Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Cell Physiol ; 238(6): 1237-1255, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37183325

RESUMO

The protection of telomere protein (POT1) is a telomere-binding protein and is an essential component of the six-membered shelterin complex, which is associated with the telomeres. POT1 directly binds to the 3' single-stranded telomeric overhang and prevents the activation of DNA damage response at telomeres thus preventing the telomere-telomere fusions and genomic instability. POT1 also plays a pivotal role in maintaining telomere length by regulating telomerase-mediated telomere elongation. Mutations in POT1 proteins result in three different telomere phenotypes, which include long, short, or aberrant telomere length. Long telomeres predispose individuals to cancer, while short or aberrant telomere phenotypes result in pro-aging diseases referred to as telomeropathies. Here, we review the function of POT1 proteins in telomere length hemostasis and how the spectrum of mutations reported in POT1 can be segregated toward developing very distinct disease phenotypes of cancer and telomeropathies.


Assuntos
Complexo Shelterina , Proteínas de Ligação a Telômeros , Telômero , Mutação/genética , Fenótipo , Telômero/genética , Telômero/metabolismo , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Humanos
2.
Semin Cancer Biol ; 68: 258-278, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32380233

RESUMO

Human malignancies are one of the major health-related issues though out the world and anticipated to rise in the future. The development of novel drugs/agents requires a huge amount of cost and time that represents a major challenge for drug discovery. In the last three decades, the number of FDA approved drugs has dropped down and this led to increasing interest in drug reposition or repurposing. The present review focuses on recent concepts and therapeutic opportunities for the utilization of antidiabetics, antibiotics, antifungal, anti-inflammatory, antipsychotic, PDE inhibitors and estrogen receptor antagonist, Antabuse, antiparasitic and cardiovascular agents/drugs as an alternative approach against human malignancies. The repurposing of approved non-cancerous drugs is an effective strategy to develop new therapeutic options for the treatment of cancer patients at an affordable cost in clinics. In the current scenario, most of the countries throughout the globe are unable to meet the medical needs of cancer patients because of the high cost of the available cancerous drugs. Some of these drugs displayed potential anti-cancer activity in preclinic and clinical studies by regulating several key molecular mechanisms and oncogenic pathways in human malignancies. The emerging pieces of evidence indicate that repurposing of drugs is crucial to the faster and cheaper discovery of anti-cancerous drugs.


Assuntos
Antineoplásicos/uso terapêutico , Descoberta de Drogas , Reposicionamento de Medicamentos/métodos , Neoplasias/tratamento farmacológico , Preparações Farmacêuticas/administração & dosagem , Animais , Humanos
3.
J Ethnopharmacol ; 322: 117573, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38110133

RESUMO

ETHNOPHARMACOLOGY RELEVANCE: Syzygium cumini (L.) Skeels (SC), an ancient medicinal plant, is used as a complementary and alternative medicine for treating diabetes mellitus and its associated complications, such as diabetic nephropathy (DN). Phytochemicals present in SC homeopathic formulations possess anti-glycemic, anti-glycation, anti-inflammatory, and antioxidant properties. Additionally, the non-enzymatic formation of advanced glycation end products (AGEs) increases during hyperglycemia in diabetes. AGEs interaction with their receptor of AGEs (RAGE) promotes inflammation via Nuclear Factor-κB (NF-κB) and the accumulation of Extracellular Matrix (ECM) proteins, contributing to the renal dysfunction in DN. However, the molecular mechanism through which SC formulations interact with the AGEs-RAGE-NF-κB pathway has not yet been investigated. AIM: This study aims to examine the impact of SC formulations on the RAGE-NF-κB pathway and ECM protein modifications in glycation-induced DN using a molecular approach. MATERIALS AND METHODS: Human serum albumin (10 mg/ml) was glycated with MGO (55 mM) in the presence of SC formulations - Mother tincture (MT), 30C, 200C for 7 days. Glycated samples were added to renal cells (HEK 293) for 24 h. Subsequently, cellular gene and protein expressions of RAGE, NF-κB, vascular endothelial growth factor (VEGF), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), collagen IV (Col IV), and fibronectin were determined using RT-qPCR and Western blot analysis. The immunofluorescence, luciferase assay, and chromatin immunoprecipitation techniques were employed to gain insights into glycation-induced NF-κB nuclear translocation, transcriptional activity, and its effect on RAGE promoter activity in SC-treated cells. RESULTS: SC formulations significantly downregulated glycation-induced elevated levels of RAGE and NF-κB. Mechanistically, SC formulations prevented NF-κB nuclear translocation, transcriptional activity, and RAGE promoter activity. Also, SC formulations significantly attenuated glycation-enhanced expressions of inflammatory cytokines (IL-6, TNF-α, and VEGF) and ECM proteins (Col IV and fibronectin). CONCLUSION: Our findings enlighten the molecular mechanism of SC in DN by targeting the AGEs-RAGE-NF-κB signaling pathway, inflammatory responses, and ECM accumulation. Hence, the study validates the protective role of SC formulations and signifies its novel potential for treating DN.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Syzygium , Humanos , NF-kappa B/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Fibronectinas , Fator A de Crescimento do Endotélio Vascular , Reação de Maillard , Interleucina-6 , Células HEK293 , Fator de Necrose Tumoral alfa
4.
Life Sci ; 339: 122416, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38216120

RESUMO

AIM: Telomerase expression is unique to cancer cells, making it a promising target for therapy. However, a major drawback of telomerase inhibition is that it affects cancer cell proliferation only when telomeres shorten, creating a lag phase post-continuous drug treatment. Acute cytotoxicity of telomerase inhibitors is dependent on their ability to induce DNA damage. p53 senses DNA damage and is the primary effector required for sensitizing cells towards apoptosis. MAIN METHODS: Isogenic p53+/+ and p53-/- ovarian cancer cell lines were generated using the CRISPR/Cas9 system and the anti-cancer effect of telomerase inhibitors MST-312 and BIBR1532 were determined. Flow cytometry, real-time PCR, and western blot were performed to study cell cycle, apoptosis, and gene expression. KEY FINDINGS: We report that MST-312 exhibits p53-dependent cytotoxicity, while BIBR1532 exhibits p53-independent cytotoxicity. Colony-forming ability also confirms the p53-dependent effect of MST-312. Re-expression of p53 in p53-/- cells could rescue MST-312 sensitivity. In p53+/+ cells, MST-312 causes S phase arrest and activation of p53-dependent target genes like anti-apoptosis markers (Fas and Puma) and cell cycle markers (p21 and cyclinB). In p53-/- cells, MST-312 causes S/G2/M arrest. BIBR1532 induces S/G2/M phase cell cycle arrest irrespective of p53 status. This correlates with the expression of the DNA damage marker (γ-H2AX). Long-term continuous treatment with MST-312 or BIBR1532 results in p53-independent telomere shortening. SIGNIFICANCE: In summary, we demonstrate that acute anti-cancer effects of MST-312 are dependent on p53 expression. Hence, it is important to consider the p53 expression status in cancer cells when selecting and administering telomerase inhibitors.


Assuntos
Aminobenzoatos , Benzamidas , Naftalenos , Neoplasias , Telomerase , Telomerase/genética , Telomerase/metabolismo , Proteína Supressora de Tumor p53/genética , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Apoptose , Neoplasias/tratamento farmacológico , Neoplasias/genética
5.
J Vis Exp ; (194)2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37154569

RESUMO

Telomeres are repetitive sequences which are present at chromosomal ends; their shortening is a characteristic feature of human somatic cells. Shortening occurs due to a problem with end replication and the absence of the telomerase enzyme, which is responsible for maintaining telomere length. Interestingly, telomeres also shorten in response to various internal physiological processes, like oxidative stress and inflammation, which may be impacted due to extracellular agents like pollutants, infectious agents, nutrients, or radiation. Thus, telomere length serves as an excellent biomarker of aging and various physiological health parameters. The TAGGG telomere length assay kit is used to quantify average telomere lengths using the telomere restriction fragment (TRF) assay and is highly reproducible. However, it is an expensive method, and because of this, it is not employed routinely for large sample numbers. Here, we describe a detailed protocol for an optimized and cost-effective measurement of telomere length using Southern blots or TRF analysis and non-radioactive chemiluminescence-based detection.


Assuntos
Envelhecimento , Telomerase , Humanos , Telômero/genética , Telômero/metabolismo , Telomerase/genética , Telomerase/metabolismo , Estresse Oxidativo
6.
Transl Oncol ; 27: 101569, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36274541

RESUMO

Quercetin is a natural flavonoid with well-established anti-proliferative activities against a variety of cancers. Telomerase inhibitor MST-312 also exhibits anti-proliferative effect on various cancer cells independent of its effect on telomere shortening. However, due to their low absorption and toxicity at higher doses, their clinical development is limited. In the present study, we examine the synergistic potential of their combination in cancer cells, which may result in a decrease in the therapeutic dosage of these compounds. We report that MST-312 and quercetin exhibit strong synergism in ovarian cancer cells with combination index range from 0.2 to 0.7. Co-treatment with MST-312 and quercetin upregulates the DNA damage and augments apoptosis when compared to treatment with either compound alone or a vehicle. We also examined the effect of these compounds on the proliferation of normal ovarian surface epithelial cells (OSEs). MST-312 has a cytoprotective impact in OSEs at lower dosages, but is inhibitory at higher doses. Quercetin did not affect the OSEs proliferation at low concentrations while at higher concentrations it is inhibitory. Notably, combination of MST-312 and quercetin had no discernible impact on OSEs. These observations have significant implications for future efforts towards maximizing efficacy in cancer therapeutics as this co-treatment specifically affects cancer cells and reduces the effective dosage of both the compounds.

7.
Anticancer Agents Med Chem ; 22(3): 469-484, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34102988

RESUMO

DNA's integrity is continuously challenged by intrinsic cellular processes and environmental agents. To overcome this genomic damage, cells have developed multiple signalling pathways collectively named as DNA Damage Response (DDR) and composed of three components: (i) sensor proteins, which detect DNA damage, (ii) mediators that relay the signal downstream and recruit the repair machinery and (iii) the repair proteins, which restore the damaged DNA. A flawed DDR and failure to repair the damage lead to the accumulation of genetic lesions and increased genomic instability, which is recognized as a hallmark of cancer. Cancer cells tend to harbor increased mutations in DDR genes and often have fewer DDR pathways than normal cells. This makes cancer cells more dependent on particular DDR pathways and thus become more susceptible to compounds inhibiting those pathways compared to normal cells, which have all the DDR pathways intact. Understanding the roles of different DDR proteins in the DNA damage response and repair pathways and the identification of their structures have paved the way for development of their inhibitors as targeted cancer therapy. In this review, we describe the major participants of various DDR pathways, their significance in carcinogenesis and focus on the inhibitors developed against several key DDR proteins.


Assuntos
Antineoplásicos/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Antineoplásicos/química , Dano ao DNA , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo
8.
J Biol Chem ; 285(7): 4554-61, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19959471

RESUMO

The abundance of cyclin-dependent kinase inhibitor p27(Kip1) during the cell cycle determines whether cells will proliferate or become quiescent. Although the post-translational regulation of p27(Kip1) is well established, its transcriptional regulation is poorly understood. Here, we report that mitogenic stimulation of quiescent HEK293 and Huh7 cells showed a rapid decline in the levels of p27(Kip1) transcript by 2.4 +/- 0.1-fold. Inhibition of the p27(Kip1) gene in response to mitogens involved transcriptional down-regulation and required newly synthesized protein(s). Mutation of the AP-1 element at position -469 in the human p27(Kip1) promoter abrogated the effect of mitogens. The recruitment of the AP-1 complex to the p27(Kip1) promoter was confirmed by in vitro DNA binding and chromatin immunoprecipitation studies. Reporter gene analysis combined with enforced expression of Jun/Fos proteins suggested the involvement of Jun/Fos heterodimer in the transrepression process. Both MAPK and phosphatidylinositol 3-kinase signaling pathways appeared to mediate p27(Kip1) transcription. Furthermore, hepatitis B virus X protein-mediated down-regulation of p27(Kip1) in a transgenic environment correlated with an increase in c-Fos levels, reiterating the physiological relevance of AP-1 in the transcriptional regulation of p27(Kip1). Collectively, our studies present the first evidence demonstrating the role of the AP-1 complex in transcriptional down-regulation of the p27(Kip1) gene following mitogenic stimulation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fator de Transcrição AP-1/fisiologia , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Cromonas/farmacologia , Biologia Computacional , Inibidor de Quinase Dependente de Ciclina p27/genética , Ensaio de Desvio de Mobilidade Eletroforética , Flavonoides/farmacologia , Citometria de Fluxo , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Multimerização Proteica/genética , Multimerização Proteica/fisiologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição AP-1/genética
9.
Cancer Lett ; 502: 120-132, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33450357

RESUMO

Long non-coding RNAs (lncRNAs) are known to regulate various biological processes including cancer. Cancer cells possess limitless replicative potential which is attained by telomere length maintenance while normal somatic cells have a limited lifespan because their telomeres shorten with every cell division ultimately triggering replicative senescence. Two lncRNAs have been observed to play a key role in telomere length maintenance. First is the lncRNA TERC (telomerase RNA component) which functions as a template for telomeric DNA synthesis in association with telomerase reverse transcriptase (TERT) which serves as the catalytic component. Together they constitute the telomerase complex which functions as a reverse transcriptase to elongate telomeres. Second lncRNA that helps in regulating telomere length is the telomeric repeat-containing RNA (TERRA) which is transcribed from the subtelomeric region and extends to the telomeric region. TERC and TERRA exhibit important functions in cancer with implications in precision oncology. In this review, we discuss various aspects of these important lncRNAs in humans and their role in cancer along with recent advancements in their anticancer therapeutic application.


Assuntos
Neoplasias/genética , RNA Longo não Codificante/genética , RNA/genética , Telomerase/genética , Telômero/metabolismo , Humanos , Neoplasias/metabolismo , Medicina de Precisão , Telomerase/metabolismo
10.
Environ Toxicol Pharmacol ; 85: 103633, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33711516

RESUMO

External environment affects cellular physiological processes and impact the stability of our genome. The most important structural components of our linear chromosomes which endure the impact by these agents, are the chromosomal ends called telomeres. Telomeres preserve the integrity of our genome by preventing end to end fusions and telomeric loss through by inhibiting DNA damage response (DDR) activation. This is accomplished by the presence of a six membered shelterin complex at telomeres. Further, telomeres cannot be replicated by normal DNA polymerase and require a special enzyme called telomerase which is expressed only in stem cells, few immune cells and germ cells. Telomeres are rich in guanine content and thus become extremely prone to damage arising due to physiological processes like oxidative stress and inflammation. External environmental factors which includes various physical, biological and chemical agents also affect telomere homeostasis by increasing oxidative stress and inflammation. In the present review, we highlight the effect of these external factors on telomerase activity and telomere length. We also discuss how the external agents affect the physiological processes, thus modulating telomere stability. Further, we describe its implication in the development of aging and its related pathologies.


Assuntos
Poluentes Ambientais/toxicidade , Telomerase/metabolismo , Telômero/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Humanos
11.
Exp Hematol ; 90: 12-17, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32858091

RESUMO

Mammalian Rap1 is a part of the telomere binding complex named shelterin and is one of the most conserved telomeric proteins. With its essential requirement in lower species to its becoming necessary in higher species, it appears to have gained and lost several functions simultaneously evolving with telomeres. Mammalian Rap1 has been reported to play a role in inflammation, metabolism, and oxidative stress. Mammalian Rap1 has also been found to regulate DNA damage response from telomeres in senescent cells. Recently our group uncovered its novel role in stem cell maintenance, and modulation of the chemotherapeutic response. Mechanistically it was found to function as an adaptor via protein-protein interactions and to modulate the response to DNA damage. In the current review we highlight newly identified functions of Rap1 in regulating telomeric and general DNA damage response with its impact at the cellular and organismal levels.


Assuntos
Dano ao DNA , Homeostase , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Células-Tronco Neoplásicas , Proteínas de Ligação a Telômeros/metabolismo , Animais , Senescência Celular , Humanos , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Complexo Shelterina , Transdução de Sinais , Telômero/genética , Telômero/metabolismo , Telômero/patologia , Proteínas de Ligação a Telômeros/genética
12.
Cancers (Basel) ; 12(7)2020 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-32674474

RESUMO

Telomeres are the ends of linear chromosomes comprised of repetitive nucleotide sequences in humans. Telomeres preserve chromosomal stability and genomic integrity. Telomere length shortens with every cell division in somatic cells, eventually resulting in replicative senescence once telomere length becomes critically short. Telomere shortening can be overcome by telomerase enzyme activity that is undetectable in somatic cells, while being active in germline cells, stem cells, and immune cells. Telomeres are bound by a shelterin complex that regulates telomere lengthening as well as protects them from being identified as DNA damage sites. Telomeres are transcribed by RNA polymerase II, and generate a long noncoding RNA called telomeric repeat-containing RNA (TERRA), which plays a key role in regulating subtelomeric gene expression. Replicative immortality and genome instability are hallmarks of cancer and to attain them cancer cells exploit telomere maintenance and telomere protection mechanisms. Thus, understanding the role of telomeres and their associated proteins in cancer initiation, progression and treatment is very important. The present review highlights the critical role of various telomeric components with recently established functions in cancer. Further, current strategies to target various telomeric components including human telomerase reverse transcriptase (hTERT) as a therapeutic approach in human malignancies are discussed.

14.
Nat Commun ; 10(1): 5349, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31836706

RESUMO

Increased levels and non-telomeric roles have been reported for shelterin proteins, including RAP1 in cancers. Herein using Rap1 null mice, we provide the genetic evidence that mammalian Rap1 plays a major role in hematopoietic stem cell survival, oncogenesis and response to chemotherapy. Strikingly, this function of RAP1 is independent of its association with the telomere or with its known partner TRF2. We show that RAP1 interacts with many members of the DNA damage response (DDR) pathway. RAP1 depleted cells show reduced interaction between XRCC4/DNA Ligase IV and DNA-PK, and are impaired in DNA Ligase IV recruitment to damaged chromatin for efficient repair. Consistent with its role in DNA damage repair, RAP1 loss decreases double-strand break repair via NHEJ in vivo, and consequently reduces B cell class switch recombination. Finally, we discover that RAP1 levels are predictive of the success of chemotherapy in breast and colon cancer.


Assuntos
Antineoplásicos/farmacologia , Carcinogênese/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , DNA Ligase Dependente de ATP/metabolismo , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Proteína Quinase Ativada por DNA/metabolismo , Fluoruracila/farmacologia , Raios gama , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/efeitos da radiação , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos da radiação , Humanos , Camundongos Knockout , Mutagênicos/toxicidade , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/efeitos da radiação , Proteínas Proto-Oncogênicas c-myc/metabolismo , Complexo Shelterina , Análise de Sobrevida
15.
Front Cell Dev Biol ; 5: 1, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28184371

RESUMO

Telomerase elongates telomeres and is crucial for maintaining genomic stability. While stem cells and cancer cells display high telomerase activity, normal somatic cells lack telomerase activity primarily due to transcriptional repression of telomerase reverse transcriptase (TERT), the catalytic component of telomerase. Transcription factor binding, chromatin status as well as epigenetic modifications at the TERT promoter regulates TERT transcription. Myc is an important transcriptional regulator of TERT that directly controls its expression by promoter binding and associating with other transcription factors. In this review, we discuss the current understanding of the molecular mechanisms behind regulation of TERT transcription by Myc. We also discuss future perspectives in investigating the regulation of Myc at TERT promoter during cancer development.

16.
Cancer Discov ; 6(11): 1276-1291, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27650951

RESUMO

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.


Assuntos
Neoplasias/genética , Telomerase/genética , Ativação Transcricional/genética , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Mutação , Neoplasias/patologia , Regiões Promotoras Genéticas , Telomerase/metabolismo
17.
J Clin Invest ; 126(10): 4045-4060, 2016 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-27643433

RESUMO

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.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , RNA de Transferência/genética , Telomerase/fisiologia , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Feminino , Células HEK293 , Humanos , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias , Regiões Promotoras Genéticas , Ligação Proteica , Biossíntese de Proteínas , Domínios e Motivos de Interação entre Proteínas , RNA Polimerase III/metabolismo , RNA de Transferência/metabolismo
18.
Cell Rep ; 15(12): 2597-607, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27292646

RESUMO

Incomplete knowledge of the mechanisms at work continues to hamper efforts to maximize reprogramming efficiency. Here, we present a systematic genome-wide RNAi screen to determine the global regulators during the early stages of human reprogramming. Our screen identifies functional repressors and effectors that act to impede or promote the reprogramming process. Repressors and effectors form close interacting networks in pathways, including RNA processing, G protein signaling, protein ubiquitination, and chromatin modification. Combinatorial knockdown of five repressors (SMAD3, ZMYM2, SFRS11, SAE1, and ESET) synergistically resulted in ∼85% TRA-1-60-positive cells. Removal of the novel splicing factor SFRS11 during reprogramming is accompanied by rapid acquisition of pluripotency-specific spliced forms. Mechanistically, SFRS11 regulates exon skipping and mutually exclusive splicing of transcripts in genes involved in cell differentiation, mRNA splicing, and chromatin modification. Our study provides insights into the reprogramming process, which comprises comprehensive and multi-layered transcriptional, splicing, and epigenetic machineries.


Assuntos
Reprogramação Celular/genética , Interferência de RNA , Células Cultivadas , Técnicas de Silenciamento de Genes , Testes Genéticos , Genoma Humano , Humanos , Cinética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Splicing de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Processamento de Serina-Arginina/metabolismo
19.
J Clin Invest ; 125(5): 2109-22, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25893605

RESUMO

Constitutively active MYC and reactivated telomerase often coexist in cancers. While reactivation of telomerase is thought to be essential for replicative immortality, MYC, in conjunction with cofactors, confers several growth advantages to cancer cells. It is known that the reactivation of TERT, the catalytic subunit of telomerase, is limiting for reconstituting telomerase activity in tumors. However, while reactivation of TERT has been functionally linked to the acquisition of several "hallmarks of cancer" in tumors, the molecular mechanisms by which this occurs and whether these mechanisms are distinct from the role of telomerase on telomeres is not clear. Here, we demonstrated that first-generation TERT-null mice, unlike Terc-null mice, show delayed onset of MYC-induced lymphomagenesis. We further determined that TERT is a regulator of MYC stability in cancer. TERT stabilized MYC levels on chromatin, contributing to either activation or repression of its target genes. TERT regulated MYC ubiquitination and proteasomal degradation, and this effect of TERT was independent of its reverse transcriptase activity and role in telomere elongation. Based on these data, we conclude that reactivation of TERT, a direct transcriptional MYC target in tumors, provides a feed-forward mechanism to potentiate MYC-dependent oncogenesis.


Assuntos
Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Telomerase/fisiologia , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Retroalimentação Fisiológica , Genes myc , Quinase 3 da Glicogênio Sintase/fisiologia , Glicogênio Sintase Quinase 3 beta , Xenoenxertos , Humanos , Linfoma não Hodgkin/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Fosforilação , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA/genética , RNA/fisiologia , Interferência de RNA , Telomerase/deficiência , Telomerase/genética , Homeostase do Telômero/genética , Fatores de Tempo , Transcrição Gênica , Ubiquitinação
20.
FEBS J ; 279(7): 1220-30, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22309289

RESUMO

Hepatitis B virus X protein (HBx) is a putative viral oncoprotein that plays an important role in various cellular processes, including modulation of the phosphatidylinositol 3-kinase/Akt signalling pathway. However, the molecular mechanism of Akt activation remains elusive. Here we show that HBx interacts with Akt1 kinase and is phosphorylated at serine 31 as indicated by mutational analysis of the Akt recognition motif (creating the HBxS31A mutant) or immunoblotting of HBx immunoprecipitates using Akt motif-specific antibody. The Akt-dependent phosphorylation of HBx was abrogated in the presence of the phosphatidylinositol 3-kinase inhibitor LY294002 or Akt1 gene silencing by specific siRNA. Co-immunoprecipitation studies provided evidence for HBx-Akt interaction in a cellular environment. This interaction was also confirmed in hepatoma HepG2.2.15 cells in which HBx was expressed at physiological levels from the integrated hepatitis B viral genome. The HBx-Akt interaction was essential for Akt signalling, and involved displacement of the Akt-bound negative regulator 'C-terminal modulator protein' by HBx. HBx-activated Akt phosphorylated its downstream target glycogen synthase kinase 3ß, leading to stabilization of ß-catenin, while p65 phosphorylation resulted in enhanced promoter recruitment and expression of target genes encoding cyclin D1 and Bcl-XL. Further, the oncogenic potential of HBx was significantly augmented in the presence of Akt in a soft agar colony formation assay. Together, these results suggest that oncogenic co-operation between HBx and Akt may be important for cell proliferation, abrogation of apoptosis and tumorigenic transformation of cells.


Assuntos
Vírus da Hepatite B/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia , Transativadores/genética , Proteínas Virais Reguladoras e Acessórias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA