin receptor signaling is required for the majority of myelin generation in development and following injury. Using a constitutive conditional knockout of all activin receptor signaling in oligodendrocyte lineage cells, we discovered this signaling to be required for myelination via regulation of oligodendrocyte differentiation and myelin compaction. These processes were found to be dependent on the activin receptor subtype Acvr2a, which is expressed during oligodendrocyte differentiation and axonal ensheathment in development and following myelin injury. During efficient myelin regeneration, Acvr2a upregulation was seen to coincide with downregulation of Acvr2b, a receptor subtype with relatively higher ligand affinity; Acvr2b was shown to be dispensable for activin receptor-driven oligodendrocyte differentiation and its overexpression was sufficient to impair the abovementioned ligand-driven responses. In actively myelinating or remyelinating areas of human perinatal brain injury and multiple sclerosis tissue, respectively, oligodendrocyte lineage cells expressing Acvr2a outnumbered those expressing Acvr2b, whereas in non-repairing lesions Acvr2b+ cells were increased. Thus, we propose that following human white matter injury, this increase in Acvr2b expression would sequester ligand and consequently impair Acvr2a-driven oligodendrocyte differentiation and myelin formation. Our results demonstrate dysregulated activin receptor signaling in common myelin disorders and reveal Acvr2a as a novel therapeutic target for myelin generation following injury across the lifespan.


Sujet(s)
Récepteur activine/métabolisme , Différenciation cellulaire/physiologie , Lignage cellulaire/physiologie , Oligodendroglie/métabolisme , Récepteur activine/génétique , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Lésions encéphaliques/métabolisme , Lésions encéphaliques/anatomopathologie , Cellules cultivées , Femelle , Humains , Mâle , Souris de lignée C57BL , Souris knockout , Sclérose en plaques/métabolisme , Sclérose en plaques/anatomopathologie , Oligodendroglie/anatomopathologie , Rat Sprague-Dawley , Techniques de culture de tissus , Structures d'échafaudage tissulaires
18.
Laryngoscope ; 127(10): E336-E339, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28543373

RÉSUMÉ

OBJECTIVES/HYPOTHESIS: A candidate variant (p.Val496Ala) of the ACSS2 gene (T > C missense, rs59088485 variant at chr20: bp37 33509608) was previously found to consistently segregate with nonsyndromic cleft lip and/or palate (NSCLP) in three Honduran families. Objectives of this study were 1) to investigate the frequency of this ACSS2 variant in Honduran unrelated NSCLP patients and unrelated unaffected controls and 2) to investigate the frequency of this variant in Colombian unrelated affected NSCLP patients and unrelated unaffected controls. STUDY DESIGN: Case-control studies. METHODS: Sanger sequencing of 99 unrelated Honduran NSCLP patients and 215 unrelated unaffected controls for the p.Val496Ala ACSS2 variant was used to determine the carrier frequency in NSCLP patients and controls. Sanger sequencing of 230 unrelated Colombian NSCLP patients and 146 unrelated unaffected controls for the p.Val496Ala ACSS2 variant was used to determine the carrier frequency in NSCLP patients and controls. RESULTS: In the Honduran population, the odds ratio of having NSCLP among carriers of the p.Val496Ala ACSS2 variant was 4.0 (P = .03), with a carrier frequency of seven of 99 (7.1%) in unrelated affected and four of 215 (1.9%) in unrelated unaffected individuals. In the Colombian population, the odds ratio of having NSCLP among carriers of the p.Val496Ala ACSS2 variant was 2.6 (P = .04), with a carrier frequency of 23 of 230 (10.0%) in unrelated affected and six of 146 (4.1%) in unrelated unaffected individuals. CONCLUSIONS: These findings support the role of ACSS2 in NSCLP in two independent Hispanic populations from Honduras and Colombia. LEVEL OF EVIDENCE: NA Laryngoscope, 127:E336-E339, 2017.


Sujet(s)
Acetate coA-ligase/génétique , Encéphale/malformations , Bec-de-lièvre/génétique , Fente palatine/génétique , Acetate coA-ligase/sang , Études cas-témoins , Enfant d'âge préscolaire , Bec-de-lièvre/sang , Fente palatine/sang , Colombie , Femelle , Fréquence d'allèle , Études d'associations génétiques , Prédisposition génétique à une maladie , Honduras , Humains , Mâle , Odds ratio , Polymorphisme de nucléotide simple , Analyse de séquence d'ADN/méthodes
19.
Laryngoscope ; 127(10): 2279-2281, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28407312

RÉSUMÉ

This case report describes the use of CO2 laser and quadrivalent human papillomavirus (HPV) vaccination in two patients with nasopharyngeal HPV11-positive recurrent respiratory papillomas (RRP). These patients initially underwent CO2 laser excision but developed recurrent lesions and underwent HPV vaccination as adjunct therapy. The recurrent lesions shrank after vaccination and were again excised with CO2 laser. Subsequently, these patients had no recurrence of lesions on long-term follow-up at 33 months or 6 years after surgery. Thus, in patients with nasopharyngeal RRP, resection with CO2 laser, and HPV vaccination as adjunct therapy should be considered in HPV11-associated cases. Laryngoscope, 127:2279-2281, 2017.


Sujet(s)
Papillomavirus humain de type 6/immunologie , Thérapie laser/méthodes , Tumeurs du rhinopharynx/thérapie , Papillome/chirurgie , Infections à papillomavirus/thérapie , Vaccins contre les papillomavirus/usage thérapeutique , Vaccination/méthodes , Adulte , Humains , Lasers à gaz/usage thérapeutique , Mâle , Adulte d'âge moyen , Papillome/diagnostic
20.
Oncotarget ; 8(13): 22203-22217, 2017 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-28108737

RÉSUMÉ

The phosphoinositide 3-kinase (PI3K) signaling pathway is the most commonly mutated pathway in head and neck squamous cell carcinoma (HNSCC). There are several drugs targeting members of the PI3K signaling pathway in development for HNSCC. In this article, we review the genetic alterations reported in the pathway pertinent to HNSCC, various agents in development targeting various mediators of the pathway, results from clinical trials, and remaining challenges in the development of PI3K pathway inhibitors.


Sujet(s)
Carcinome épidermoïde/traitement médicamenteux , Tumeurs de la tête et du cou/traitement médicamenteux , Thérapie moléculaire ciblée , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Carcinome épidermoïde/enzymologie , Carcinome épidermoïde/anatomopathologie , Tumeurs de la tête et du cou/enzymologie , Tumeurs de la tête et du cou/anatomopathologie , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE