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










Publication year range
1.
Curr Opin Pharmacol ; 76: 102460, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38776747

ABSTRACT

Shelterin proteins regulate genomic stability by preventing inappropriate DNA damage responses (DDRs) at telomeres. Unprotected telomeres lead to persistent DDR causing cell cycle inhibition, growth arrest, and apoptosis. Cancer cells rely on DDR to protect themselves from DNA lesions and exogenous DNA-damaging agents such as chemotherapy and radiotherapy. Therefore, targeting DDR machinery is a promising strategy to increase the sensitivity of cancer cells to existing cancer therapies. However, the success of these DDR inhibitors depends on other mutations, and over time, patients develop resistance to these therapies. This suggests the need for alternative approaches. One promising strategy is co-inhibiting shelterin proteins with DDR molecules, which would offset cellular fitness in DNA repair in a mutation-independent manner. This review highlights the associations and dependencies of the shelterin complex with the DDR proteins and discusses potential co-inhibition strategies that might improve the therapeutic potential of current inhibitors.


Subject(s)
Antineoplastic Agents , DNA Damage , DNA Repair , Neoplasms , Humans , Neoplasms/drug therapy , DNA Repair/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Telomere/drug effects , Telomere/metabolism , Telomere-Binding Proteins/metabolism , Molecular Targeted Therapy
2.
Nucleic Acids Res ; 51(1): 1-16, 2023 01 11.
Article in English | MEDLINE | ID: mdl-35697349

ABSTRACT

Transcriptional reactivation of hTERT is the limiting step in tumorigenesis. While mutations in hTERT promoter present in 19% of cancers are recognized as key drivers of hTERT reactivation, mechanisms by which wildtype hTERT (WT-hTERT) promoter is reactivated, in majority of human cancers, remain unknown. Using primary colorectal cancers (CRC) we identified Tert INTeracting region 2 (T-INT2), the critical chromatin region essential for reactivating WT-hTERT promoter in CRCs. Elevated ß-catenin and JunD level in CRC facilitates chromatin interaction between hTERT promoter and T-INT2 that is necessary to turn on hTERTexpression. Pharmacological screens uncovered salinomycin, which inhibits JunD mediated hTERT-T-INT2 interaction that is required for the formation of a stable transcription complex on the hTERT promoter. Our results showed for the first time how known CRC alterations, such as APC, lead to WT-hTERT promoter reactivation during stepwise-tumorigenesis and provide a new perspective for developing cancer-specific drugs.


Healthy and cancer cells harbor the same DNA sequence, but reactivation of the Human Telomerase Reverse Transcriptase (hTERT) gene is observed only in cancer cells. How does that happen was not known for over three decades of research? This study identifies a specific DNA structure that forms only in cancer cells and brings the necessary molecular machinery into the correct position to activate the hTERT gene. The detailed mechanism of hTERT activation provided in this study will be instrumental in designing cancer cell-specific hTERT inhibitors, especially since all the other ways of inhibiting telomerase failed in the clinic.


Subject(s)
Colorectal Neoplasms , Telomerase , Humans , Carcinogenesis , Chromatin/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Promoter Regions, Genetic , Telomerase/antagonists & inhibitors , Telomerase/genetics , Transcription, Genetic
3.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35027447

ABSTRACT

Cancer-specific hTERT promoter mutations reported in 19% of cancers result in enhanced telomerase activity. Understanding the distinctions between transcriptional regulation of wild-type (WT) and mutant (Mut) hTERT promoters may open up avenues for development of inhibitors which specially block hTERT expression in cancer cells. To comprehensively identify physiological regulators of WT- or Mut-hTERT promoters, we generated several isogenic reporter cells driven by endogenous hTERT loci. Genome-wide CRISPR-Cas9 and small interfering RNA screens using these isogenic reporter lines identified specific regulators of Mut-hTERT promoters. We validate and characterize one of these hits, namely, MED12, a kinase subunit of mediator complex. We demonstrate that MED12 specifically drives expression of hTERT from the Mut-hTERT promoter by mediating long-range chromatin interaction between the proximal Mut-hTERT promoter and T-INT1 distal regulatory region 260 kb upstream. Several hits identified in our screens could serve as potential therapeutic targets, inhibition of which may specifically block Mut-hTERT promoter driven telomerase reactivation in cancers.


