237 results on '"Tergaonkar, V."'
Search Results
2. Mitochondrial uncoupling reveals a novel therapeutic opportunity for p53-defective cancers
- Author
-
Kumar, R., Coronel, L., Somalanka, B., Raju, A., Aning, O. A., An, O., Ho, Y. S., Chen, S., Mak, S. Y., Hor, P. Y., Yang, H., Lakshmanan, M., Itoh, H., Tan, S. Y., Lim, Y. K., Wong, A. P. C., Chew, S. H., Huynh, T. H., Goh, B. C., Lim, C. Y., Tergaonkar, V., and Cheok, C. F.
- Published
- 2018
- Full Text
- View/download PDF
3. Mouse models for core binding factor leukemia
- Author
-
Chin, D W L, Watanabe-Okochi, N, Wang, C Q, Tergaonkar, V, and Osato, M
- Published
- 2015
- Full Text
- View/download PDF
4. Cbfb deficiency results in differentiation blocks and stem/progenitor cell expansion in hematopoiesis
- Author
-
Wang, C Q, Chin, D W L, Chooi, J Y, Chng, W J, Taniuchi, I, Tergaonkar, V, and Osato, M
- Published
- 2015
- Full Text
- View/download PDF
5. RUNX1 meets MLL: epigenetic regulation of hematopoiesis by two leukemia genes
- Author
-
Koh, C P, Wang, C Q, Ng, C E L, Ito, Y, Araki, M, Tergaonkar, V, Huang, G, and Osato, M
- Published
- 2013
- Full Text
- View/download PDF
6. Nuclear factor κB (NF-κB) suppresses food intake and energy expenditure in mice by directly activating the Pomc promoter
- Author
-
Shi, X., Wang, X., Li, Q., Su, M., Chew, E., Wong, E. T., Lacza, Z., Radda, G. K., Tergaonkar, V., and Han, W.
- Published
- 2013
- Full Text
- View/download PDF
7. Coding and non-coding roles of MOCCI (C15ORF48) coordinate to regulate host inflammation and immunity
- Author
-
Lee, CQE, Kerouanton, B, Chothani, S, Zhang, S, Chen, Y, Mantri, CK, Hock, DH, Lim, R, Nadkarni, R, Vinh, TH, Lim, D, Chew, WL, Zhong, FL, Stroud, DA, Schafer, S, Tergaonkar, V, St John, AL, Rackham, OJL, Ho, L, Lee, CQE, Kerouanton, B, Chothani, S, Zhang, S, Chen, Y, Mantri, CK, Hock, DH, Lim, R, Nadkarni, R, Vinh, TH, Lim, D, Chew, WL, Zhong, FL, Stroud, DA, Schafer, S, Tergaonkar, V, St John, AL, Rackham, OJL, and Ho, L
- Abstract
Mito-SEPs are small open reading frame-encoded peptides that localize to the mitochondria to regulate metabolism. Motivated by an intriguing negative association between mito-SEPs and inflammation, here we screen for mito-SEPs that modify inflammatory outcomes and report a mito-SEP named "Modulator of cytochrome C oxidase during Inflammation" (MOCCI) that is upregulated during inflammation and infection to promote host-protective resolution. MOCCI, a paralog of the NDUFA4 subunit of cytochrome C oxidase (Complex IV), replaces NDUFA4 in Complex IV during inflammation to lower mitochondrial membrane potential and reduce ROS production, leading to cyto-protection and dampened immune response. The MOCCI transcript also generates miR-147b, which targets the NDUFA4 mRNA with similar immune dampening effects as MOCCI, but simultaneously enhances RIG-I/MDA-5-mediated viral immunity. Our work uncovers a dual-component pleiotropic regulation of host inflammation and immunity by MOCCI (C15ORF48) for safeguarding the host during infection and inflammation.
- Published
- 2021
8. The control of cell energy metabolism by NF-κB transcription factors: W09.001
- Author
-
Mauro, C., Leow, S. C., Anso, E., Rocha, S., Thotakura, A. K., Tornatore, L., Moretti, M., De Smaele, E., Beg, A. A., Tergaonkar, V., Chandel, N. S., and Franzoso, G.
- Published
- 2012
9. The expanding roles of long non-coding RNAs in the regulation of cancer stem cells
- Author
-
Ma, Z., Wang, Y., Xin, H., Wang, L., Arfuso, Frank, Dharmarajan, A., Kumar, A., Wang, H., Tang, F., Warrier, Sudha, Tergaonkar, V., Sethi, G., Ma, Z., Wang, Y., Xin, H., Wang, L., Arfuso, Frank, Dharmarajan, A., Kumar, A., Wang, H., Tang, F., Warrier, Sudha, Tergaonkar, V., and Sethi, G.
- Abstract
Long non-coding RNAs (lncRNAs) are a novel class of gene regulators playing multifaceted roles in physiological processes as well as pathological conditions such as cancer. Cancer stem cells (CSCs) are a small subset of tumor cells that constitute the origin and development of various malignant tumors. CSCs have been identified in a wide spectrum of human tumors and could act as a critical link underlying the processes of tumor metastasis and recurrence. Mounting evidence indicates that lncRNAs are aberrantly expressed in diverse CSCs and regulate CSC properties at different molecular levels. Here, we very briefly summarize the recent findings on the potential roles of lncRNAs in regulating various functions of CSCs, and elaborate on how can lncRNAs impact CSC properties via interacting with other macromolecules at the epigenetic, transcriptional, and post-transcriptional levels. This mini-review also highlights the understanding of the modular regulatory principles of lncRNA interactions in CSCs.
- Published
- 2019
10. Accumulation of JAK activation loop phosphorylation is linked to type I JAK inhibitor withdrawal syndrome in myelofibrosis
- Author
-
Tvorogov, D, Thomas, D, Liau, NPD, Dottore, M, Barry, EF, Lathi, M, Kan, WL, Hercus, TR, Stomski, F, Hughes, TP, Tergaonkar, V, Parker, MW, Ross, DM, Majeti, R, Babon, JJ, Lopez, AF, Tvorogov, D, Thomas, D, Liau, NPD, Dottore, M, Barry, EF, Lathi, M, Kan, WL, Hercus, TR, Stomski, F, Hughes, TP, Tergaonkar, V, Parker, MW, Ross, DM, Majeti, R, Babon, JJ, and Lopez, AF
- Abstract
Treatment of patients with myelofibrosis with the type I JAK (Janus kinase) inhibitor ruxolitinib paradoxically induces JAK2 activation loop phosphorylation and is associated with a life-threatening cytokine-rebound syndrome if rapidly withdrawn. We developed a time-dependent assay to mimic ruxolitinib withdrawal in primary JAK2V617F and CALR mutant myelofibrosis patient samples and observed notable activation of spontaneous STAT signaling in JAK2V617F samples after drug washout. Accumulation of ruxolitinib-induced JAK2 phosphorylation was dose dependent and correlated with rebound signaling and the presence of a JAK2V617F mutation. Ruxolitinib prevented dephosphorylation of a cryptic site involving Tyr1007/1008 in JAK2 blocking ubiquitination and degradation. In contrast, a type II JAK inhibitor, CHZ868, did not induce JAK2 phosphorylation, was not associated with withdrawal signaling, and was superior in the eradication of flow-purified JAK2V617F mutant CD34+ progenitors after drug washout. Type I inhibitor-induced loop phosphorylation may act as a pathogenic signaling node released upon drug withdrawal, especially in JAK2V617F patients.
- Published
- 2018
11. Rare variants in Fanconi anemia genes are enriched in acute myeloid leukemia
- Author
-
Maung, KZY, Leo, PJ, Bassal, M, Casolari, DA, Gray, JX, Bray, SC, Pederson, S, Singhal, D, Samaraweera, SE, Tran, N, Cildir, G, Marshall, M, Ewing, A, Duncan, EL, Brown, MA, Saal, R, Tergaonkar, V, To, LB, Marlton, P, Gill, D, Lewis, I, Deans, AJ, Brown, AL, D'Andrea, RJ, Gonda, TJ, Maung, KZY, Leo, PJ, Bassal, M, Casolari, DA, Gray, JX, Bray, SC, Pederson, S, Singhal, D, Samaraweera, SE, Tran, N, Cildir, G, Marshall, M, Ewing, A, Duncan, EL, Brown, MA, Saal, R, Tergaonkar, V, To, LB, Marlton, P, Gill, D, Lewis, I, Deans, AJ, Brown, AL, D'Andrea, RJ, and Gonda, TJ
- Published
- 2018
12. Evidence for the Involvement of the Master Transcription Factor NF-?B in Cancer Initiation and Progression.
- Author
-
Puar, Y., Shanmugam, M., Fan, L., Arfuso, Frank, Sethi, G., Tergaonkar, V., Puar, Y., Shanmugam, M., Fan, L., Arfuso, Frank, Sethi, G., and Tergaonkar, V.
- Abstract
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-?B) is responsible for the regulation of a large number of genes that are involved in important physiological processes, including survival, inflammation, and immune responses. At the same time, this transcription factor can control the expression of a plethora of genes that promote tumor cell proliferation, survival, metastasis, inflammation, invasion, and angiogenesis. The aberrant activation of this transcription factor has been observed in several types of cancer and is known to contribute to aggressive tumor growth and resistance to therapeutic treatment. Although NF-?B has been identified to be a major contributor to cancer initiation and development, there is evidence revealing its role in tumor suppression. This review briefly highlights the major mechanisms of NF-?B activation, the role of NF-?B in tumor promotion and suppression, as well as a few important pharmacological strategies that have been developed to modulate NF-?B function.
- Published
- 2018
13. Wanted DEAD/H or Alive: Helicases Winding Up in Cancers.
- Author
-
Cai, W., Xiong Chen, Z., Rane, G., Satendra Singh, S., Choo, Z., Wang, C., Yuan, Y., Zea Tan, T., Arfuso, Frank, Yap, C., Pongor, L., Yang, H., Lee, M., Cher Goh, B., Sethi, G., Benoukraf, T., Tergaonkar, V., Prem Kumar, A., Cai, W., Xiong Chen, Z., Rane, G., Satendra Singh, S., Choo, Z., Wang, C., Yuan, Y., Zea Tan, T., Arfuso, Frank, Yap, C., Pongor, L., Yang, H., Lee, M., Cher Goh, B., Sethi, G., Benoukraf, T., Tergaonkar, V., and Prem Kumar, A.
- Abstract
Cancer is one of the most studied areas of human biology over the past century. Despite having attracted much attention, hype, and investments, the search to find a cure for cancer remains an uphill battle. Recent discoveries that challenged the central dogma of molecular biology not only further increase the complexity but also demonstrate how various types of noncoding RNAs such as microRNA and long noncoding RNA, as well as their related processes such as RNA editing, are important in regulating gene expression. Parallel to this aspect, an increasing number of reports have focused on a family of proteins known as DEAD/H-box helicases involved in RNA metabolism, regulation of long and short noncoding RNAs, and novel roles as "editing helicases" and their association with cancers. This review summarizes recent findings on the roles of RNA helicases in various cancers, which are broadly classified into adult solid tumors, childhood solid tumors, leukemia, and cancer stem cells. The potential small molecule inhibitors of helicases and their therapeutic value are also discussed. In addition, analyzing next-generation sequencing data obtained from public portals and reviewing existing literature, we provide new insights on the potential of DEAD/H-box helicases to act as pharmacological drug targets in cancers.
- Published
- 2017
14. Aberrant activation of the GIMAP enhancer by oncogenic transcription factors in T-cell acute lymphoblastic leukemia
- Author
-
Liau, W S, primary, Tan, S H, additional, Ngoc, P C T, additional, Wang, C Q, additional, Tergaonkar, V, additional, Feng, H, additional, Gong, Z, additional, Osato, M, additional, Look, A T, additional, and Sanda, T, additional
- Published
- 2016
- Full Text
- View/download PDF
15. Antisense oligonucleotide-mediated MDM4 exon 6 skipping impairs tumor growth
- Author
-
Dewaele, M, Tabaglio, T, Willekens, K, Bezzi, M, Teo, SX, Low, DHP, Koh, CM, Rambow, F, Fiers, M, Rogiers, A, Radaelli, E, Al-Haddawi, M, Tan, SY, Hermans, E, Amant, F, Yan, H, Lakshmanan, M, Koumar, RC, Lim, ST, Derheimer, FA, Campbell, RM, Bonday, Z, Tergaonkar, V, Shackleton, M, Blattner, C, Marine, J-C, Guccione, E, Dewaele, M, Tabaglio, T, Willekens, K, Bezzi, M, Teo, SX, Low, DHP, Koh, CM, Rambow, F, Fiers, M, Rogiers, A, Radaelli, E, Al-Haddawi, M, Tan, SY, Hermans, E, Amant, F, Yan, H, Lakshmanan, M, Koumar, RC, Lim, ST, Derheimer, FA, Campbell, RM, Bonday, Z, Tergaonkar, V, Shackleton, M, Blattner, C, Marine, J-C, and Guccione, E
- Abstract
MDM4 is a promising target for cancer therapy, as it is undetectable in most normal adult tissues but often upregulated in cancer cells to dampen p53 tumor-suppressor function. The mechanisms that underlie MDM4 upregulation in cancer cells are largely unknown. Here, we have shown that this key oncogenic event mainly depends on a specific alternative splicing switch. We determined that while a nonsense-mediated, decay-targeted isoform of MDM4 (MDM4-S) is produced in normal adult tissues as a result of exon 6 skipping, enhanced exon 6 inclusion leads to expression of full-length MDM4 in a large number of human cancers. Although this alternative splicing event is likely regulated by multiple splicing factors, we identified the SRSF3 oncoprotein as a key enhancer of exon 6 inclusion. In multiple human melanoma cell lines and in melanoma patient-derived xenograft (PDX) mouse models, antisense oligonucleotide-mediated (ASO-mediated) skipping of exon 6 decreased MDM4 abundance, inhibited melanoma growth, and enhanced sensitivity to MAPK-targeting therapeutics. Additionally, ASO-based MDM4 targeting reduced diffuse large B cell lymphoma PDX growth. As full-length MDM4 is enhanced in multiple human tumors, our data indicate that this strategy is applicable to a wide range of tumor types. We conclude that enhanced MDM4 exon 6 inclusion is a common oncogenic event and has potential as a clinically compatible therapeutic target.
- Published
- 2016
16. RNA helicase DP103 and TAK1: a new connection in cancer
- Author
-
Shin, E., Osato, M., Kumar, Alan Prem, Tergaonkar, V., Shin, E., Osato, M., Kumar, Alan Prem, and Tergaonkar, V.
- Published
- 2015
17. Cancer prevention and therapy through the modulation of transcription factors by bioactive natural compounds
- Author
-
Shanmugam, M., Lee, J., Chai, E., Kanchi, M., Kar, S., Arfuso, Frank, Dharmarajan, A., Kumar, A., Ramar, P., Looi, C., Mustafa, M., Tergaonkar, V., Bishayee, A., Ahn, K., Sethi, G., Shanmugam, M., Lee, J., Chai, E., Kanchi, M., Kar, S., Arfuso, Frank, Dharmarajan, A., Kumar, A., Ramar, P., Looi, C., Mustafa, M., Tergaonkar, V., Bishayee, A., Ahn, K., and Sethi, G.
- Abstract
The association between chronic inflammation and cancer development has been well documented. One of the major obstacles in cancer treatment is the persistent autocrine and paracrine activation of pro-inflammatory transcription factors such as nuclear factor-?B, signal transducer and activator of transcription 3, activator protein 1, fork head box protein M1, and hypoxia-inducible factor 1a in a wide variety of tumor cell lines and patient specimens. This, in turn, leads to an accelerated production of cellular adhesion molecules, inflammatory cytokines, chemokines, anti-apoptotic molecules, and inducible nitric oxide synthase. Numerous medicinal plant-derived compounds have made a tremendous impact in drug discovery research endeavors, and have been reported to modulate the activation of diverse oncogenic transcription factors in various tumor models. Moreover, novel therapeutic combinations of standard chemotherapeutic drugs with these agents have significantly improved patient survival by making cancer cells more susceptible to chemotherapy and radiotherapy. In this review, we critically analyze the existing literature on the modulation of diverse transcription factors by various natural compounds and provide views on new directions for accelerating the discovery of novel drug candidates derived from Mother Nature.
- Published
- 2015
18. Paracrine regulation in mesenchymal stem cells: the role of Rap1
- Author
-
Zhang, Y, primary, Zhang, Z, additional, Gao, F, additional, Tse, H-F, additional, Tergaonkar, V, additional, and Lian, Q, additional
- Published
- 2015
- Full Text
- View/download PDF
19. Rap1-mediated nuclear factor-kappaB (NF-κB) activity regulates the paracrine capacity of mesenchymal stem cells in heart repair following infarction
- Author
-
Zhang, Y, primary, Chiu, S, additional, Liang, X, additional, Gao, F, additional, Zhang, Z, additional, Liao, S, additional, Liang, Y, additional, Chai, Y-H, additional, Low, D J H, additional, Tse, H-F, additional, Tergaonkar, V, additional, and Lian, Q, additional
- Published
- 2015
- Full Text
- View/download PDF
20. Hematopoiesis specific loss of Cdk2 and Cdk4 results in increased erythrocyte size and delayed platelet recovery following stress
- Author
-
Jayapal, S. R., primary, Wang, C. Q., additional, Bisteau, X., additional, Caldez, M. J., additional, Lim, S., additional, Tergaonkar, V., additional, Osato, M., additional, and Kaldis, P., additional
- Published
- 2015
- Full Text
- View/download PDF
21. Inhibition of RAP1 Enhances Corneal Recovery Following Alkali Injury
- Author
-
Poon, M. W., primary, Yan, L., additional, Jiang, D., additional, Qin, P., additional, Tse, H.-f., additional, Wong, I. Y., additional, Wong, D. S. H., additional, Tergaonkar, V., additional, and Lian, Q., additional
- Published
- 2015
- Full Text
- View/download PDF
22. γ-tocotrienol inhibits angiogenesis-dependent growth of human hepatocellular carcinoma through abrogation of AKT/mTOR pathway in an orthotopic mouse model
- Author
-
Siveen, K., Ahn, K., Ong, T., Shanmugam, M., Li, F., Yap, W., Kumar, Alan Prem, Fong, C., Tergaonkar, V., Hui, K., Sethi, G., Siveen, K., Ahn, K., Ong, T., Shanmugam, M., Li, F., Yap, W., Kumar, Alan Prem, Fong, C., Tergaonkar, V., Hui, K., and Sethi, G.
- Abstract
Angiogenesis is one of the key hallmarks of cancer. In this study, we investigated whether γ-tocotrienol can abrogate angiogenesis-mediated tumor growth in hepatocellular carcinoma (HCC) and if so, through what molecular mechanisms. We observed that γ-tocotrienol inhibited vascular endothelial growth factor (VEGF)-induced migration, invasion, tube formation and viability of HUVECs in vitro. Moreover, γ-tocotrienol reduced the number of capillary sprouts from matrigel embedded rat thoracic aortic ring in a dose-dependent manner. Also, in chick chorioallantoic membrane assay, γ-tocotrienol significantly reduced the blood vessels formation. We further noticed that γ-tocotrienol blocked angiogenesis in an in vivo matrigel plug assay. Furthermore, γ-tocotrienol inhibited VEGF-induced autophosphorylation of VEGFR2 in HUVECs and also suppressed the constitutive activation of AKT/mammalian target of rapamycin (mTOR) signal transduction cascades in HUVECs as well as in HCC cells. Interestingly, γ-tocotrienol was also found to significantly reduce the tumor growth in an orthotopic HCC mouse model and inhibit tumor-induced angiogenesis in HCC patient xenografts through the suppression of various biomarkers of proliferation and angiogenesis. Taken together, our findings strongly suggest that γ-tocotrienol might be a promising anti-angiogenic drug with significant antitumor activity in HCC.
- Published
- 2014
23. DEAD-box helicase DP103 defines metastatic potential of human breast cancers
- Author
-
Shin, E., Hay, H., Lee, M., Goh, J., Tan, T., Sen, Y., Lim, S., Yousef, E., Ong, H., Thike, A., Kong, X., Wu, Z., Mendoz, E., Sun, W., Salto-Tellez, M., Lim, C., Lobie, P., Lim, Y., Yap, C., Zeng, Q., Sethi, G., Tan, P., Goh, B., Miller, L., Thiery, J., Zhu, T., Gaboury, L., Hui, K., Yip, G., Miyamoto, S., Kumar, Alan Prem, Tergaonkar, V., Shin, E., Hay, H., Lee, M., Goh, J., Tan, T., Sen, Y., Lim, S., Yousef, E., Ong, H., Thike, A., Kong, X., Wu, Z., Mendoz, E., Sun, W., Salto-Tellez, M., Lim, C., Lobie, P., Lim, Y., Yap, C., Zeng, Q., Sethi, G., Tan, P., Goh, B., Miller, L., Thiery, J., Zhu, T., Gaboury, L., Hui, K., Yip, G., Miyamoto, S., Kumar, Alan Prem, and Tergaonkar, V.
- Abstract
Despite advancement in breast cancer treatment, 30% of patients with early breast cancers experience relapse with distant metastasis. It is a challenge to identify patients at risk for relapse; therefore, the identification of markers and therapeutic targets for metastatic breast cancers is imperative. Here, we identified DP103 as a biomarker and metastasis-driving oncogene in human breast cancers and determined that DP103 elevates matrix metallopeptidase 9 (MMP9) levels, which are associated with metastasis and invasion through activation of NF-κB. In turn, NF-κB signaling positively activated DP103 expression. Furthermore, DP103 enhanced TGF-β–activated kinase-1 (TAK1) phosphorylation of NF-κB–activating IκB kinase 2 (IKK2), leading to increased NF-κB activity. Reduction of DP103 expression in invasive breast cancer cells reduced phosphorylation of IKK2, abrogated NF-κB–mediated MMP9 expression, and impeded metastasis in a murine xenograft model. In breast cancer patient tissues, elevated levels of DP103 correlated with enhanced MMP9, reduced overall survival, and reduced survival after relapse. Together, these data indicate that a positive DP103/NF-κB feedback loop promotes constitutive NF-κB activation in invasive breast cancers and activation of this pathway is linked to cancer progression and the acquisition of chemotherapy resistance. Furthermore, our results suggest that DP103 has potential as a therapeutic target for breast cancer treatment.
- Published
- 2014
24. Negative regulation of signal transducer and activator of transcription-3 signalling cascade by lupeol inhibits growth and induces apoptosis in hepatocellular carcinoma cells
- Author
-
Siveen, K., Nguyen, A., Lee, J., Li, F., Singh, S., Kumar, Alan Prem, Low, G., Jha, S., Tergaonkar, V., Ahn, K., Sethi, G., Siveen, K., Nguyen, A., Lee, J., Li, F., Singh, S., Kumar, Alan Prem, Low, G., Jha, S., Tergaonkar, V., Ahn, K., and Sethi, G.
- Abstract
Background: Constitutive activation of signal transducer and activator of transcription signalling 3 (STAT3) has been linked with survival, proliferation and angiogenesis in a wide variety of malignancies including hepatocellular carcinoma (HCC). Methods: We evaluated the effect of lupeol on STAT3 signalling cascade and its regulated functional responses in HCC cells. Results: Lupeol suppressed constitutive activation of STAT3 phosphorylation at tyrosine 705 residue effectively in a dose- and time-dependent manner. The phosphorylation of Janus-activated kinases (JAKs) 1 and 2 and Src was also suppressed by lupeol. Pervanadate treatment reversed the downregulation of phospho-STAT3 induced by lupeol, thereby indicating the involvement of a phosphatase. Indeed, we observed that treatment with lupeol increased the protein and mRNA levels of SHP-2, and silencing of SHP-2 abolished the inhibitory effects of lupeol on STAT3 activation. Treatment with lupeol also downregulated the expression of diverse STAT3-regulated genes and decreased the binding of STAT3 to VEGF promoter. Moreover, the proliferation of various HCC cells was significantly suppressed by lupeol, being associated with substantial induction of apoptosis. Depletion of SHP-2 reversed the observed antiproliferative and pro-apoptotic effects of lupeol. Conclusions: Lupeol exhibited its potential anticancer effects in HCC through the downregulation of STAT3-induced pro-survival signalling cascade.
- Published
- 2014
25. Cbfb deficiency results in differentiation blocks and stem/progenitor cell expansion in hematopoiesis
- Author
-
Wang, C Q, primary, Chin, D W L, additional, Chooi, J Y, additional, Chng, W J, additional, Taniuchi, I, additional, Tergaonkar, V, additional, and Osato, M, additional
- Published
- 2014
- Full Text
- View/download PDF
26. Negative regulation of signal transducer and activator of transcription-3 signalling cascade by lupeol inhibits growth and induces apoptosis in hepatocellular carcinoma cells
- Author
-
Siveen, K S, primary, Nguyen, A H, additional, Lee, J H, additional, Li, F, additional, Singh, S S, additional, Kumar, A P, additional, Low, G, additional, Jha, S, additional, Tergaonkar, V, additional, Ahn, K S, additional, and Sethi, G, additional
- Published
- 2014
- Full Text
- View/download PDF
27. Multifaceted link between cancer and inflammation
- Author
-
Sethi, G., Shanmugam, M., Ramachandran, L., Kumar, Alan Prem, Tergaonkar, V., Sethi, G., Shanmugam, M., Ramachandran, L., Kumar, Alan Prem, and Tergaonkar, V.
- Abstract
Increasing evidence from epidemiological, preclinical and clinical studies suggests that dysregulated inflammatory response plays a pivotal role in a multitude of chronic ailments including cancer. The molecular mechanism(s) by which chronic inflammation drives cancer initiation and promotion include increased production of pro-inflammatory mediators, such as cytokines, chemokines, reactive oxygen intermediates, increased expression of oncogenes, COX-2 (cyclo-oxygenase-2), 5-LOX (5-lipoxygenase) and MMPs (matrix metalloproteinases), and pro-inflammatory transcription factors such as NF-κB (nuclear factor κB), STAT3 (signal transducer and activator of transcription 3), AP-1 (activator protein 1) and HIF-1α (hypoxia-inducible factor 1α) that mediate tumour cell proliferation, transformation, metastasis, survival, invasion, angiogenesis, chemoresistance and radioresistance. These inflammation-associated molecules are activated by a number of environmental and lifestyle-related factors including infectious agents, tobacco, stress, diet, obesity and alcohol, which together are thought to drive as much as 90% of all cancers. The present review will focus primarily on the role of various inflammatory intermediates responsible for tumour initiation and progression, and discuss in detail the critical link between inflammation and cancer.
- Published
- 2012
28. Celastrol suppresses growth and induces apoptosis of human hepatocellular carcinoma through the modulation of STAT3/JAK2 signaling cascade In Vitro and In Vivo
- Author
-
Rajendran, P., Li, F., Shanmugam, M., Kannaiyan, R., Goh, J., Wong, K., Wang, W., Khin, E., Tergaonkar, V., Kumar, Alan Prem, Luk, J., Sethi, G., Rajendran, P., Li, F., Shanmugam, M., Kannaiyan, R., Goh, J., Wong, K., Wang, W., Khin, E., Tergaonkar, V., Kumar, Alan Prem, Luk, J., and Sethi, G.
- Abstract
Cumulative evidences(s) have established that the constitutive activation of STAT3 plays a pivotal role in the proliferation, survival, metastasis, and angiogenesis and thus can contribute directly to the pathogenesis of hepatocellular carcinoma (HCC). Thus, novel agents that can inhibit STAT3 activation have potential for both prevention and treatment of HCCs. The effect of celastrol on STAT3 activation, associated protein kinases, STAT3-regulated gene products, cellular proliferation, and apoptosis was investigated. The in vivo effect of celastrol on the growth of human HCC xenograft tumors in athymic nu/nu mice was also examined. We observed that celastrol inhibited both constitutive and inducible STAT3 activation, and the suppression was mediated through the inhibition of activation of upstream kinases c-Src, as well as Janus-activated kinase-1 and -2. Vanadate treatment reversed the celastrol-induced modulation of STAT3, suggesting the involvement of a tyrosine phosphatase. The inhibition of STAT3 activation by celastrol led to the suppression of various gene products involved in proliferation, survival, and angiogenesis. Celastrol also inhibited the proliferation and induced apoptosis in HCC cells. Finally, when administered intraperitoneally, celastrol inhibited STAT3 activation in tumor tissues and the growth of human HCC xenograft tumors in athymic nu/nu mice without any side effects. Overall, our results suggest for the first time that celastrol exerts its antiproliferative and proapoptotic effects through suppression of STAT3 signaling in HCC both in vitro and in vivo.
- Published
- 2012
29. The Control of Cell Energy Metabolism by NF-Kb Transcription Factors
- Author
-
Mauro, C., primary, Leow, S. C., additional, Anso, E., additional, Rocha, S., additional, Thotakura, A. K., additional, Tornatore, L., additional, Moretti, M., additional, De Smaele, E., additional, Beg, A. A., additional, Tergaonkar, V., additional, Chandel, N. S., additional, and Franzoso, G., additional
- Published
- 2012
- Full Text
- View/download PDF
30. Aberrant activation of the GIMAPenhancer by oncogenic transcription factors in T-cell acute lymphoblastic leukemia
- Author
-
Liau, W S, Tan, S H, Ngoc, P C T, Wang, C Q, Tergaonkar, V, Feng, H, Gong, Z, Osato, M, Look, A T, and Sanda, T
- Abstract
The transcription factor TAL1/SCLis one of the most prevalent oncogenes in T-cell acute lymphoblastic leukemia (T-ALL), a malignant disorder resulting from leukemic transformation of thymus T-cell precursors. TAL1 is normally expressed in hematopoietic stem cells (HSCs) but is silenced in immature thymocytes. We hypothesize that TAL1 contributes to leukemogenesis by activating genes that are normally repressed in immature thymocytes. Herein, we identified a novel TAL1-regulated super-enhancer controlling the GIMAPlocus, which resides within an insulated chromosomal locus in T-ALL cells. The GIMAPgenes are expressed in HSCs and mature T cells but are downregulated during the immature stage of thymocyte differentiation. The GIMAPenhancer is activated by TAL1, RUNX1 and GATA3 in human T-ALL cells but is repressed by E-proteins. Overexpression of human GIMAPgenes in immature thymocytes alone does not induce tumorigenesis but accelerates leukemia development in zebrafish. Our results demonstrate that aberrant activation of the GIMAPenhancer contributes to T-cell leukemogenesis.
- Published
- 2017
- Full Text
- View/download PDF
31. R-Roscovitine simultaneously targets both the p53 and NF-κB pathways and causes potentiation of apoptosis: implications in cancer therapy
- Author
-
Dey, A, primary, Wong, E T, additional, Cheok, C F, additional, Tergaonkar, V, additional, and Lane, D P, additional
- Published
- 2007
- Full Text
- View/download PDF
32. Nutlin-3 inhibits the NFκB Pathway in a p53 Dependent Manner: Implications in Lung Cancer Therapy
- Author
-
Dey, Anwesha, primary, Wong, E.T., additional, Bist, P., additional, Tergaonkar, V., additional, and Lane, David, additional
- Published
- 2007
- Full Text
- View/download PDF
33. Pleiotropic effects of human papillomavirus type 16 E6 oncogene expression in human epithelial cell lines.
- Author
-
Mythily, D V, primary, Krishna, S, additional, and Tergaonkar, V, additional
- Published
- 1999
- Full Text
- View/download PDF
34. Cytokeratin patterns of expression in human epithelial cell lines correlate with transcriptional activity of the human papillomavirus type 16 upstream regulatory region.
- Author
-
Tergaonkar, V, primary, Mythily, D V, additional, and Krishna, S, additional
- Published
- 1997
- Full Text
- View/download PDF
35. R-Roscovitine simultaneously targets both the p53 and NF-κB pathways and causes potentiation of apoptosis: implications in cancer therapy.
- Author
-
Dey, A., Wong, E. T., Cheok, C. F., Tergaonkar, V., and Lane, D. P.
- Subjects
PURINES ,ANTINEOPLASTIC agents ,CANCER treatment ,APOPTOSIS ,NF-kappa B - Abstract
Seliciclib (CYC202, R-Roscovitine) is a 2, 6, 9-substituted purine analog that is currently in phase II clinical trials as an anticancer agent. We show in this study that R-Roscovitine can downregulate nuclear factor-kappa B (NF-κB) activation in response to tumor necrosis factor (TNF)α and interleukin 1. Activation of p53-dependent transcription is not compromised when R-Roscovitine is combined with TNFα. We characterize the molecular mechanism governing NF-κB repression and show that R-Roscovitine inhibits the IκB kinase (IKK) kinase activity, which leads to defective IκBα phosphorylation, degradation and hence nuclear function of NF-κB. We further show that the downregulation of the NF-κB pathway is also at the level of p65 modification and that the phosphorylation of p65 at Ser 536 is repressed by R-Roscovitine. Consistent with repression of canonical IKK signaling pathway, the induction of NF-κB target genes monocyte chemoattractant protein, intercellular adhesion molecule-1, cyclooxygenase-2 and IL-8 is also inhibited by R-Roscovitine. We further show that treatment of cells with TNFα and R-Roscovitine causes potentiation of cell death. Based on these results, we suggest the potential use of R-Roscovitine as a bitargeted anticancer drug that functions by simultaneously causing p53 activation and NF-κB suppression. This study also provides mechanistic insight into the molecular mechanism of action of R-Roscovitine, thereby possibly explaining its anti-inflammatory properties.Cell Death and Differentiation (2008) 15, 263–273; doi:10.1038/sj.cdd.4402257; published online 2 November 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
36. Human papillomavirus type 16 E6-enhanced susceptibility of L929 cells to tumor necrosis factor alpha correlates with increased accumulation of reactive oxygen species.
- Author
-
Liu, Y, Tergaonkar, V, Krishna, S, and Androphy, E J
- Abstract
Human papillomavirus type 16 (HPV-16) E6 has been shown to prevent or enhance apoptosis depending on the stimulus and cell type. Here we present evidence that HPV-16 E6 sensitized murine fibrosarcoma L929 cells to tumor necrosis factor alpha (TNF)-induced cytolysis. The E6-enhanced cytolysis correlated with a precedent increase in reactive oxygen species (ROS) level and antioxidant treatment could completely block the E6-dependent sensitization. These findings represent the first demonstration of a link between a viral oncogene-sensitized cytolysis and ROS. Previous studies have shown conflicting results regarding whether TNF-induced cytolysis of L929 cells is through necrosis or apoptosis. Here we report that, although L929 cells underwent DNA fragmentation after exposure to TNF, they retained the morphology of intact nuclei while gaining permeability to propidium iodide, features characteristic of necrosis rather than apoptosis. We confirmed that the broad spectrum caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone markedly increased the susceptibility of L929 cells to TNF, and further demonstrated that E6 enhanced this susceptibility, which again correlated with increased ROS accumulation. We showed that the expression of E6 in L929 cells did not alter the stability of p53, and the cells retained a p53 response to actinomycin D. Furthermore, two E6 mutants defective for p53 degradation in other systems exhibited differential effects on TNF sensitization. These results suggest that the enhancement of TNF-induced L929 cytolysis by E6 is independent of p53 degradation. We also found that TNF-induced activation of NF-kappaB did not account for the enhanced TNF susceptibility by E6.
- Published
- 1999
37. γ-tocotrienol inhibits angiogenesis-dependent growth of human hepatocellular carcinoma through abrogation of AKT/mTOR pathway in an orthotopic mouse model
- Author
-
Kodappully Sivaraman Siveen, Ahn, K. S., Ong, T. H., Shanmugam, M. K., Li, F., Yap, W. N., Kumar, A. P., Fong, C. W., Tergaonkar, V., Hui, K. M., and Sethi, G.
