Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Mol Cancer ; 23(1): 114, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811984

RÉSUMÉ

BACKGROUND: Prostate cancer develops through malignant transformation of the prostate epithelium in a stepwise, mutation-driven process. Although activator protein-1 transcription factors such as JUN have been implicated as potential oncogenic drivers, the molecular programs contributing to prostate cancer progression are not fully understood. METHODS: We analyzed JUN expression in clinical prostate cancer samples across different stages and investigated its functional role in a Pten-deficient mouse model. We performed histopathological examinations, transcriptomic analyses and explored the senescence-associated secretory phenotype in the tumor microenvironment. RESULTS: Elevated JUN levels characterized early-stage prostate cancer and predicted improved survival in human and murine samples. Immune-phenotyping of Pten-deficient prostates revealed high accumulation of tumor-infiltrating leukocytes, particularly innate immune cells, neutrophils and macrophages as well as high levels of STAT3 activation and IL-1ß production. Jun depletion in a Pten-deficient background prevented immune cell attraction which was accompanied by significant reduction of active STAT3 and IL-1ß and accelerated prostate tumor growth. Comparative transcriptome profiling of prostate epithelial cells revealed a senescence-associated gene signature, upregulation of pro-inflammatory processes involved in immune cell attraction and of chemokines such as IL-1ß, TNF-α, CCL3 and CCL8 in Pten-deficient prostates. Strikingly, JUN depletion reversed both the senescence-associated secretory phenotype and senescence-associated immune cell infiltration but had no impact on cell cycle arrest. As a result, JUN depletion in Pten-deficient prostates interfered with the senescence-associated immune clearance and accelerated tumor growth. CONCLUSIONS: Our results suggest that JUN acts as tumor-suppressor and decelerates the progression of prostate cancer by transcriptional regulation of senescence- and inflammation-associated genes. This study opens avenues for novel treatment strategies that could impede disease progression and improve patient outcomes.


Sujet(s)
Évolution de la maladie , Phosphohydrolase PTEN , Tumeurs de la prostate , Microenvironnement tumoral , Mâle , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Animaux , Souris , Humains , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Microenvironnement tumoral/immunologie , Phénotype sécrétoire associé à la sénescence , Protéines proto-oncogènes c-jun/métabolisme , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Vieillissement de la cellule/génétique , Modèles animaux de maladie humaine
2.
Mol Cancer ; 22(1): 133, 2023 08 12.
Article de Anglais | MEDLINE | ID: mdl-37573301

RÉSUMÉ

Prostate cancer (PCa) is a common and fatal type of cancer in men. Metastatic PCa (mPCa) is a major factor contributing to its lethality, although the mechanisms remain poorly understood. PTEN is one of the most frequently deleted genes in mPCa. Here we show a frequent genomic co-deletion of PTEN and STAT3 in liquid biopsies of patients with mPCa. Loss of Stat3 in a Pten-null mouse prostate model leads to a reduction of LKB1/pAMPK with simultaneous activation of mTOR/CREB, resulting in metastatic disease. However, constitutive activation of Stat3 led to high LKB1/pAMPK levels and suppressed mTORC1/CREB pathway, preventing mPCa development. Metformin, one of the most widely prescribed therapeutics against type 2 diabetes, inhibits mTORC1 in liver and requires LKB1 to mediate glucose homeostasis. We find that metformin treatment of STAT3/AR-expressing PCa xenografts resulted in significantly reduced tumor growth accompanied by diminished mTORC1/CREB, AR and PSA levels. PCa xenografts with deletion of STAT3/AR nearly completely abrogated mTORC1/CREB inhibition mediated by metformin. Moreover, metformin treatment of PCa patients with high Gleason grade and type 2 diabetes resulted in undetectable mTORC1 levels and upregulated STAT3 expression. Furthermore, PCa patients with high CREB expression have worse clinical outcomes and a significantly increased risk of PCa relapse and metastatic recurrence. In summary, we have shown that STAT3 controls mPCa via LKB1/pAMPK/mTORC1/CREB signaling, which we have identified as a promising novel downstream target for the treatment of lethal mPCa.


Sujet(s)
Diabète de type 2 , Metformine , Tumeurs de la prostate , Animaux , Humains , Mâle , Souris , AMP-Activated Protein Kinases/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Metformine/pharmacologie , Récidive tumorale locale , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme
3.
Mol Metab ; 72: 101711, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36958422

RÉSUMÉ

PURPOSE: Heart diseases are the leading cause of death worldwide. Metabolic interventions via ketogenic diets (KDs) have been used for decades to treat epilepsy, and more recently, also diabetes and obesity, as common comorbidities of heart diseases. However, recent reports linked KDs, based on long-chain triglycerides (LCTs), to cardiac fibrosis and a reduction of heart function in rodents. As intervention using medium-chain triglycerides (MCTs) was recently shown to be beneficial in murine cardiac reperfusion injury, the question arises as to what extent the fatty acid (FA)-composition in a KD alters molecular markers of FA-oxidation (FAO) and modulates cardiac fibrotic outcome. METHODS: The effects of LCT-KD as well as an LCT/MCT mix (8:1 ketogenic ratio) on cardiac tissue integrity and the plasma metabolome were assessed in adult male C57/BL6NRJ mice after eight weeks on the respective diet. RESULTS: Both KDs resulted in increased amount of collagen fibers and cardiac tissue was immunologically indistinguishable between groups. MCT supplementation resulted in i) profound changes in plasma metabolome, ii) reduced hydroxymethylglutaryl-CoA synthase upregulation, and mitofusin 2 downregulation, iii) abrogation of LCT-induced mitochondrial enlargement, and iv) enhanced FAO profile. Contrary to literature, mitochondrial biogenesis was unaffected by KDs. We propose that the observed tissue remodeling is caused by the accumulation of 4-hydroxy-2-nonenal protein adducts, despite an inconspicuous nuclear factor (erythroid-derived 2)-like 2 pathway. CONCLUSION: We conclude that regardless of the generally favorable effects of MCTs, they cannot inhibit 4-hydroxy-2-nonenal adduct formation and fibrotic tissue formation in this setting. Furthermore, we support the burgeoning concern about the effect of KDs on the cardiac safety profile.


Sujet(s)
Régime cétogène , Cardiopathies , Mâle , Souris , Animaux , Régime cétogène/effets indésirables , Régime cétogène/méthodes , Triglycéride/métabolisme , Acides gras , Fibrose
4.
Mol Cancer ; 21(1): 89, 2022 03 30.
Article de Anglais | MEDLINE | ID: mdl-35354467

RÉSUMÉ

BACKGROUND: Frequent truncation mutations of the histone lysine N-methyltransferase KMT2C have been detected by whole exome sequencing studies in various cancers, including malignancies of the prostate. However, the biological consequences of these alterations in prostate cancer have not yet been elucidated. METHODS: To investigate the functional effects of these mutations, we deleted the C-terminal catalytic core motif of Kmt2c specifically in mouse prostate epithelium. We analysed the effect of Kmt2c SET domain deletion in a Pten-deficient PCa mouse model in vivo and of truncation mutations of KMT2C in a large number of prostate cancer patients. RESULTS: We show here for the first time that impaired KMT2C methyltransferase activity drives proliferation and PIN formation and, when combined with loss of the tumour suppressor PTEN, triggers loss of senescence, metastatic dissemination and dramatically reduces life expectancy. In Kmt2c-mutated tumours we show enrichment of proliferative MYC gene signatures and loss of expression of the cell cycle repressor p16INK4A. In addition, we observe a striking reduction in disease-free survival of patients with KMT2C-mutated prostate cancer. CONCLUSIONS: We identified truncating events of KMT2C as drivers of proliferation and PIN formation. Loss of PTEN and KMT2C in prostate cancer results in loss of senescence, metastatic dissemination and reduced life expectancy. Our data demonstrate the prognostic significance of KMT2C mutation status in prostate cancer patients. Inhibition of the MYC signalling axis may be a viable treatment option for patients with KMT2C truncations and therefore poor prognosis.


Sujet(s)
Methyltransferases , Tumeurs de la prostate , Animaux , Inhibiteur p16 de kinase cycline-dépendante/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Protéines de liaison à l'ADN/physiologie , Humains , Mâle , Methyltransferases/génétique , Souris , Mutation , Tumeurs de la prostate/métabolisme ,
5.
Oncogene ; 40(6): 1091-1105, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33323974

RÉSUMÉ

Metastatic melanoma is hallmarked by its ability of phenotype switching to more slowly proliferating, but highly invasive cells. Here, we tested the impact of signal transducer and activator of transcription 3 (STAT3) on melanoma progression in association with melanocyte inducing transcription factor (MITF) expression levels. We established a mouse melanoma model for deleting Stat3 in melanocytes with specific expression of human hyperactive NRASQ61K in an Ink4a-deficient background, two frequent driver mutations in human melanoma. Mice devoid of Stat3 showed early disease onset with higher proliferation in primary tumors, but displayed significantly diminished lung, brain, and liver metastases. Whole-genome expression profiling of tumor-derived cells also showed a reduced invasion phenotype, which was further corroborated by 3D melanoma model analysis. Notably, loss or knockdown of STAT3 in mouse or human cells resulted in the upregulation of MITF and induction of cell proliferation. Mechanistically we show that STAT3-induced CAAT Box Enhancer Binding Protein (CEBP) expression was sufficient to suppress MITF transcription. Epigenetic analysis by ATAC-seq confirmed that CEBPa/b binding to the MITF enhancer region silenced the MITF locus. Finally, by classification of patient-derived melanoma samples, we show that STAT3 and MITF act antagonistically and hence contribute differentially to melanoma progression. We conclude that STAT3 is a driver of the metastatic process in melanoma and able to antagonize MITF via direct induction of CEBP family member transcription.


Sujet(s)
Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Mélanome/génétique , Facteur de transcription associé à la microphtalmie/génétique , Facteur de transcription STAT-3/génétique , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mélanocytes/effets des médicaments et des substances chimiques , Mélanome/anatomopathologie , Souris , Métastase tumorale , Transduction du signal/effets des médicaments et des substances chimiques
7.
Leukemia ; 33(3): 696-709, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30131584

RÉSUMÉ

TYK2 is a member of the JAK family of tyrosine kinases that is involved in chromosomal translocation-induced fusion proteins found in anaplastic large cell lymphomas (ALCL) that lack rearrangements activating the anaplastic lymphoma kinase (ALK). Here we demonstrate that TYK2 is highly expressed in all cases of human ALCL, and that in a mouse model of NPM-ALK-induced lymphoma, genetic disruption of Tyk2 delays the onset of tumors and prolongs survival of the mice. Lymphomas in this model lacking Tyk2 have reduced STAT1 and STAT3 phosphorylation and reduced expression of Mcl1, a pro-survival member of the BCL2 family. These findings in mice are mirrored in human ALCL cell lines, in which TYK2 is activated by autocrine production of IL-10 and IL-22 and by interaction with specific receptors expressed by the cells. Activated TYK2 leads to STAT1 and STAT3 phosphorylation, activated expression of MCL1 and aberrant ALCL cell survival. Moreover, TYK2 inhibitors are able to induce apoptosis in ALCL cells, regardless of the presence or absence of an ALK-fusion. Thus, TYK2 is a dependency that is required for ALCL cell survival through activation of MCL1 expression. TYK2 represents an attractive drug target due to its essential enzymatic domain, and TYK2-specific inhibitors show promise as novel targeted inhibitors for ALCL.


Sujet(s)
Lymphome à grandes cellules anaplasiques/génétique , Protéine Mcl-1/génétique , Facteur de transcription STAT-1/génétique , TYK2 Kinase/génétique , Kinase du lymphome anaplasique/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/génétique , Humains , Lymphome à grandes cellules anaplasiques/traitement médicamenteux , Souris , Phosphorylation/effets des médicaments et des substances chimiques , Phosphorylation/génétique , Inhibiteurs de protéines kinases/pharmacologie , Protein-tyrosine kinases/génétique , Facteur de transcription STAT-3/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Translocation génétique/effets des médicaments et des substances chimiques , Translocation génétique/génétique
8.
Int J Cancer ; 143(11): 2943-2954, 2018 12 01.
Article de Anglais | MEDLINE | ID: mdl-29987839

RÉSUMÉ

Persistent activation of hedgehog (HH)/GLI signaling accounts for the development of basal cell carcinoma (BCC), a very frequent nonmelanoma skin cancer with rising incidence. Targeting HH/GLI signaling by approved pathway inhibitors can provide significant therapeutic benefit to BCC patients. However, limited response rates, development of drug resistance, and severe side effects of HH pathway inhibitors call for improved treatment strategies such as rational combination therapies simultaneously inhibiting HH/GLI and cooperative signals promoting the oncogenic activity of HH/GLI. In this study, we identified the interleukin-6 (IL6) pathway as a novel synergistic signal promoting oncogenic HH/GLI via STAT3 activation. Mechanistically, we provide evidence that signal integration of IL6 and HH/GLI occurs at the level of cis-regulatory sequences by co-binding of GLI and STAT3 to common HH-IL6 target gene promoters. Genetic inactivation of Il6 signaling in a mouse model of BCC significantly reduced in vivo tumor growth by interfering with HH/GLI-driven BCC proliferation. Our genetic and pharmacologic data suggest that combinatorial HH-IL6 pathway blockade is a promising approach to efficiently arrest cancer growth in BCC patients.


Sujet(s)
Carcinome basocellulaire/métabolisme , Carcinome basocellulaire/anatomopathologie , Protéines Hedgehog/métabolisme , Interleukine-6/métabolisme , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie , Animaux , Carcinogenèse/métabolisme , Prolifération cellulaire/physiologie , Humains , Souris , Souris transgéniques , Transduction du signal/physiologie , Transactivateurs/métabolisme
9.
Blood ; 131(12): 1337-1349, 2018 03 22.
Article de Anglais | MEDLINE | ID: mdl-29352038

RÉSUMÉ

Chronic lymphocytic leukemia (CLL) outgrowth depends on signals from the microenvironment. We have previously found that in vitro reconstitution of this microenvironment induces specific variant isoforms of the adhesion molecule CD44, which confer human CLL with high affinity to hyaluronan (HA). Here, we determined the in vivo contribution of standard CD44 and its variants to leukemic B-cell homing and proliferation in Tcl1 transgenic mice with a B-cell-specific CD44 deficiency. In these mice, leukemia onset was delayed and leukemic infiltration of spleen, liver, and lungs, but not of bone marrow, was decreased. Competitive transplantation revealed that CLL homing to spleen and bone marrow required functional CD44. Notably, enrichment of CD44v6 variants particularly in spleen enhanced CLL engraftment and proliferation, along with increased HA binding. We recapitulated CD44v6 induction in the human disease and revealed the involvement of MAPK and NF-κB signaling upon CD40 ligand and B-cell receptor stimulation by in vitro inhibition experiments and chromatin immunoprecipitation assays. The investigation of downstream signaling after CD44v6-HA engagement uncovered the activation of extracellular signal-regulated kinase and p65. Consequently, anti-CD44v6 treatment reduced leukemic cell proliferation in vitro in human and mouse, confirming the general nature of the findings. In summary, we propose a CD44-NF-κB-CD44v6 circuit in CLL, allowing tumor cells to gain HA binding capacity and supporting their proliferation.


Sujet(s)
Prolifération cellulaire , Antigènes CD44/métabolisme , Leucémie chronique lymphocytaire à cellules B/métabolisme , Système de signalisation des MAP kinases , Protéines tumorales/métabolisme , Microenvironnement tumoral , Animaux , Humains , Antigènes CD44/génétique , Acide hyaluronique/génétique , Acide hyaluronique/métabolisme , Leucémie chronique lymphocytaire à cellules B/génétique , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Souris , Souris transgéniques , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Protéines tumorales/génétique , Récepteurs pour l'antigène des lymphocytes B/génétique , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Rate/métabolisme , Rate/anatomopathologie
10.
Int J Cancer ; 142(5): 968-975, 2018 03 01.
Article de Anglais | MEDLINE | ID: mdl-29055107

RÉSUMÉ

Aberrant activation of Hedgehog (HH)/GLI signaling is causally involved in numerous human malignancies, including basal cell carcinoma (BCC) and medulloblastoma. HH pathway antagonists targeting smoothened (SMO), an essential effector of canonical HH/GLI signaling, show significant clinical success in BCC patients and have recently been approved for the treatment of advanced and metastatic BCC. However, rapid and frequent development of drug resistance to SMO inhibitors (SMOi) together with severe side effects caused by prolonged SMOi treatment call for alternative treatment strategies targeting HH/GLI signaling downstream of SMO. In this study, we report that 4SC-202, a novel clinically validated inhibitor of class I histone deacetylases (HDACs), efficiently blocks HH/GLI signaling. Notably, 4SC-202 treatment abrogates GLI activation and HH target gene expression in both SMOi-sensitive and -resistant cells. Mechanistically, we propose that the inhibition of HDACs 1/2/3 is crucial for targeting oncogenic HH/GLI signaling, and that class I HDAC inhibitors either in combination with SMOi or as second-line therapy may improve the treatment options for HH-associated malignancies with SMOi resistance.


Sujet(s)
Benzamides/pharmacologie , Carcinome basocellulaire/traitement médicamenteux , Résistance aux médicaments antinéoplasiques , Protéines Hedgehog/antagonistes et inhibiteurs , Histone deacetylases/composition chimique , Récepteur Smoothened/antagonistes et inhibiteurs , Protéine à doigt de zinc GLI1/antagonistes et inhibiteurs , Animaux , Apoptose , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Carcinome basocellulaire/métabolisme , Carcinome basocellulaire/anatomopathologie , Prolifération cellulaire , Protéines Hedgehog/métabolisme , Histone deacetylases/métabolisme , Humains , Souris , Souris de lignée NOD , Souris SCID , Transduction du signal , Récepteur Smoothened/métabolisme , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe , Protéine à doigt de zinc GLI1/métabolisme
11.
Cell Commun Signal ; 15(1): 8, 2017 01 25.
Article de Anglais | MEDLINE | ID: mdl-28122581

RÉSUMÉ

Treatment of acute myeloid leukemia (AML), an aggressive and heterogeneous hematological malignancy, remains a challenge. Despite advances in our understanding of the complex genetics and biology of AML pathophysiology, these findings have been translated to the clinic with only limited success, and poor outcomes persist for the majority of patients. Thus, novel treatment strategies are clearly needed for achieving deeper and prolonged remissions and for avoiding the development of resistance. Due to its profound role in (cancer) stem cell biology and differentiation, the Hedgehog (HH)/Glioma-associated Oncogene Homolog (GLI) signaling pathway may be an attractive novel therapeutic target in AML. In this review, we aim to provide a critical and concise overview of the currently known potential and challenges of HH/GLI targeting. We describe the biological role of the HH/GLI pathway in AML pathophysiology. We specifically focus on ways of targeting non-canonical HH/GLI signaling in AML, particularly in combination with standard treatment regimens, which may overcome some hurdles observed with approved HH pathway inhibitors in solid tumors.


Sujet(s)
Protéines Hedgehog/métabolisme , Leucémie aigüe myéloïde/métabolisme , Thérapie moléculaire ciblée , Transduction du signal , Protéine à doigt de zinc GLI1/métabolisme , Animaux , Humains , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie
12.
Cancer Res ; 76(8): 2186-96, 2016 04 15.
Article de Anglais | MEDLINE | ID: mdl-26837762

RÉSUMÉ

The proliferation of chronic lymphocytic leukemia (CLL) cells requires communication with the lymphoid organ microenvironment. Integrin-linked kinase (ILK) is a multifunctional intracellular adaptor protein that transmits extracellular signals to regulate malignant cell motility, metastasis, and cell-cycle progression, but is poorly characterized in hematologic malignancies. In this study, we investigated the role of ILK in the context of CLL and observed high ILK expression in patient samples, particularly in tumor cells harboring prognostic high-risk markers such as unmutated IGHV genes, high Zap70, or CD38 expression, or a signature of recent proliferation. We also found increased numbers of Ki67 (MKI67)-positive cells in regions of enhanced ILK expression in lymph nodes from CLL patients. Using coculture conditions mimicking the proliferative lymph node microenvironment, we detected a parallel induction of ILK and cyclin D1 (CCND1) expression in CLL cells that was dependent on the activation of NF-κB signaling by soluble TNFα. The newly synthesized ILK protein colocalized to centrosomal structures and was required for correct centrosome clustering and mitotic spindle organization. Furthermore, we established a mouse model of CLL in which B-cell-specific genetic ablation of ILK resulted in decelerated leukemia development due to reduced organ infiltration and proliferation of CLL cells. Collectively, our findings describe a TNFα-NF-κB-mediated mechanism by which ILK expression is induced in the lymph node microenvironment and propose that ILK promotes leukemogenesis by enabling CLL cells to cope with centrosomal defects acquired during malignant transformation. Cancer Res; 76(8); 2186-96. ©2016 AACR.


Sujet(s)
Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Tissu lymphoïde/enzymologie , Facteur de transcription NF-kappa B/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Animaux , Prolifération cellulaire , Humains , Leucémie chronique lymphocytaire à cellules B/enzymologie , Leucémie chronique lymphocytaire à cellules B/métabolisme , Tissu lymphoïde/anatomopathologie , Souris , Souris transgéniques , Pronostic , Protein-Serine-Threonine Kinases/génétique , Transduction du signal
13.
Oncotarget ; 7(6): 7134-48, 2016 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-26784250

RÉSUMÉ

A wide range of human malignancies displays aberrant activation of Hedgehog (HH)/GLI signaling, including cancers of the skin, brain, gastrointestinal tract and hematopoietic system. Targeting oncogenic HH/GLI signaling with small molecule inhibitors of the essential pathway effector Smoothened (SMO) has shown remarkable therapeutic effects in patients with advanced and metastatic basal cell carcinoma. However, acquired and de novo resistance to SMO inhibitors poses severe limitations to the use of SMO antagonists and urgently calls for the identification of novel targets and compounds.Here we report on the identification of the Dual-Specificity-Tyrosine-Phosphorylation-Regulated Kinase 1B (DYRK1B) as critical positive regulator of HH/GLI signaling downstream of SMO. Genetic and chemical inhibition of DYRK1B in human and mouse cancer cells resulted in marked repression of HH signaling and GLI1 expression, respectively. Importantly, DYRK1B inhibition profoundly impaired GLI1 expression in both SMO-inhibitor sensitive and resistant settings. We further introduce a novel small molecule DYRK1B inhibitor, DYRKi, with suitable pharmacologic properties to impair SMO-dependent and SMO-independent oncogenic GLI activity. The results support the use of DYRK1B antagonists for the treatment of HH/GLI-associated cancers where SMO inhibitors fail to demonstrate therapeutic efficacy.


Sujet(s)
Carcinome basocellulaire/anatomopathologie , Résistance aux médicaments antinéoplasiques , Protéines Hedgehog/métabolisme , Tumeurs du pancréas/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-tyrosine kinases/métabolisme , Récepteur Smoothened/métabolisme , Protéine à doigt de zinc GLI1/métabolisme , Animaux , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Carcinome basocellulaire/traitement médicamenteux , Carcinome basocellulaire/génétique , Carcinome basocellulaire/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Facteurs de transcription Forkhead/physiologie , Protéines Hedgehog/antagonistes et inhibiteurs , Protéines Hedgehog/génétique , Humains , Souris , Souris nude , Cellules NIH 3T3 , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/génétique , ARN messager/génétique , Petit ARN interférent/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie , Récepteur Smoothened/antagonistes et inhibiteurs , Récepteur Smoothened/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Protéine à doigt de zinc GLI1/antagonistes et inhibiteurs , Protéine à doigt de zinc GLI1/génétique ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...