Subject(s)
Mutation , Promoter Regions, Genetic , Telomerase/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Chromatin , DNA-Binding Proteins , Gene Editing , Gene Expression Regulation, Neoplastic , Humans , Mediator Complex/genetics , Mediator Complex/metabolism , Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Telomerase/metabolism , Transcription Factors , Transcription, Genetic
4.
Genome Biol ; 22(1): 226, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34399797

ABSTRACT

Chromatin interactions play important roles in regulating gene expression. However, the availability of genome-wide chromatin interaction data is limited. We develop a computational method, chromatin interaction neural network (ChINN), to predict chromatin interactions between open chromatin regions using only DNA sequences. ChINN predicts CTCF- and RNA polymerase II-associated and Hi-C chromatin interactions. ChINN shows good across-sample performances and captures various sequence features for chromatin interaction prediction. We apply ChINN to 6 chronic lymphocytic leukemia (CLL) patient samples and a published cohort of 84 CLL open chromatin samples. Our results demonstrate extensive heterogeneity in chromatin interactions among CLL patient samples.


Subject(s)
Chromatin , Machine Learning , Neural Networks, Computer , Base Sequence , Computational Biology , Genome , Humans , Leukemia/genetics
5.
Cell Mol Life Sci ; 78(9): 4235-4257, 2021 May.
Article in English | MEDLINE | ID: mdl-33599797

ABSTRACT

Reactivation of telomerase is a major hallmark observed in 90% of all cancers. Yet paradoxically, enhanced telomerase activity does not correlate with telomere length and cancers often possess short telomeres; suggestive of supplementary non-canonical roles that telomerase might play in the development of cancer. Moreover, studies have shown that aberrant expression of shelterin proteins coupled with their release from shortening telomeres can further promote cancer by mechanisms independent of their telomeric role. While targeting telomerase activity appears to be an attractive therapeutic option, this approach has failed in clinical trials due to undesirable cytotoxic effects on stem cells. To circumvent this concern, an alternative strategy could be to target the molecules involved in the non-canonical functions of telomeric proteins. In this review, we will focus on emerging evidence that has demonstrated the non-canonical roles of telomeric proteins and their impact on tumorigenesis. Furthermore, we aim to address current knowledge gaps in telomeric protein functions and propose future research approaches that can be undertaken to achieve this.


Subject(s)
Neoplasms/pathology , Telomere-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/pathology , Humans , Neoplasms/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
6.
Gut ; 70(10): 1857-1871, 2021 10.
Article in English | MEDLINE | ID: mdl-33239342

ABSTRACT

OBJECTIVE: NFκB is the key modulator in inflammatory disorders. However, the key regulators that activate, fine-tune or shut off NFκB activity in inflammatory conditions are poorly understood. In this study, we aim to investigate the roles that NFκB-specific long non-coding RNAs (lncRNAs) play in regulating inflammatory networks. DESIGN: Using the first genetic-screen to identify NFκB-specific lncRNAs, we performed RNA-seq from the p65-/- and Ikkß-/- mouse embryonic fibroblasts and report the identification of an evolutionary conserved lncRNA designated mNAIL (mice) or hNAIL (human). hNAIL is upregulated in human inflammatory disorders, including UC. We generated mNAILΔNFκB mice, wherein deletion of two NFκB sites in the proximal promoter of mNAIL abolishes its induction, to study its function in colitis. RESULTS: NAIL regulates inflammation via sequestering and inactivating Wip1, a known negative regulator of proinflammatory p38 kinase and NFκB subunit p65. Wip1 inactivation leads to coordinated activation of p38 and covalent modifications of NFκB, essential for its genome-wide occupancy on specific targets. NAIL enables an orchestrated response for p38 and NFκB coactivation that leads to differentiation of precursor cells into immature myeloid cells in bone marrow, recruitment of macrophages to inflamed area and expression of inflammatory genes in colitis. CONCLUSION: NAIL directly regulates initiation and progression of colitis and its expression is highly correlated with NFκB activity which makes it a perfect candidate to serve as a biomarker and a therapeutic target for IBD and other inflammation-associated diseases.


Subject(s)
Colitis/genetics , Colitis/metabolism , RNA, Long Noncoding/metabolism , Transcription Factor RelA/metabolism , Animals , Biomarkers/metabolism , Disease Progression , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System , Mice , Protein Phosphatase 2C/metabolism
7.
J Clin Invest ; 126(10): 4045-4060, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27643433

ABSTRACT

Transcriptional reactivation of telomerase reverse transcriptase (TERT) reconstitutes telomerase activity in the majority of human cancers. Here, we found that ectopic TERT expression increases cell proliferation, while acute reductions in TERT levels lead to a dramatic loss of proliferation without any change in telomere length, suggesting that the effects of TERT could be telomere independent. We observed that TERT determines the growth rate of cancer cells by directly regulating global protein synthesis independently of its catalytic activity. Genome-wide TERT binding across 5 cancer cell lines and 2 embryonic stem cell lines revealed that endogenous TERT, driven by mutant promoters or oncogenes, directly associates with the RNA polymerase III (pol III) subunit RPC32 and enhances its recruitment to chromatin, resulting in increased RNA pol III occupancy and tRNA expression in cancers. TERT-deficient mice displayed marked delays in polyomavirus middle T oncogene-induced (PyMT-induced) mammary tumorigenesis, increased survival, and reductions in tRNA levels. Ectopic expression of either RPC32 or TERT restored tRNA levels and proliferation defects in TERT-depleted cells. Finally, we determined that levels of TERT and tRNA correlated in breast and liver cancer samples. Together, these data suggest the existence of a unifying mechanism by which TERT enhances translation in cells to regulate cancer cell proliferation.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , RNA, Transfer/genetics , Telomerase/physiology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Female , HEK293 Cells , Humans , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Transplantation , Promoter Regions, Genetic , Protein Binding , Protein Biosynthesis , Protein Interaction Domains and Motifs , RNA Polymerase III/metabolism , RNA, Transfer/metabolism
8.
Cancer Discov ; 6(11): 1276-1291, 2016 11.
Article in English | MEDLINE | ID: mdl-27650951

ABSTRACT

Cancer-specific TERT promoter mutations (-146C>T and -124C>T) have been linked to reactivation of the epigenetically silenced telomerase reverse transcriptase gene (TERT). Understanding how these single-nucleotide alterations drive TERT reactivation is a fundamental unanswered question and is key for making successful therapeutics. We show that unlike wild-type promoters, recruitment of the transcription factor GABPA specifically to mutant TERT promoters mediates long-range chromatin interaction and enrichment of active histone marks, and hence drives TERT transcription. CRISPR-mediated reversal of mutant TERT promoters, or deletion of its long-range interacting chromatin, abrogates GABPA binding and long-range interactions, leading to depletion of active histone marks, loss of POL2 recruitment, and suppression of TERT transcription. In contrast, de novo introduction of a TERT promoter mutation enables GABPA binding and upregulation of TERT via long-range interactions, acquisition of active histone marks, and subsequent POL2 recruitment. This study provides a unifying mechanistic insight into activation of mutant TERT promoters across various human cancers. SIGNIFICANCE: This study identifies a key mechanism by which cancer-specific mutant TERT promoters cause reactivation of TERT Because the mechanism uncovered here is not utilized by promoters that drive TERT in normal cells, this mechanism could be exploited to make inhibitors which have the potential to block telomerase function and hence the progression of up to 90% of human cancers. Cancer Discov; 6(11); 1276-91. ©2016 AACR.See related commentary by Min and Shay, p. 1212This article is highlighted in the In This Issue feature, p. 1197.


Subject(s)
Neoplasms/genetics , Telomerase/genetics , Transcriptional Activation/genetics , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplasms/pathology , Promoter Regions, Genetic , Telomerase/metabolism
9.
Cell Mol Life Sci ; 73(8): 1659-70, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26846696

ABSTRACT

Activation of telomerase is a critical step in the development of about 85 % of human cancers. Levels of Tert, which encodes the reverse transcriptase subunit of telomerase, are limiting in normal somatic cells. Tert is subjected to transcriptional, post-transcriptional and epigenetic regulation, but the precise mechanism of how telomerase is re-activated in cancer cells is poorly understood. Reactivation of the Tert promoter involves multiple changes which evolve during cancer progression including mutations and chromosomal re-arrangements. Newly described non-coding mutations in the Tert promoter region of many cancer cells (19 %) in two key positions, C250T and C228T, have added another layer of complexity to telomerase reactivation. These mutations create novel consensus sequences for transcription factors which can enhance Tert expression. In this review, we will discuss gene structure and function of Tert and provide insights into the mechanisms of Tert reactivation in cancers, highlighting the contribution of recently identified Tert promoter mutations.


Subject(s)
Neoplasms/pathology , Promoter Regions, Genetic/genetics , Telomerase/genetics , Telomerase/metabolism , Enzyme Activation/physiology , Epigenesis, Genetic , Humans , Mutation/genetics , Neoplasms/genetics
10.
FEBS Lett ; 589(9): 974-84, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25749370

ABSTRACT

Besides its canonical function of catalyzing the formation of telomeric repeats, many groups have recently reported non-canonical functions of hTERT in particular, and telomerase in general. Regulating transcription is the central basis of non-canonical functions of telomerase. However, unlike reverse transcriptase activity of telomerase that requires only a few molecules of enzymatically active hTERT, non-canonical functions of hTERT or other telomerase components theoretically require several hundred copies. Here, we provide the first direct quantification of all the telomerase components in human cancer cell lines. We demonstrate that telomerase components do not exist in a 1:1 stoichiometric ratio, and there are several hundred copies of hTERT in cells. This provides the molecular basis of hTERT to function in other signaling cascades, including transcription.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Telomerase/genetics , Telomerase/metabolism , Blotting, Western , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Neoplasms/enzymology , Neoplasms/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
11.
Nat Commun ; 6: 6184, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25630468

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally. The identity and role of cell surface molecules driving complex biological events leading to HCC progression are poorly understood, hence representing major lacunae in HCC therapies. Here, combining SILAC quantitative proteomics and biochemical approaches, we uncover a critical oncogenic role of Agrin, which is overexpressed and secreted in HCC. Agrin enhances cellular proliferation, migration and oncogenic signalling. Mechanistically, Agrin's extracellular matrix sensor activity provides oncogenic cues to regulate Arp2/3-dependent ruffling, invadopodia formation and epithelial-mesenchymal transition through sustained focal adhesion integrity that drives liver tumorigenesis. Furthermore, Agrin signalling through Lrp4-muscle-specific tyrosine kinase (MuSK) forms a critical oncogenic axis. Importantly, antibodies targeting Agrin reduced oncogenic signalling and tumour growth in vivo. Together, we demonstrate that Agrin is frequently upregulated and important for oncogenic property of HCC, and is an attractive target for antibody therapy.


Subject(s)
Agrin/metabolism , Carcinoma, Hepatocellular/metabolism , Focal Adhesions/metabolism , Liver Neoplasms/metabolism , Oncogenes , Animals , Antibodies, Blocking/pharmacology , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Endocytosis/drug effects , Epithelial-Mesenchymal Transition/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Gene Knockdown Techniques , Integrins/metabolism , Isotope Labeling , LDL-Receptor Related Proteins/metabolism , Liver Neoplasms/pathology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Pseudopodia/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/metabolism , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
12.
Cell Rep ; 7(6): 1876-86, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24931609

ABSTRACT

Although much is known about the molecular players in insulin signaling, there is scant information about transcriptional regulation of its key components. We now find that NUCKS is a transcriptional regulator of the insulin signaling components, including the insulin receptor (IR). Knockdown of NUCKS leads to impaired insulin signaling in endocrine cells. NUCKS knockout mice exhibit decreased insulin signaling and increased body weight/fat mass along with impaired glucose tolerance and reduced insulin sensitivity, all of which are further exacerbated by a high-fat diet (HFD). Genome-wide ChIP-seq identifies metabolism and insulin signaling as NUCKS targets. Importantly, NUCKS is downregulated in individuals with a high body mass index and in HFD-fed mice, and conversely, its levels increase upon starvation. Altogether, NUCKS is a physiological regulator of energy homeostasis and glucose metabolism that works by regulating chromatin accessibility and RNA polymerase II recruitment to the promoters of IR and other insulin pathway modulators.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/metabolism , Glucose/metabolism , Insulin/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Animals , Body Weight , Diabetes Mellitus, Type 2/genetics , Homeostasis , Humans , Insulin Resistance , Mice , Mice, Knockout , Nuclear Proteins/genetics , Phosphoproteins/genetics , Signal Transduction , Transcriptional Activation
13.
Trends Mol Med ; 19(8): 487-500, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23746697

ABSTRACT

Inflammation is indispensable for host homeostasis against invading pathogens and efficient wound healing upon tissue malfunction and has to be tightly controlled by various mechanisms to limit excess responses harmful to host tissues. A myriad of disease conditions ranging from type 2 diabetes (T2D) to neurodegenerative and cardiovascular disorders are now shown to progress due to persistent, unresolved inflammation in metabolic tissues such as adipose, liver, pancreas, muscle, and brain. However, their underlying mechanisms are incompletely understood. The actions of innate and adaptive immune cells in these ailments are increasingly appreciated so much so that a new research area called 'immunometabolism' has emerged. In this review, we will highlight the fundamental roles of various immune cells in adipose tissue during the initiation and progression of obesity-induced inflammation and discuss potential anti-inflammatory therapies from different mechanistic points of view.


Subject(s)
Adipose Tissue/immunology , Immune System/immunology , Obesity/immunology , Animals , Chronic Disease , Humans , Immune System/cytology
14.
PLoS One ; 7(11): e49252, 2012.
Article in English | MEDLINE | ID: mdl-23152883

ABSTRACT

One of the heat shock family protein (Hsp) expressing bacteria is the gram negative, periodontal pathogen Aggregatibacter actinomycetemcomitans (Aa). A. actinomycetemcomitans' Hsp is a 64-kDa GroEL-protein, which has been shown to influence the host cells. In this study we used recombinant A. actinomycetemcomitans GroEL (rAaGroEL) protein as a model antigen to study GroEL-mediated T cell immune response. Human peripheral mononuclear cells (PBMCs), when stimulated with recombinant rAaGroEL, expressed early activation marker CD69 and IL-2R (CD25). CD25 and CD69 expressions were higher in CD4+ T cells compared to CD8+ T cells. rAaGroEL-responding CD4+ T cells expressed IL-10, IFNγ and TNFα cytokines. Interestingly, there were also IL-10 and IFNγ double cytokine producing CD4+ T cells. Additionally, IFNγ expressing CD4+ T cells were also T-bet positive. Altogether the results suggest that rAaGroEL protein affects CD4+ T cells to differentiate into IFNγ IL10-secreting T-bet+ Th1 cells.


Subject(s)
Aggregatibacter actinomycetemcomitans/metabolism , Chaperonin 60/pharmacology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , T-Box Domain Proteins/metabolism , Th1 Cells/cytology , Humans , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Recombinant Proteins/pharmacology , Th1 Cells/drug effects , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...