Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
BMC Res Notes ; 16(1): 26, 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36864454

ABSTRACT

In recent decades, advances in methods in molecular biology and genetics have revolutionized multiple areas of the life and health sciences. However, there remains a global need for the development of more refined and effective methods across these fields of research. In this current Collection, we aim to showcase articles presenting novel molecular biology and genetics techniques developed by scientists from around the world.


Subject(s)
Molecular Biology , Physicians , Humans
3.
Oncogene ; 41(30): 3778-3790, 2022 07.
Article in English | MEDLINE | ID: mdl-35761036

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the deadliest cancers. The retinoblastoma protein (RB1), a regulator of cell proliferation, is functionally inactivated in HCC by CYCLIN D/E-mediated phosphorylation. However, the mechanism of RB1-inactivation is unclear because only small percentages of HCCs exhibit amplification of CYCLIN D/E or mutations in the CDK-inhibitory genes. We show that FOXM1, which is overexpressed and critical for HCC, plays essential roles in inactivating RB1 and suppressing RB1-induced senescence of the HCC cells. Mechanistically, FOXM1 binds RB1 and DNMT3B to repress the expression of FOXO1, leading to a decrease in the levels of the CDK-inhibitors, creating an environment for phosphorylation and inactivation of RB1. Consistent with that, inhibition of FOXM1 causes increased expression of FOXO1 with consequent activation of RB1, leading to senescence of the HCC cells, in vitro and in vivo. Also, repression-deficient mutants of FOXM1 induce senescence that is blocked by depletion of RB1 or FOXO1. We provide evidence that human HCCs rely upon this FOXM1-FOXO1 axis for phosphorylation and inactivation of RB1. The observations demonstrate the existence of a new autoregulatory loop of RB1-inactivation in HCC involving a FOXM1-FOXO1 axis that is required for phosphorylation of RB1 and for aggressive progression of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cellular Senescence , Cyclin D/metabolism , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
Cancer Res ; 82(13): 2458-2471, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35583996

ABSTRACT

The transcription factor Forkhead box M1 (FoxM1) is overexpressed in breast cancers and correlates with poor prognosis. Mechanistically, FoxM1 associates with CBP to activate transcription and with Rb to repress transcription. Although the activating function of FoxM1 in breast cancer has been well documented, the significance of its repressive activity is poorly understood. Using CRISPR-Cas9 engineering, we generated a mouse model that expresses FoxM1-harboring point mutations that block binding to Rb while retaining its ability to bind CBP. Unlike FoxM1-null mice, mice harboring Rb-binding mutant FoxM1 did not exhibit significant developmental defects. The mutant mouse line developed PyMT-driven mammary tumors that were deficient in lung metastasis, which was tumor cell-intrinsic. Single-cell RNA-seq of the tumors revealed a deficiency in prometastatic tumor cells and an expansion of differentiated alveolar type tumor cells, and further investigation identified that loss of the FoxM1/Rb interaction caused enhancement of the mammary alveolar differentiation program. The FoxM1 mutant tumors also showed increased Pten expression, and FoxM1/Rb was found to activate Akt signaling by repressing Pten. In human breast cancers, expression of FoxM1 negatively correlated with Pten mRNA. Furthermore, the lack of tumor-infiltrating cells in FoxM1 mutant tumors appeared related to decreases in pro-metastatic tumor cells that express factors required for infiltration. These observations demonstrate that the FoxM1/Rb-regulated transcriptome is critical for the plasticity of breast cancer cells that drive metastasis, identifying a prometastatic role of Rb when bound to FoxM1. SIGNIFICANCE: This work provides new insights into how the interaction between FoxM1 and Rb facilitates the evolution of metastatic breast cancer cells by altering the transcriptome.


Subject(s)
Breast Neoplasms , Forkhead Box Protein M1/metabolism , Forkhead Transcription Factors , Animals , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Female , Forkhead Box Protein M1/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Signal Transduction , Transcription, Genetic
5.
Life Sci ; 288: 120157, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34801511