38. Aberrant Activation of the GIMAP Enhancer by Oncogenic Transcription Factors in T-cell Acute Lymphoblastic Leukemia
- Author
-
Phuong Cao Thi Ngoc, Chelsia Qiuxia Wang, Hui Feng, Vinay Tergaonkar, Wei-Siang Liau, Takaomi Sanda, Shi Hao Tan, Zhiyuan Gong, A T Look, Motomi Osato, Liau, WS, Tan, SH, Ngoc, PCT, Wang, CQ, Tergaonkar, V, Feng, H, Gong, Z, Osato, Motomi, Look, AT, and Sanda, T
- Subjects
0301 basic medicine ,Cancer Research ,T cell ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,GTP-Binding Proteins ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,leukemia development ,Animals ,Humans ,Enhancer ,Transcription factor ,Zebrafish ,Membrane Proteins ,GIMAP enhancer ,Hematology ,medicine.disease ,Mice, Inbred C57BL ,Haematopoiesis ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Oncology ,RUNX1 ,chemistry ,030220 oncology & carcinogenesis ,Multigene Family ,Cancer research ,T-cell acute lymphoblastic leukemia ,TAL1 ,Transcription Factors - Abstract
The transcription factor TAL1/SCL is one of the most prevalent oncogenes in T-cell acute lymphoblastic leukemia (T-ALL), a malignant disorder resulting from leukemic transformation of thymus T-cell precursors. TAL1 is normally expressed in hematopoietic stem cells (HSCs) but is silenced in immature thymocytes. We hypothesize that TAL1 contributes to leukemogenesis by activating genes that are normally repressed in immature thymocytes. Herein, we identified a novel TAL1-regulated super-enhancer controlling the GIMAP locus, which resides within an insulated chromosomal locus in T-ALL cells. The GIMAP genes are expressed in HSCs and mature T cells but are downregulated during the immature stage of thymocyte differentiation. The GIMAP enhancer is activated by TAL1, RUNX1 and GATA3 in human T-ALL cells but is repressed by E-proteins. Overexpression of human GIMAP genes in immature thymocytes alone does not induce tumorigenesis but accelerates leukemia development in zebrafish. Our results demonstrate that aberrant activation of the GIMAP enhancer contributes to T-cell leukemogenesis. Refereed/Peer-reviewed
- Published
- 2016
39. Integrin-α9 overexpression underlies the niche-independent maintenance of leukemia stem cells in acute myeloid leukemia.
- Author
-
Niibori-Nambu A, Wang CQ, Chin DWL, Chooi JY, Hosoi H, Sonoki T, Tham CY, Nah GSS, Cirovic B, Tan DQ, Takizawa H, Sashida G, Goh Y, Tng J, Fam WN, Fullwood MJ, Suda T, Yang H, Tergaonkar V, Taniuchi I, Li S, Chng WJ, and Osato M
- Subjects
- Animals, Humans, Mice, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor Alpha 2 Subunit metabolism, Gene Expression Regulation, Leukemic, Integrin alpha Chains metabolism, Integrin alpha Chains genetics, Mice, Inbred C57BL, Osteopontin genetics, Osteopontin metabolism, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins c-myc genetics, Signal Transduction, Stem Cell Niche, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology
- Abstract
Leukemia stem cells (LSCs) are widely believed to reside in well-characterized bone marrow (BM) niches; however, the capacity of the BM niches to accommodate LSCs is insufficient, and a significant proportion of LSCs are instead maintained in regions outside the BM. The molecular basis for this niche-independent behavior of LSCs remains elusive. Here, we show that integrin-α9 overexpression (ITGA9 OE) plays a pivotal role in the extramedullary maintenance of LSCs by molecularly mimicking the niche-interacting status, through the binding with its soluble ligand, osteopontin (OPN). Retroviral insertional mutagenesis conducted on leukemia-prone Runx-deficient mice identified Itga9 OE as a novel leukemogenic event. Itga9 OE activates Akt and p38MAPK signaling pathways. The elevated Myc expression subsequently enhances ribosomal biogenesis to overcome the cell integrity defect caused by the preexisting Runx alteration. The Itga9-Myc axis, originally discovered in mice, was further confirmed in multiple human acute myeloid leukemia (AML) subtypes, other than RUNX leukemias. In addition, ITGA9 was shown to be a functional LSC marker of the best prognostic value among 14 known LSC markers tested. Notably, the binding of ITGA9 with soluble OPN, a known negative regulator against HSC activation, induced LSC dormancy, while the disruption of ITGA9-soluble OPN interaction caused rapid cell propagation. These findings suggest that the ITGA9 OE increases both actively proliferating leukemia cells and dormant LSCs in a well-balanced manner, thereby maintaining LSCs. The ITGA9 OE would serve as a novel therapeutic target in AML., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
40. Iron-(Fe3+)-Dependent Reactivation of Telomerase Drives Colorectal Cancers.
- Author
-
Shanmugam R, Majee P, Shi W, Ozturk MB, Vaiyapuri TS, Idzham K, Raju A, Shin SH, Fidan K, Low JL, Chua JYH, Kong YC, Qi OY, Tan E, Chok AY, Seow-En I, Wee I, Macalinao DC, Chong DQ, Chang HY, Lee F, Leow WQ, Murata-Hori M, Xiaoqian Z, Shumei C, Tan CSH, Dasgupta R, Tan IB, and Tergaonkar V
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Telomerase metabolism, Telomerase genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Iron metabolism
- Abstract
Over-consumption of iron-rich red meat and hereditary or genetic iron overload are associated with an increased risk of colorectal carcinogenesis, yet the mechanistic basis of how metal-mediated signaling leads to oncogenesis remains enigmatic. Using fresh colorectal cancer samples we identify Pirin, an iron sensor, that overcomes a rate-limiting step in oncogenesis, by reactivating the dormant human telomerase reverse transcriptase (hTERT) subunit of the telomerase holoenzyme in an iron-(Fe3+)-dependent manner and thereby drives colorectal cancers. Chemical genetic screens combined with isothermal dose-response fingerprinting and mass spectrometry identified a small molecule SP2509 that specifically inhibits Pirin-mediated hTERT reactivation in colorectal cancers by competing with iron-(Fe3+) binding. Our findings, first to document how metal ions reactivate telomerase, provide a molecular mechanism for the well-known association between red meat and increased incidence of colorectal cancers. Small molecules like SP2509 represent a novel modality to target telomerase that acts as a driver of 90% of human cancers and is yet to be targeted in clinic. Significance: We show how iron-(Fe3+) in collusion with genetic factors reactivates telomerase, providing a molecular mechanism for the association between iron overload and increased incidence of colorectal cancers. Although no enzymatic inhibitors of telomerase have entered the clinic, we identify SP2509, a small molecule that targets telomerase reactivation and function in colorectal cancers., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
41. Super-silencer perturbation by EZH2 and REST inhibition leads to large loss of chromatin interactions and reduction in cancer growth.
- Author
-
Zhang Y, Chen K, Tang SC, Cai Y, Nambu A, See YX, Fu C, Raju A, Lebeau B, Ling Z, Chan JJ, Tay Y, Mutwil M, Lakshmanan M, Tucker-Kellogg G, Chng WJ, Tenen DG, Osato M, Tergaonkar V, and Fullwood MJ
- Abstract
Human silencers have been shown to regulate developmental gene expression. However, the functional importance of human silencers needs to be elucidated, such as whether they can form 'super-silencers' and whether they are linked to cancer progression. Here, we show two silencer components of the FGF18 gene can cooperate through compensatory chromatin interactions to form a super-silencer. Double knockout of two silencers exhibited synergistic upregulation of FGF18 expression and changes in cell identity. To perturb the super-silencers, we applied combinational treatment of an enhancer of zeste homolog 2 inhibitor GSK343, and a repressor element 1-silencing transcription factor inhibitor, X5050 ('GR'). Interestingly, GR led to severe loss of topologically associated domains and loops, which were associated with reduced CTCF and TOP2A mRNA levels. Moreover, GR synergistically upregulated super-silencer-controlled genes related to cell cycle, apoptosis and DNA damage, leading to anticancer effects in vivo. Overall, our data demonstrated a super-silencer example and showed that GR can disrupt super-silencers, potentially leading to cancer ablation., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
42. Autophagy in aging-related diseases and cancer: Principles, regulatory mechanisms and therapeutic potential.
- Author
-
Wu N, Zheng W, Zhou Y, Tian Y, Tang M, Feng X, Ashrafizadeh M, Wang Y, Niu X, Tambuwala M, Wang L, Tergaonkar V, Sethi G, Klionsky D, Huang L, and Gu M
- Subjects
- Humans, Animals, Autophagy physiology, Neoplasms therapy, Neoplasms pathology, Neoplasms metabolism, Aging physiology, Aging pathology, Aging metabolism
- Abstract
Macroautophagy/autophagy is primarily accountable for the degradation of damaged organelles and toxic macromolecules in the cells. Regarding the essential function of autophagy for preserving cellular homeostasis, changes in, or dysfunction of, autophagy flux can lead to disease development. In the current paper, the complicated function of autophagy in aging-associated pathologies and cancer is evaluated, highlighting the underlying molecular mechanisms that can affect longevity and disease pathogenesis. As a natural biological process, a reduction in autophagy is observed with aging, resulting in an accumulation of cell damage and the development of different diseases, including neurological disorders, cardiovascular diseases, and cancer. The MTOR, AMPK, and ATG proteins demonstrate changes during aging, and they are promising therapeutic targets. Insulin/IGF1, TOR, PKA, AKT/PKB, caloric restriction and mitochondrial respiration are vital for lifespan regulation and can modulate or have an interaction with autophagy. The specific types of autophagy, such as mitophagy that degrades mitochondria, can regulate aging by affecting these organelles and eliminating those mitochondria with genomic mutations. Autophagy and its specific types contribute to the regulation of carcinogenesis and they are able to dually enhance or decrease cancer progression. Cancer hallmarks, including proliferation, metastasis, therapy resistance and immune reactions, are tightly regulated by autophagy, supporting the conclusion that autophagy is a promising target in cancer therapy., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
43. Mast cells and the gut-liver Axis: Implications for liver disease progression and therapy.
- Author
-
Nair B, Kamath AJ, Tergaonkar V, Sethi G, and Nath LR
- Subjects
- Humans, Animals, Non-alcoholic Fatty Liver Disease metabolism, Non-alcoholic Fatty Liver Disease therapy, Non-alcoholic Fatty Liver Disease pathology, Mast Cells metabolism, Gastrointestinal Microbiome, Liver Diseases pathology, Disease Progression, Liver pathology, Liver metabolism
- Abstract
The role of mast cells, traditionally recognized for their involvement in immediate hypersensitivity reactions, has garnered significant attention in liver diseases. Studies have indicated a notable increase in mast cell counts following hepatic injury, underscoring their potential contribution to liver disorder pathogenesis. Predominantly situated in connective tissue that envelops the hepatic veins, bile ducts, and arteries, mast cells are central to both initiating and perpetuating liver disorders. Additionally, they are crucial for maintaining gastrointestinal barrier function. The gut-liver axis emphasizes the complex, two-way communication between the gut microbiome and the liver. Past research has implicated gut microbiota and their metabolites in the progression of hepatic disorders. This review sheds light on how mast cells are activated in various liver conditions such as alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), viral hepatitis, hepatic fibrogenesis, and hepatocellular carcinoma. It also briefly explores the connection between the gut microbiome and mast cell activation in these hepatic conditions., Competing Interests: Declaration of competing interest The authors declare no conflict of interest., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
44. Versatile function of NF-ĸB in inflammation and cancer.
- Author
-
Ma Q, Hao S, Hong W, Tergaonkar V, Sethi G, Tian Y, and Duan C
- Abstract
Nuclear factor-kappaB (NF-ĸB) plays a crucial role in both innate and adaptive immune systems, significantly influencing various physiological processes such as cell proliferation, migration, differentiation, survival, and stemness. The function of NF-ĸB in cancer progression and response to chemotherapy has gained increasing attention. This review highlights the role of NF-ĸB in inflammation control, biological mechanisms, and therapeutic implications in cancer treatment. NF-ĸB is instrumental in altering the release of inflammatory factors such as TNF-α, IL-6, and IL-1β, which are key in the regulation of carcinogenesis. Specifically, in conditions including colitis, NF-ĸB upregulation can intensify inflammation, potentially leading to the development of colorectal cancer. Its pivotal role extends to regulating the tumor microenvironment, impacting components such as macrophages, fibroblasts, T cells, and natural killer cells. This regulation influences tumorigenesis and can dampen anti-tumor immune responses. Additionally, NF-ĸB modulates cell death mechanisms, notably by inhibiting apoptosis and ferroptosis. It also has a dual role in stimulating or suppressing autophagy in various cancers. Beyond these functions, NF-ĸB plays a role in controlling cancer stem cells, fostering angiogenesis, increasing metastatic potential through EMT induction, and reducing tumor cell sensitivity to chemotherapy and radiotherapy. Given its oncogenic capabilities, research has focused on natural products and small molecule compounds that can suppress NF-ĸB, offering promising avenues for cancer therapy., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
45. PHF2 regulates genome topology and DNA replication in neural stem cells via cohesin.
- Author
-
Feng J, Chuah YH, Liang Y, Cipta NO, Zeng Y, Warrier T, Elfar GARE, Yoon J, Grinchuk OV, Tay EXY, Lok KZ, Zheng ZQ, Khong ZJ, Chong ZS, Teo J, Sanford EM, Neo CJY, Chiu HY, Leung JY, Wang LC, Lim YT, Zhao T, Sobota RM, Crasta KC, Tergaonkar V, Taneja R, Ng SY, Cheok CF, Ling SC, Loh YH, and Ong DST
- Subjects
- Animals, Mice, Chromatin metabolism, Replication Origin, Histone Demethylases metabolism, Histone Demethylases genetics, Nuclear Proteins metabolism, Nuclear Proteins genetics, Genome genetics, CCCTC-Binding Factor metabolism, CCCTC-Binding Factor genetics, Mice, Knockout, Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, Cohesins, Chromosomal Proteins, Non-Histone metabolism, Chromosomal Proteins, Non-Histone genetics, Neural Stem Cells metabolism, Neural Stem Cells cytology, DNA Replication, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics
- Abstract
Cohesin plays a crucial role in the organization of topologically-associated domains (TADs), which influence gene expression and DNA replication timing. Whether epigenetic regulators may affect TADs via cohesin to mediate DNA replication remains elusive. Here, we discover that the histone demethylase PHF2 associates with RAD21, a core subunit of cohesin, to regulate DNA replication in mouse neural stem cells (NSC). PHF2 loss impairs DNA replication due to the activation of dormant replication origins in NSC. Notably, the PHF2/RAD21 co-bound genomic regions are characterized by CTCF enrichment and epigenomic features that resemble efficient, active replication origins, and can act as boundaries to separate adjacent domains. Accordingly, PHF2 loss weakens TADs and chromatin loops at the co-bound loci due to reduced RAD21 occupancy. The observed topological and DNA replication defects in PHF2 KO NSC support a cohesin-dependent mechanism. Furthermore, we demonstrate that the PHF2/RAD21 complex exerts little effect on gene regulation, and that PHF2's histone-demethylase activity is dispensable for normal DNA replication and proliferation of NSC. We propose that PHF2 may serve as a topological accessory to cohesin for cohesin localization to TADs and chromatin loops, where cohesin represses dormant replication origins directly or indirectly, to sustain DNA replication in NSC., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
46. Tumour microenvironment programming by an RNA-RNA-binding protein complex creates a druggable vulnerability in IDH-wild-type glioblastoma.
- Author
-
Wu L, Zhao Z, Shin YJ, Yin Y, Raju A, Vaiyapuri TS, Idzham K, Son M, Lee Y, Sa JK, Chua JYH, Unal B, Zhai Y, Fan W, Huang L, Hu H, Gunaratne J, Nam DH, Jiang T, and Tergaonkar V
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Mutation, Antineoplastic Agents pharmacology, Xenograft Model Antitumor Assays, Cell Proliferation, Gene Expression Regulation, Neoplastic, Glioblastoma pathology, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma drug therapy, Tumor Microenvironment, Brain Neoplasms pathology, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms drug therapy, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase metabolism, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics
- Abstract
Patients with IDH-wild-type glioblastomas have a poor five-year survival rate along with limited treatment efficacy due to immune cell (glioma-associated microglia and macrophages) infiltration promoting tumour growth and resistance. To enhance therapeutic options, our study investigated the unique RNA-RNA-binding protein complex LOC-DHX15. This complex plays a crucial role in driving immune cell infiltration and tumour growth by establishing a feedback loop between cancer and immune cells, intensifying cancer aggressiveness. Targeting this complex with blood-brain barrier-permeable small molecules improved treatment efficacy, disrupting cell communication and impeding cancer cell survival and stem-like properties. Focusing on RNA-RNA-binding protein interactions emerges as a promising approach not only for glioblastomas without the IDH mutation but also for potential applications beyond cancer, offering new avenues for developing therapies that address intricate cellular relationships in the body., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
47. Pushing the Frontiers of Cancer Research: Highlights from the Frontiers in Cancer Science Conference 2023.
- Author
-
Lee YF, Chen L, Chew V, Chow EK, Deng LW, Hunziker W, Lee ASG, Leong G, Ngeow J, Pervaiz S, Sabapathy K, Skanderup AJ, Sundar R, Tay Y, Virshup DM, Wong SH, Tergaonkar V, and Tam WL
- Subjects
- Humans, Cell Death, Drug Delivery Systems, Research, Neoplasms therapy
- Abstract
The 15th annual Frontiers in Cancer Science (FCS) conference gathered scientific experts who shared the latest research converging upon several themes of cancer biology. These themes included the dysregulation of metabolism, cell death, and other signaling processes in cancer cells; using patient "omics" datasets and single-cell and spatial approaches to investigate heterogeneity, understand therapy resistance, and identify targets; innovative strategies for inhibiting tumors, including rational drug combinations and improved drug delivery mechanisms; and advances in models that can facilitate screening for cancer vulnerabilities and drug testing. We hope the insights from this meeting will stimulate further progress in the field., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
48. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy.
- Author
-
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, and Han P
- Subjects
- Humans, Tumor Microenvironment, Immunotherapy, Cell Differentiation, Nanoparticles therapeutic use, Neoplasms therapy
- Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
49. Cellular heterogeneity in TNF/TNFR1 signalling: live cell imaging of cell fate decisions in single cells.
- Author
-
Preedy MK, White MRH, and Tergaonkar V
- Subjects
- Signal Transduction, NF-kappa B metabolism, Apoptosis, Receptors, Tumor Necrosis Factor, Type I metabolism, Tumor Necrosis Factor-alpha pharmacology, Tumor Necrosis Factor-alpha metabolism
- Abstract
Cellular responses to TNF are inherently heterogeneous within an isogenic cell population and across different cell types. TNF promotes cell survival by activating pro-inflammatory NF-κB and MAPK signalling pathways but may also trigger apoptosis and necroptosis. Following TNF stimulation, the fate of individual cells is governed by the balance of pro-survival and pro-apoptotic signalling pathways. To elucidate the molecular mechanisms driving heterogenous responses to TNF, quantifying TNF/TNFR1 signalling at the single-cell level is crucial. Fluorescence live-cell imaging techniques offer real-time, dynamic insights into molecular processes in single cells, allowing for detection of rapid and transient changes, as well as identification of subpopulations, that are likely to be missed with traditional endpoint assays. Whilst fluorescence live-cell imaging has been employed extensively to investigate TNF-induced inflammation and TNF-induced cell death, it has been underutilised in studying the role of TNF/TNFR1 signalling pathway crosstalk in guiding cell-fate decisions in single cells. Here, we outline the various opportunities for pathway crosstalk during TNF/TNFR1 signalling and how these interactions may govern heterogenous responses to TNF. We also advocate for the use of live-cell imaging techniques to elucidate the molecular processes driving cell-to-cell variability in single cells. Understanding and overcoming cellular heterogeneity in response to TNF and modulators of the TNF/TNFR1 signalling pathway could lead to the development of targeted therapies for various diseases associated with aberrant TNF/TNFR1 signalling, such as rheumatoid arthritis, metabolic syndrome, and cancer., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
50. BRD4 isoforms have distinct roles in tumour progression and metastasis in rhabdomyosarcoma.
- Author
-
Das D, Leung JY, Balamurugan S, Tergaonkar V, Loh AHP, Chiang CM, and Taneja R
- Subjects
- Humans, Nuclear Proteins genetics, Nuclear Proteins metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, Protein Isoforms genetics, Bromodomain Containing Proteins, Transcription Factors genetics, Transcription Factors metabolism, Rhabdomyosarcoma genetics
- Abstract
BRD4, a bromodomain and extraterminal (BET) protein, is deregulated in multiple cancers and has emerged as a promising drug target. However, the function of the two main BRD4 isoforms (BRD4-L and BRD4-S) has not been analysed in parallel in most cancers. This complicates determining therapeutic efficacy of pan-BET inhibitors. In this study, using functional and transcriptomic analysis, we show that BRD-L and BRD4-S isoforms play distinct roles in fusion negative embryonal rhabdomyosarcoma. BRD4-L has an oncogenic role and inhibits myogenic differentiation, at least in part, by activating myostatin expression. Depletion of BRD4-L in vivo impairs tumour progression but does not impact metastasis. On the other hand, depletion of BRD4-S has no significant impact on tumour growth, but strikingly promotes metastasis in vivo. Interestingly, BRD4-S loss results in the enrichment of BRD4-L and RNA Polymerase II at integrin gene promoters resulting in their activation. In fusion positive alveolar rhabdomyosarcoma, BRD4-L is unrestricted in its oncogenic role, with no evident involvement of BRD4-S. Our work unveils isoform-specific functions of BRD4 in rhabdomyosarcoma., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.