Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892054

RÉSUMÉ

MicroRNAs (miRNAs) are small RNA molecules that regulate more than 30% of genes in humans. Recent studies have revealed that miRNAs play a crucial role in tumorigenesis. Large sets of miRNAs in human tumors are under-expressed compared to normal tissues. Furthermore, experiments have shown that interference with miRNA processing enhances tumorigenesis. Multiple studies have documented the causal role of miRNAs in cancer, and miRNA-based anticancer therapies are currently being developed. This review primarily focuses on two key points: (1) miRNAs and their role in human cancer and (2) the regulation of tumor suppressors by miRNAs. The review discusses (a) the regulation of the tumor suppressor p53 by miRNA, (b) the critical role of the miR-144/451 cluster in regulating the Itch-p63-Ago2 pathway, and (c) the regulation of PTEN by miRNAs. Future research and the perspectives of miRNA in cancer are also discussed. Understanding these pathways will open avenues for therapeutic interventions targeting miRNA regulation.


Sujet(s)
Régulation de l'expression des gènes tumoraux , microARN , Tumeurs , Humains , microARN/génétique , microARN/métabolisme , Tumeurs/génétique , Tumeurs/thérapie , Tumeurs/métabolisme , Animaux , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
2.
Mol Ther Nucleic Acids ; 31: 466-481, 2023 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-36865087

RÉSUMÉ

The tumor suppressor p53 plays a critical role in cancer pathogenesis, and regulation of p53 expression is essential for maintaining normal cell growth. UBE4B is an E3/E4 ubiquitin ligase involved in a negative-feedback loop with p53. UBE4B is required for Hdm2-mediated p53 polyubiquitination and degradation. Thus, targeting the p53-UBE4B interactions is a promising anticancer strategy for cancer therapy. In this study, we confirm that while the UBE4B U box does not bind to p53, it is essential for the degradation of p53 and acts in a dominant-negative manner, thereby stabilizing p53. C-terminal UBE4B mutants lose their ability to degrade p53. Notably, we identified one SWIB/Hdm2 motif of UBE4B that is vital for p53 binding. Furthermore, the novel UBE4B peptide activates p53 functions, including p53-dependent transactivation and growth inhibition, by blocking the p53-UBE4B interactions. Our findings indicate that targeting the p53-UBE4B interaction presents a novel approach for p53 activation therapy in cancer.

3.
Exp Mol Med ; 54(10): 1658-1669, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-36207426

RÉSUMÉ

Antitumor therapeutic strategies that fundamentally rely on the induction of DNA damage to eradicate and inhibit the growth of cancer cells are integral approaches to cancer therapy. Although DNA-damaging therapies advance the battle with cancer, resistance, and recurrence following treatment are common. Thus, searching for vulnerabilities that facilitate the action of DNA-damaging agents by sensitizing cancer cells is an active research area. Therefore, it is crucial to decipher the detailed molecular events involved in DNA damage responses (DDRs) to DNA-damaging agents in cancer. The tumor suppressor p53 is active at the hub of the DDR. Researchers have identified an increasing number of genes regulated by p53 transcriptional functions that have been shown to be critical direct or indirect mediators of cell fate, cell cycle regulation, and DNA repair. Posttranslational modifications (PTMs) primarily orchestrate and direct the activity of p53 in response to DNA damage. Many molecules mediating PTMs on p53 have been identified. The anticancer potential realized by targeting these molecules has been shown through experiments and clinical trials to sensitize cancer cells to DNA-damaging agents. This review briefly acknowledges the complexity of DDR pathways/networks. We specifically focus on p53 regulators, protein kinases, and E3/E4 ubiquitin ligases and their anticancer potential.


Sujet(s)
Tumeurs , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Altération de l'ADN , Réparation de l'ADN , Protein kinases/métabolisme , Ubiquitin-protein ligases/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/génétique
4.
Cells ; 11(9)2022 04 30.
Article de Anglais | MEDLINE | ID: mdl-35563821

RÉSUMÉ

Hepatoblastoma (HB) is a rare primary malignancy of the developing fetal liver. Its course is profoundly influenced by genetics, in the context of sporadic mutation or genetic syndromes. Conventionally, subtypes of HB are histologically determined based on the tissue type that is recapitulated by the tumor and the direction of its differentiation. This classification is being reevaluated based on advances on molecular pathology. The therapeutic approach comprises surgical intervention, chemotherapy (in a neoadjuvant or post-operative capacity), and in some cases, liver transplantation. Although diagnostic modalities and treatment options are evolving, some patients experience complications, including relapse, metastatic spread, and suboptimal response to chemotherapy. As yet, there is no consistent framework with which such outcomes can be predicted. N6-methyladenosine (m6A) is an RNA modification with rampant involvement in the normal processing of cell metabolism and neoplasia. It has been observed to impact the development of a variety of cancers via its governance of gene expression. M6A-associated genes appear prominently in HB. Literature data seem to underscore the role of m6A in promotion and clinical course of HB. Illuminating the pathogenetic mechanisms that drive HB are promising additions to the understanding of the clinically aggressive tumor behavior, given its potential to predict disease course and response to therapy. Implicated genes may also act as targets to facilitate the evolving personalized cancer therapy. Here, we explore the role of m6A and its genetic associates in the promotion of HB, and the impact this may have on the management of this neoplastic disease.


Sujet(s)
Hépatoblastome , Tumeurs du foie , Transplantation hépatique , Adénosine/analogues et dérivés , Hépatoblastome/traitement médicamenteux , Hépatoblastome/génétique , Hépatoblastome/anatomopathologie , Humains , Tumeurs du foie/anatomopathologie , Récidive tumorale locale
5.
Cell Death Dis ; 13(4): 397, 2022 04 22.
Article de Anglais | MEDLINE | ID: mdl-35459267

RÉSUMÉ

As a key component of the RNA-induced silencing complex (RISC), Argonaute2 (Ago2) exhibits a dual function regulatory role in tumor progression. However, the mechanistic basis of differential regulation remains elusive. p63 is a homolog of the tumor suppressor p53. p63 isoforms play a critical role in tumorigenesis and metastasis. Herein, we show that p63 isoforms physically interact with and stabilize Ago2. Expression of p63 isoforms increases the levels of Ago2 protein, while depletion of p63 isoforms by shRNA decreases Ago2 protein levels. p63 strongly guides Ago2 dual functions in vitro and in vivo. Ectopic expression of the miR-144/451 cluster increases p63 protein levels; TAp63 transactivates the miR-144/451 cluster, forming a positive feedback loop. Notably, miR-144 activates p63 by directly targeting Itch, an E3 ligase of p63. Ectopic expression of miR-144 induces apoptosis in H1299 cells. miR-144 enhances TAp63 tumor suppressor function and inhibits cell invasion. Our findings uncover a novel function of p63 linking the miRNA-144 cluster and the Ago2 pathway. FACTS AND QUESTIONS: Identification of Ago2 as a p63 target. Ago2 exhibits a dual function regulatory role in tumor progression; however, the molecular mechanism of Ago2 regulation remains unknown. p63 strongly guides Ago2 dual functions in vitro and in vivo. Unraveling a novel function of p63 links the miRNA-144 cluster and the Ago2 pathway.


Sujet(s)
microARN , Tumeurs , Protéines Argonaute , Humains , microARN/génétique , microARN/métabolisme , Tumeurs/métabolisme , Isoformes de protéines/métabolisme , Complexe réprimant l'expression de l'ARN/métabolisme , Facteurs de transcription , Protéines suppresseurs de tumeurs , Ubiquitin-protein ligases/métabolisme
6.
Cancer Lett ; 527: 41-48, 2022 02 28.
Article de Anglais | MEDLINE | ID: mdl-34902523

RÉSUMÉ

Triple-negative breast cancer (TNBC) is a subtype of breast cancer (BCa) in which estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2) are not expressed. Although TNBC cases account for approximately 15% of all BCa cases, TNBC patients' prognosis is poor compared with that of other BCa subtypes. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) plays an important role in cell proliferation and migration by negatively regulating the PI3K/Akt pathway. PTEN is one of the most commonly inactivated tumor suppressors in BCa. PTEN inactivity is associated with larger tumor sizes, multiple lymph node metastases, and an aggressive triple-negative phenotype. This review primarily focuses on two key points: (1) PTEN and its function. (2) The regulation of tumor suppressor PTEN in TNBC. We provide a summary of genomic alterations of PTEN in BCa. We further discuss the transcriptional regulation of PTEN and how PTEN is regulated by posttranscription and posttranslational modification, as well as by protein interactions. Finally, we discuss the perspectives of the PTEN protein in TNBC.


Sujet(s)
Génomique/méthodes , Phosphohydrolase PTEN/métabolisme , Tumeurs du sein triple-négatives/génétique , Femelle , Humains
8.
Nucleic Acids Res ; 49(5): 2740-2758, 2021 03 18.
Article de Anglais | MEDLINE | ID: mdl-33619536

RÉSUMÉ

The major clinical problem in human cancer is metastasis. Metastases are the cause of 90% of human cancer deaths. TAp63 is a critical suppressor of tumorigenesis and metastasis. ΔNp63 acts as a dominant-negative inhibitor to block the function of p53 and TAp63. Although several ubiquitin E3 ligases have been reported to regulate p63 stability, the mechanism of p63 regulation remains partially understood. Herein, we show that CHIP, an E3 ligase with a U-box domain, physically interacts with p63 and promotes p63 degradation. Notably, Hsp70 depletion by siRNA stabilizes TAp63 in H1299 cells and destabilizes ΔNp63 in SCC9 cells. Loss of Hsp70 results in a reduction in the TAp63-CHIP interaction in H1299 cells and an increase in the interaction between ΔNp63 and CHIP in SCC9 cells. Our results reveal that Hsp70 acts as a molecular switch to control CHIP-mediated ubiquitination and degradation of p63 isoforms. Furthermore, regulation of p63 by the Hsp70-CHIP axis contributes to the migration and invasion of tumor cells. Hence, our findings demonstrate that Hsp70 is a crucial regulator of CHIP-mediated ubiquitination and degradation of p63 isoforms and identify a new pathway for maintaining TAp63 or ΔNp63 stability in cancers.


Sujet(s)
Protéines du choc thermique HSP70/métabolisme , Facteurs de transcription/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination , Animaux , Apoptose , Lignée cellulaire tumorale , Mouvement cellulaire , Cellules cultivées , Protéines du choc thermique HSP70/physiologie , Humains , Souris , Invasion tumorale , Tumeurs/métabolisme , Tumeurs/mortalité , Transactivateurs/métabolisme , Activation de la transcription
9.
Carcinogenesis ; 42(4): 650-662, 2021 04 30.
Article de Anglais | MEDLINE | ID: mdl-33569599

RÉSUMÉ

Pirh2 is an E3 ligase belonging to the RING-H2 family and shown to bind, ubiquitinate and downregulate p73 tumor suppressor function without altering p73 protein levels. AIP4, an E3 ligase belonging to the HECT domain family, has been reported to be a negative regulatory protein that promotes p73 ubiquitination and degradation. Herein, we found that Pirh2 is a key regulator of AIP4 that inhibits p73 function. Pirh2 physically interacts with AIP4 and significantly downregulates AIP4 expression. This downregulation is shown to involve the ubiquitination of AIP4 by Pirh2. Importantly, we demonstrated that the ectopic expression of Pirh2 inhibits the AIP4-p73 negative regulatory pathway, which was restored when depleting endogenous Pirh2 utilizing Pirh2-siRNAs. We further observed that Pirh2 decreases AIP4-mediated p73 ubiquitination. At the translational level and specifically regarding p73 cell cycle arrest function, Pirh2 still ensures the arrest of p73-mediated G1 despite AIP4 expression. Our study reveals a novel link between two E3 ligases previously thought to be unrelated in regulating the same effector substrate, p73. These findings open a gateway to explain how E3 ligases differentiate between regulating multiple substrates that may belong to the same family of proteins, as it is the case for the p53 and p73 proteins.


Sujet(s)
Protéines de répression/génétique , Protéine tumorale p73/génétique , Ubiquitin-protein ligases/génétique , Ubiquitination/génétique , Points de contrôle du cycle cellulaire/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes/génétique , Humains , Liaison aux protéines/génétique
10.
J Cancer ; 11(17): 5007-5023, 2020.
Article de Anglais | MEDLINE | ID: mdl-32742448

RÉSUMÉ

Osteosarcoma affects both adolescents and adults, and some improvement in the survival rate for affected patients has been reached in the last decade. Still, non-specificity and systemic toxicity may limit traditional therapeutic approaches to some extent. The insulin growth factor 1 (IGF1) and its binding protein (IGFBP3) have been implicated in the tumorigenesis. Nanoparticles, such as graphene oxide (GO), can provide an effective treatment for cancer as they can specifically target cancer cells while reducing undesired side effects. This study aimed to evaluate the toxicity of GO on osteosarcoma in vitro using tumor cell lines with and without knocking out the IGF and IGFBP3 genes. Human osteosarcoma cell lines, U2OS and SAOS2, and the normal osteoblast cell line hFOB1.19 were used. The IGF1 and IGFBP3 genes were eliminated using CRISPR/Cas9. Tumor cells were cultured and treated with GO. Apoptosis and reactive oxygen species (ROS) were analyzed by Annexin V-FITC and ROS assays. The nuclear factor erythroid 2-related factor 2 (NRF2), which is a crucial regulator of cellular resistance to oxidants, was investigated by Western blotting. We found a significantly higher rate of apoptosis in the OS than hFOB1.19, especially in U2OS cells in which IGF1 and IGFBP3 were knocked out. ROS increase due to GO exposure was remarkably time and concentration-dependent. Based on the rate of apoptosis, ROS, Nrf-2 decrease, and cytomorphological changes, GO has a significant cytotoxic effect against OS. Targeting the IGF1 and IGFBP3 signaling pathway may strengthen GO-related cytotoxicity with the potential to increase the survival of patients affected by this tumor.

11.
Carcinogenesis ; 39(9): 1185-1196, 2018 09 21.
Article de Anglais | MEDLINE | ID: mdl-29985991

RÉSUMÉ

Triple negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis and high mortality rate. The tumor suppressor phosphatase and tensin homolog deleted on chromosome 10 (PTEN) plays an important role in cell proliferation and cell migration by negatively regulating the PI3K/Akt pathway. PTEN is downregulated by microRNAs in multiple cancers. However, few microRNAs have been reported to directly target PTEN in TNBC. In this study, microRNAs predicted to target PTEN were screened by immunoblotting and luciferase reporter assays. Expression levels of microRNA-498 (miR-498) were measured by TaqMan microRNA assays. We performed clonogenic, cell cycle and scratch wound assays to examine the oncogenic role of miR-498. We demonstrated that miR-498 directly targeted the 3'untranslated region of PTEN mRNA and reduced PTEN protein levels in TNBC cells. Compared with the non-tumorigenic breast epithelial cell line MCF-10A, TNBC cell lines overexpressed miR-498. Moreover, miR-498 promoted cell proliferation and cell cycle progression in TNBC cells in a PTEN-dependent manner. Suppressing miR-498 overexpression impaired the oncogenic effects of miR-498 on cell proliferation and cell migration. This study identified a novel microRNA (miR-498) overexpressed in TNBC cells and its oncogenic role in suppressing PTEN. These results provide new insight into the downregulation of PTEN and indicate a potential therapeutic target for treating TNBC.


Sujet(s)
Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , microARN/génétique , Phosphohydrolase PTEN/génétique , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Régions 3' non traduites/génétique , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Régulation négative/génétique , Cellules HEK293 , Humains , Cellules MCF-7 , Phosphohydrolase PTEN/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme
12.
Cancer Lett ; 401: 20-32, 2017 08 10.
Article de Anglais | MEDLINE | ID: mdl-28483517

RÉSUMÉ

The p53 protein plays a critical role in preventing tumor development. Although numerous factors have been shown to directly or indirectly regulate p53, the mechanism of how microRNAs (miRNAs) modulate p53 remains unclear. Here, we identified miR-1301, a microRNA that regulates the activity and function of p53, by directly targeting the ubiquitination factor E4B (UBE4B), an E3 and E4 ubiquitin ligase. Notably, ectopic expression of miR-1301 inhibits dissemination and metastasis of tumor cells in a p53-dependent manner. Depletion of miR-1301 downregulates p53 function. Our results reveal that there is an inverse correlation between miR-1301 and UBE4B expression and p53 status in prostate cancer. Furthermore, low miR-1301 expression is often associated with incomplete methylation of its gene in human prostate tumors. Together, our results provide the first report indicating that miR-1301 functions as a tumor suppressor that inhibits tumor cell migration and invasion in multiple human cancer cells by regulating the UBE4B-p53 pathway.


Sujet(s)
Mouvement cellulaire , microARN/métabolisme , Tumeurs/enzymologie , Protéine p53 suppresseur de tumeur/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Ubiquitin-protein ligase complexes/métabolisme , Apoptose , Cycle cellulaire , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Cellules HCT116 , Humains , microARN/génétique , Invasion tumorale , Tumeurs/génétique , Tumeurs/anatomopathologie , Régions promotrices (génétique) , Transduction du signal , Transfection , Protéine p53 suppresseur de tumeur/génétique , Protéines suppresseurs de tumeurs/génétique , Ubiquitin-protein ligase complexes/génétique , Ubiquitin-protein ligases
13.
Int J Mol Sci ; 17(12)2016 Dec 06.
Article de Anglais | MEDLINE | ID: mdl-27929429

RÉSUMÉ

The protein p63 has been identified as a homolog of the tumor suppressor protein p53 and is capable of inducing apoptosis, cell cycle arrest, or senescence. p63 has at least six isoforms, which can be divided into two major groups: the TAp63 variants that contain the N-terminal transactivation domain and the ΔNp63 variants that lack the N-terminal transactivation domain. The TAp63 variants are generally considered to be tumor suppressors involved in activating apoptosis and suppressing metastasis. ΔNp63 variants cannot induce apoptosis but can act as dominant negative inhibitors to block the function of TAp53, TAp73, and TAp63. p63 is rarely mutated in human tumors and is predominately regulated at the post-translational level by phosphorylation and ubiquitination. This review focuses primarily on regulation of p63 by the ubiquitin E-3 ligase family of enzymes via ubiquitination and proteasome-mediated degradation, and introduces a new key regulator of the p63 protein.


Sujet(s)
Protéines membranaires/métabolisme , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme , Animaux , Protéines du cycle cellulaire , Humains , Protéines membranaires/génétique , Protéines nucléaires/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination
14.
Oncotarget ; 7(3): 2823-36, 2016 Jan 19.
Article de Anglais | MEDLINE | ID: mdl-26673821

RÉSUMÉ

Phosphorylation of p53 is a key mechanism responsible for the activation of its tumor suppressor functions in response to various stresses. In unstressed cells, p53 is rapidly turned over and is maintained at a low basal level. After DNA damage or other forms of cellular stress, the p53 level increases, and the protein becomes metabolically stable. However, the mechanism of phosphorylated p53 regulation is unclear. In this study, we studied the kinetics of UBE4B, Hdm2, Pirh2, Cop1 and CHIP induction in response to p53 activation. We show that UBE4B coimmunoprecipitates with phosphorylated p53 at serines 15 and 392. Notably, the affinity between UBE4B and Hdm2 is greatly decreased after DNA damage. Furthermore, we observe that UBE4B promotes endogenous phospho-p53(S15) and phospho-p53(S392) degradation in response to IR. We demonstrate that UBE4B and Hdm2 repress p53S15A, p53S392A, and p53-2A(S15A, S392A) functions, including p53-dependent transactivation and growth inhibition. Overall, our results reveal that UBE4B plays an important role in regulating phosphorylated p53 following DNA damage.


Sujet(s)
Protéines proto-oncogènes c-mdm2/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Ubiquitin-protein ligase complexes/métabolisme , Sites de fixation , Lignée cellulaire tumorale , Altération de l'ADN/génétique , Activation enzymatique , Cellules HCT116 , Humains , Cellules MCF-7 , Phosphorylation , Liaison aux protéines , Ubiquitin-protein ligases , Ubiquitination/physiologie
15.
Oncotarget ; 6(25): 21479-92, 2015 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-26025930

RÉSUMÉ

The protein p73, a homologue of the tumor suppressor protein p53, is capable of inducing apoptosis and cell cycle arrest. MDM2 is transcriptionally activated by p73 and represses the functions of p73, including p73-dependent transactivation and growth suppression. However, the molecular mechanism of this repression is unknown. In this study, we show that MDM2 mediates p73 ubiquitination. MDM2 mainly utilizes K11, K29 and K63-linked chains to mediate p73 ubiquitination in vivo and in vitro. However, MDM2 is unable to promote p73 degradation in most tested cell lines. Surprisingly, we observe that overexpression of Mdm2 promotes p73 degradation mainly through Itch in Mdm2-null MEFs. We further find that Itch interacts with the transfected Mdm2 in Mdm2-null cells. Moreover, our findings reveal that the E3 ligase activity of MDM2 is required to repress p73-dependent apoptosis and cell cycle arrest but not p73-dependent transcriptional activity. Furthermore, the data suggest a link between p73 ubiquitination/MDM2 E3 ligase activity and p73 biological functions.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines nucléaires/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Animaux , Apoptose , Cycle cellulaire , Points de contrôle du cycle cellulaire/génétique , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Fibroblastes/métabolisme , Gènes suppresseurs de tumeur , Cellules HEK293 , Humains , Souris , Protéines nucléaires/antagonistes et inhibiteurs , Petit ARN interférent/métabolisme , Protéines de répression/métabolisme , Protéine tumorale p73 , Protéines suppresseurs de tumeurs/antagonistes et inhibiteurs , Ubiquitin-protein ligases/métabolisme , Ubiquitination
16.
Pathol Oncol Res ; 21(4): 901-8, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25740074

RÉSUMÉ

Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (CCA) are the two most common primary liver malignancies in adult patients. The molecular mechanisms underlying the pathogenesis of HCC and CCA are still poorly understood. Sonic hedgehog (SHH) signaling plays an essential role during mammalian development, i.e., promoting organ growth, tissue differentiation, and cell polarity. The upregulation of SHH has been observed during carcinogenesis, including colorectal carcinoma. Our aim was to investigate the expression pattern of SHH in HCC and CCA. We investigated 40 malignant tumors of the liver, including 21 HCC and 19 of intrahepatic CCA cases by immunohistochemistry (IHC) using a polyclonal antibody against SHH and Avidin-Biotin Complex method. We also investigated the co-localization of SHH and Bone morphogenetic protein 4 (BMP4) in CCA using indirect double IHC. Moreover, we examined whether SHH is expressed in two HCC cell lines HepG2 and HuH-7 and three CCA cell lines OZ, HuCCT1 and HuH28. We found that SHH was expressed in 15 out of 21 cases (71.4 %) of HCC and 100 % of CCA cases by immunohistochemistry. SHH expression showed a positive trend in liver tumors (HCC, CCA) with high grade (G2-G3). SHH localized to the epithelial cells, while BMP4 was expressed in the stromal cells in CCA by double IHC. However, both HCC and CCA cell lines showed SHH expression by Western blot analysis. In conclusion, SHH seems to be an interesting marker of de-differentiation in liver tumors and the simultaneous epithelial-mesenchymal expression may be an intriguing prompt to investigate cross-talks between SHH and BMP4.


Sujet(s)
Tumeurs des canaux biliaires/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Carcinome hépatocellulaire/métabolisme , Cholangiocarcinome/métabolisme , Protéines Hedgehog/métabolisme , Tumeurs du foie/métabolisme , Adulte , Tumeurs des canaux biliaires/anatomopathologie , Technique de Western , Carcinome hépatocellulaire/anatomopathologie , Cholangiocarcinome/anatomopathologie , Humains , Techniques immunoenzymatiques , Tumeurs du foie/anatomopathologie , Pronostic , Études rétrospectives , Cellules cancéreuses en culture
17.
PLoS One ; 8(12): e82803, 2013.
Article de Anglais | MEDLINE | ID: mdl-24367557

RÉSUMÉ

Pirh2 is a p53 inducible gene that encodes a RING-H2 domain and is proposed to be a main regulator of p53 protein, thus fine tuning the DNA damage response. Pirh2 interacts physically with p53 and promotes its MDM2-independent ubiquitination and subsequent degradation as well as participates in an auto-regulatory feedback loop that controls p53 function. Pirh2 also self-ubiquitinates. Interestingly, Pirh2 is overexpressed in a wide range of human tumors. In this study, we investigated the domains and residues essential for Pirh2 self-ubiquitination. Deletions were made in each of the three major domains of Pirh2: the N-terminal domain (NTD), Ring domain (RING), and C-terminal domain (CTD). The effects of these deletions on Pirh2 self-ubiquitination were then assessed using in vitro ubiquitination assays. Our results demonstrate that the RING domain is essential, but not sufficient, for Pirh2 self-ubiquitination and that residues 240-250 of the C-terminal domain are also essential. Our results demonstrate that Pirh2 mediated p53 polyubiquitination occurs mainly through the K48 residue of ubiquitin in vitro. Our data further our understanding of the mechanism of Pirh2 self-ubiquitination and may help identify valuable therapeutic targets that play roles in reducing the effects of the overexpression of Pirh2, thus maximizing p53's response to DNA damage.


Sujet(s)
Ubiquitin-protein ligases/composition chimique , Ubiquitin-protein ligases/métabolisme , Lignée cellulaire , Humains , Structure tertiaire des protéines , Ubiquitination
18.
Cell Cycle ; 12(10): 1569-77, 2013 May 15.
Article de Anglais | MEDLINE | ID: mdl-23603988

RÉSUMÉ

Several reports have pointed to the negative involvement of p53 in transcriptional regulation of the human immunodeficiency virus type 1 long-terminal repeat (HIV-1 LTR). We recently demonstrated that through their physical interaction, cdk9 phosphorylates p53 on Ser-392, leading to p53 stability and accumulation. As a result, p53 stalled transcriptional elongation of the HIV-1 LTR and significantly reduced HIV-1 replication in primary microglia and astrocytes. Therefore, we sought to identify the mechanisms used by cdk9 to allow this p53 function. Using western blot analysis, we found that cdk9 promotes inhibition and phosphorylation of Mdm2 on Ser-395, thus preventing degradation of p53, a protein that is directly involved in promoting p53 ubiquitination. On the other hand, we showed that cdk9 phosphorylates Pirh2 on Ser-211 and Thr-217 residues through their physical interaction. Phosphorylation of Pirh2 renders it inactive and may contribute to p53-inhibition of transcriptional elongation of the HIV-1 LTR. Hence, we suggest that phosphorylation of Pirh2 may be a novel target for the inhibition of HIV-1 gene expression.


Sujet(s)
Kinase-9 cycline-dépendante/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Ubiquitin-protein ligases/métabolisme , Séquence d'acides aminés , Lignée cellulaire , Kinase-9 cycline-dépendante/antagonistes et inhibiteurs , Kinase-9 cycline-dépendante/génétique , Répétition terminale longue du VIH/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Humains , Données de séquences moléculaires , Phosphorylation , Cartographie d'interactions entre protéines , Protéines proto-oncogènes c-mdm2/métabolisme , Interférence par ARN , Petit ARN interférent/métabolisme , Transfection , Protéine p53 suppresseur de tumeur/génétique , Ubiquitin-protein ligases/antagonistes et inhibiteurs , Ubiquitin-protein ligases/génétique , Ubiquitination
19.
Int J Mol Sci ; 13(12): 16865-79, 2012 Dec 10.
Article de Anglais | MEDLINE | ID: mdl-23222733

RÉSUMÉ

Neuronal survival and death of neurons are considered a fundamental mechanism in the regulation of the nervous system during early development of the system and in adulthood. Defects in this mechanism are highly problematic and are associated with many neurodegenerative diseases. Because neuronal programmed death is apoptotic in nature, indicating that apoptosis is a key regulatory process, the p53 family members (p53, p73, p63) act as checkpoints in neurons due to their role in apoptosis. The complexity of this system is due to the existence of different naturally occurring isoforms that have different functions from the wild types (WT), varying from apoptotic to anti-apoptotic effects. In this review, we focus on the role of UBE4B (known as Ube4b or Ufd2a in mouse), an E3/E4 ligase that triggers substrate polyubiquitination, as a master regulatory ligase associated with the p53 family WT proteins and isoforms in regulating neuronal survival. UBE4B is also associated with other pathways independent of the p53 family, such as polyglutamine aggregation and Wallerian degeneration, both of which are critical in neurodegenerative diseases. Many of the hypotheses presented here are gateways to understanding the programmed death/survival of neurons regulated by UBE4B in normal physiology, and a means of introducing potential therapeutic approaches with implications in treating several neurodegenerative diseases.


Sujet(s)
Neurones/physiologie , Protéines suppresseurs de tumeurs/physiologie , Ubiquitin-protein ligase complexes/physiologie , Animaux , Mort cellulaire/génétique , Survie cellulaire/génétique , Régulation de l'expression des gènes , Gènes p53 , Humains , Souris , Ubiquitin-protein ligases
20.
J Clin Pathol ; 65(10): 945-8, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22639408

RÉSUMÉ

AIMS: To assess the expression of Twisted gastrulation (TWSG1) protein, which regulates the activity of bone morphogenetic proteins (BMPs) in the extracellular space in malignant epithelial tumours of the liver. METHODS: Thirteen hepatocellular carcinoma (HCC) samples and 12 intrahepatic cholangiocellular carcinoma (CCA) samples were compiled into diagnosis-specific tissue microarrays. Sections were immunostained with a monoclonal antibody against TWSG1 and a polyclonal antibody against BMP4. Human cell lines were also used, including one HCC cell line (HepG2), three CCA cell lines (OZ, Huh-28, HuCCT-1) and a Papova-immortalised normal hepatocyte cell line (THLE-3) for western blot analysis (WBA). RESULTS: Immunostaining and WBA showed a stronger TWSG1 expression in CCA than in HCC. The difference in expression was significant (p<0.05), and the immunohistochemical signal was particularly evident in the malignant epithelial areas close to desmoplastic stroma in CCA and in the areas of glandular differentiation in HCC. No expression was seen in normal hepatocytes. Interestingly, BMP4 was fully expressed in CCA and only partly in HCC. WBA showed a band for BMP4 in both CCA and HCC cell lines. CONCLUSIONS: TWSG1 is expressed in both malignant epithelial carcinomas, although the level of expression is higher in CCA than in HCC and seems to correlate at least partially with BMP4 expression.


Sujet(s)
Protéine morphogénétique osseuse de type 4/métabolisme , Carcinome hépatocellulaire/métabolisme , Cholangiocarcinome/métabolisme , Tumeurs du foie/métabolisme , Protéines tumorales/métabolisme , Adulte , Sujet âgé , Technique de Western , Lignée cellulaire tumorale , Femelle , Hépatocytes/métabolisme , Humains , Mâle , Adulte d'âge moyen , Protéines/métabolisme , Études rétrospectives , Analyse sur puce à tissus
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...