ABSTRACT

AIM: High risk Human Papillomavirus (HPV) is an infectious pathogen implicated in a variety of cancers with poor clinical outcome. The mechanism of HPV induced cellular transformation and its intervention remains to be elucidated. Human ADA3 (hADA3), a cellular target of HPV16 E6, is an essential and conserved component of the ADA transcriptional coactivator complex. High risk HPV-E6 binds and functionally inactivates hADA3 to initiate oncogenesis. The aim of this study was to identify the interaction interface between hADA3 and HPV16E6 for designing inhibitory peptides that can potentially disrupt the hADA3-E6 interaction. MATERIAL METHODS: The present investigation employed structure-based in silico tools supported by biochemical validation, in vivo interaction studies and analysis of posttranslational modifications. KEY FINDINGS: First 3D-model of hADA3 was proposed and domains involved in the oncogenic interaction between hADA3 and HPV16E6 were delineated. Rationally designed peptide disrupted hADA3-E6 interaction and impeded malignant properties of cervical cancer cells. SIGNIFICANCE: Intervention of hADA3-E6 interaction thus promises to be a potential strategy to combat HPV induced oncogenic conditions like cervical cancer. The investigation provides mechanistic insights into HPV pathogenesis and shows promise in developing novel therapeutics to treat HPV induced cancers.


Subject(s)
Oncogene Proteins, Viral/antagonists & inhibitors , Papillomavirus Infections/complications , Peptide Fragments/pharmacology , Protein Interaction Domains and Motifs/drug effects , Repressor Proteins/antagonists & inhibitors , Sumoylation , Transcription Factors/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Cell Communication , Cell Transformation, Neoplastic , Female , Humans , Oncogene Proteins, Viral/metabolism , Papillomaviridae/isolation & purification , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Protein Conformation , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
6.
Oncogene ; 39(8): 1784-1796, 2020 02.
Article in English | MEDLINE | ID: mdl-31740787

ABSTRACT

Cancers in the oral/head & neck region (HNSCC) are aggressive due to high incidence of recurrence and distant metastasis. One prominent feature of aggressive HNSCC is the presence of severely hypoxic regions in tumors and activation of hypoxia-inducible factors (HIFs). In this study, we report that the XPE gene product DDB2 (damaged DNA binding protein 2), a nucleotide excision repair protein, is upregulated by hypoxia. Moreover, DDB2 inhibits HIF1α in HNSCC cells. It inhibits HIF1α in both normoxia and hypoxia by reducing mRNA expression. Knockdown of DDB2 enhances the expression of angiogenic markers and promotes tumor growth in a xenograft model. We show that DDB2 binds to an upstream promoter element in the HIF1Α gene and promotes histone H3K9 trimethylation around the binding site by recruiting Suv39h1. Also, we provide evidence that DDB2 has a significant suppressive effect on expression of the endogenous markers of hypoxia that are also prognostic indicators in HNSCC. Together, these results describe a new mechanism of hypoxia regulation that opposes expression of HIF1Α mRNA and the hypoxia-response genes.


Subject(s)
DNA-Binding Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Hypoxia , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Squamous Cell Carcinoma of Head and Neck/genetics
7.
Mol Cancer Res ; 17(5): 1063-1074, 2019 05.
Article in English | MEDLINE | ID: mdl-30814128

ABSTRACT

The forkhead box transcription factor FoxM1 is essential for hepatocellular carcinoma (HCC) development, and its overexpression coincides with poor prognosis. Here, we show that the mechanisms by which FoxM1 drives HCC progression involve overcoming the inhibitory effects of the liver differentiation gene FoxA2. First, the expression patterns of FoxM1 and FoxA2 in human HCC are opposite. We show that FoxM1 represses expression of FoxA2 in G1 phase. Repression of FoxA2 in G1 phase is important, as it is capable of inhibiting expression of the pluripotency genes that are expressed mainly in S-G2 phases. Using a transgenic mouse model for oncogenic Ras-driven HCC, we provide genetic evidence for a repression of FoxA2 by FoxM1. Conversely, FoxA2 inhibits expression of FoxM1 and inhibits FoxM1-induced tumorigenicity. Also, FoxA2 inhibits Ras-induced HCC progression that involves FoxM1. IMPLICATIONS: The observations provide strong genetic evidence for an opposing role of FoxM1 and FoxA2 in HCC progression. Moreover, FoxM1 drives high-grade HCC progression partly by inhibiting the hepatocyte differentiation gene FoxA2.


Subject(s)
Carcinoma, Hepatocellular/pathology , Forkhead Box Protein M1/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Liver Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Transgenic , Neoplasm Grading , Neoplasms, Experimental , Retinoblastoma Protein/metabolism , DNA Methyltransferase 3B
8.
Sci Rep ; 8(1): 15850, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30374061

ABSTRACT

A correction has been published and is appended to both the HTML and PDF versions of this paper. The error has not been fixed in the paper.

9.
FEBS Lett ; 591(21): 3507-3522, 2017 11.
Article in English | MEDLINE | ID: mdl-28948618

ABSTRACT

Cytoglobin (Cygb) is a cellular haemoprotein belonging to the globin family with ambiguous biological functions. Downregulation of Cygb in many cancers is indicative of its tumour-suppressive role. This is the first report showing the cell cycle regulation of Cygb, which was found to peak at G1 and rapidly decline in S phase. Importantly, Skp2-mediated degradation of Cygb was identified as the key mechanism for controlling its oscillating levels during the cell cycle. Moreover, overexpression of Cygb stimulates hypophosphorylation of Rb causing delayed cell cycle progression. Overall, the study reveals a novel mechanism for the regulated expression of Cygb and also assigns a new role to Cygb in cell cycle control.


Subject(s)
G1 Phase/physiology , Globins/metabolism , Proteolysis , S Phase/physiology , S-Phase Kinase-Associated Proteins/metabolism , Cell Cycle , Cytoglobin , HEK293 Cells , Humans , Phosphorylation/physiology , Retinoblastoma Protein/metabolism
10.
Sci Rep ; 7: 46017, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28387346

ABSTRACT

FoxM1b is a cell cycle-regulated transcription factor, whose over-expression is a marker for poor outcome in cancers. Its transcriptional activation function requires phosphorylation by Cdk1 or Cdk2 that primes FoxM1b for phosphorylation by Plk1, which triggers association with the co-activator CBP. FoxM1b also possesses transcriptional repression function. It represses the mammary differentiation gene GATA3 involving DNMT3b and Rb. We investigated what determines the two distinct functions of FoxM1b: activation and repression. We show that Rb binds to the C-terminal activation domain of FoxM1b. Analyses with phospho-defective and phospho-mimetic mutants of FoxM1b identified a critical role of the Plk1 phosphorylation sites in regulating the binding of FoxM1b to Rb and DNMT3b. That is opposite of what was seen for the interaction of FoxM1b with CBP. We show that, in addition to GATA3, FoxM1b also represses the mammary luminal differentiation marker FoxA1 by promoter-methylation, and that is regulated by the Plk1 phosphorylation sites in FoxM1b. Our results show that the Plk1 phosphorylation sites in FoxM1b serve as a regulator for its repressor function, and they provide insights into how FoxM1b inhibits differentiation genes and activates proliferation genes during cancer progression.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Box Protein M1/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Binding Sites , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Forkhead Box Protein M1/chemistry , GATA3 Transcription Factor/genetics , Humans , MCF-7 Cells , Mutation/genetics , Peptide Fragments/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Domains , Sialoglycoproteins/metabolism , DNA Methyltransferase 3B , Polo-Like Kinase 1
11.
Open Biol ; 6(9)2016 09.
Article in English | MEDLINE | ID: mdl-27605378

ABSTRACT

Human ADA3, the evolutionarily conserved transcriptional co-activator, remains the unified part of multiple cellular functions, including regulation of nuclear receptor functions, cell proliferation, apoptosis, senescence, chromatin remodelling, genomic stability and chromosomal maintenance. The past decade has witnessed exciting findings leading to considerable expansion in research related to the biology and regulation of hADA3. Embryonic lethality in homozygous knockout Ada3 mouse signifies the importance of this gene product during early embryonic development. Moreover, the fact that it is a novel target of Human Papillomavirus E6 oncoprotein, one of the most prevalent causal agents behind cervical cancer, helps highlight some of the crucial aspects of HPV-mediated oncogenesis. These findings imply the central involvement of hADA3 in regulation of various cellular functional losses accountable for the genesis of malignancy and viral infections. Recent reports also provide evidence for post-translational modifications of hADA3 leading to its instability and contributing to the malignant phenotype of cervical cancer cells. Furthermore, its association with poor prognosis of breast cancer suggests intimate association in the pathogenesis of the disease. Here, we present the first review on hADA3 with a comprehensive outlook on the molecular and functional roles of hADA3 to provoke further interest for more elegant and intensive studies exploring assorted aspects of this protein.


Subject(s)
Carcinogenesis , Embryonic Development , Oncogene Proteins, Viral/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Cell Proliferation , Female , Humans , Mice , Oncogene Proteins, Viral/genetics , Papillomaviridae/metabolism , Protein Processing, Post-Translational , Transcription Factors/genetics , Transcriptional Activation , Uterine Cervical Neoplasms/pathology
12.
Int J Biochem Cell Biol ; 58: 28-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25462159

ABSTRACT

The oncogenic transcription factor Forkhead box M1b (FoxM1b), a key regulator of cell cycle, is often overexpressed in many human cancers. Interestingly, posttranslational modifications are known to play important role in regulating the levels and activity of FoxM1b. The purpose of the present study was to characterize the SUMOylation of FoxM1b and identify the functional consequences including viral pathogenesis. Here, we report that FoxM1b interacts with SUMOylating enzymes Ubc9 and PIAS1 and acts as a substrate for SUMOylation. We also show that SUMOylation facilitates FoxM1b protein destabilization and nucleocytoplasmic shuttling. More importantly, we provide the first evidence for a role of E7 oncoprotein in high risk human papillomavirus (HPV) mediated upregulation of FoxM1b. The elevated expression of FoxM1 was determined to be posttranscriptional and was attributed to decreased SUMOylation of FoxM1b in the E7-expressing cells. Moreover, we demonstrate the involvement of SUMOylation in regulation of FoxM1 and present biochemical evidence that HPV16 E7 oncoprotein can modulate SUMOylation of FoxM1b by impairing its interaction with Ubc9. Together, these results provide a novel connection between SUMOylation of FoxM1b and HPV carcinogenesis. The findings may have important implications in the discovery of future anti-cancer therapeutics.


Subject(s)
Forkhead Transcription Factors/metabolism , Papillomavirus E7 Proteins/genetics , Blotting, Western , Cell Line , Cell Line, Tumor , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , HeLa Cells , Humans , Immunoprecipitation , Sumoylation/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
13.
DNA Repair (Amst) ; 24: 107-112, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25269893

ABSTRACT

Cytoglobin (Cygb) is an emerging tumor suppressor gene silenced by promoter hypermethylation in many human tumors. So far, the precise molecular mechanism underlying its tumor suppressive function remains poorly understood. Here, we identified Cygb as a genotoxic stress-responsive hemoprotein upregulated upon sensing cellular DNA damage. Our studies demonstrated that Cygb physically associates with and stabilizes p53, a key cellular DNA damage signaling factor. We provide evidence that Cygb extends the half-life of p53 by blocking its ubiquitination and subsequent degradation. We show that, upon DNA damage, cells overexpressing Cygb displayed proliferation defect by rapid accumulation of p53 and its target gene p21, while Cygb knockdown cells failed to efficiently arrest in G1 phase in response to DNA insult. These results suggest a possible involvement of Cygb in mediating cellular response to DNA damage and thereby contributing in the maintenance of genomic integrity. Our study thus presents a novel insight into the mechanistic role of Cygb in tumor suppression.


Subject(s)
DNA Damage , G1 Phase Cell Cycle Checkpoints , Globins/metabolism , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoglobin , DNA Damage/drug effects , Doxorubicin/pharmacology , Etoposide/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Globins/genetics , HEK293 Cells/drug effects , Humans , Protein Stability , Tumor Suppressor Protein p53/genetics , Ubiquitination
14.
Carcinogenesis ; 35(8): 1830-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24795430

ABSTRACT

Despite significant research, our understanding of the molecular mechanisms of Human Papilloma Virus (HPV) induced cancers remains incomplete. Majority of invasive cervical cancers are caused by high-risk HPV 16 and 18. Two potent HPV oncoproteins, E6 and E7, promote human malignancies by disrupting the activities of key regulators of cell proliferation and apoptosis. Recent investigations have identified hADA3, a transcriptional coactivator protein as a target of high-risk HPV16E6. However, the mechanism of degradation of hADA3 by E6 and its contribution in HPV induced carcinogenesis is poorly understood. Here, we showed that E6-mediated proteolysis of hADA3 is responsible for maintaining low levels of hADA3 in HPV-positive cervical cancer cell lines. We demonstrate that HPV16E6 targets hADA3 for ubiquitin-mediated degradation via E6AP ubiquitin ligase. We also show that hADA3 undergoes accelerated SUMOylation in the presence of HPV16E6. Our data represent the first evidence that hADA3 is posttranslationally modified by SUMOylation, which makes it unstable and establishes a link between SUMOylation and E6-mediated ubiquitination of hADA3. Furthermore, depletion of Ubc9 prevented rapid degradation of hADA3 in E6 expressing cervical cancer cells and overexpression of hADA3 resulted in suppression of proliferation and migration abilities of SiHa cells. Overall, this study underscores the importance of posttranslational modifications in HPV16E6-mediated downregulation of hADA3 thereby unveiling a novel mechanism by which HPV induces oncogenesis.


Subject(s)
Human papillomavirus 16/physiology , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/metabolism , Repressor Proteins/metabolism , Sumoylation , Transcription Factors/metabolism , Uterine Cervical Neoplasms/metabolism , Blotting, Western , Cell Proliferation , Female , Humans , Immunoprecipitation , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Proteolysis , Risk Factors , Tumor Cells, Cultured , Ubiquitination , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL