137 results on '"Burma, S"'
Search Results
2. DNA double-strand breaks cooperate with loss of Ink4 and Arf tumor suppressors to generate glioblastomas with frequent Met amplification
- Author
-
Camacho, C V, Todorova, P K, Hardebeck, M C, Tomimatsu, N, Gil del Alcazar, C R, Ilcheva, M, Mukherjee, B, McEllin, B, Vemireddy, V, Hatanpaa, K, Story, M D, Habib, A A, Murty, V V, Bachoo, R, and Burma, S
- Published
- 2015
- Full Text
- View/download PDF
3. EGFR wild type antagonizes EGFRvIII-mediated activation of Met in glioblastoma
- Author
-
Li, L, Puliyappadamba, V T, Chakraborty, S, Rehman, A, Vemireddy, V, Saha, D, Souza, R F, Hatanpaa, K J, Koduru, P, Burma, S, Boothman, D A, and Habib, A A
- Published
- 2015
- Full Text
- View/download PDF
4. An EGFR wild type–EGFRvIII–HB-EGF feed-forward loop regulates the activation of EGFRvIII
- Author
-
Li, L, Chakraborty, S, Yang, C-R, Hatanpaa, K J, Cipher, D J, Puliyappadamba, V T, Rehman, A, Jiwani, A J, Mickey, B, Madden, C, Raisanen, J, Burma, S, Saha, D, Wang, Z, Pingle, S C, Kesari, S, Boothman, D A, and Habib, A A
- Published
- 2014
- Full Text
- View/download PDF
5. SP-0352 Targeting radiation-induced senescence to augment glioblastoma therapy
- Author
-
Burma, S.
- Published
- 2023
- Full Text
- View/download PDF
6. Baculovirus vector-mediated expression of heterologous genes in insect cells
- Author
-
Sridhar, P., Awasthi, A. K., Azim, A. A., Burma, S., Habib, S., Jain, A., Mukherjee, B., Ranjan, A., and Hasnain, Seyed E.
- Published
- 1994
- Full Text
- View/download PDF
7. A Six Year Trend in Treatment Outcomes among Tuberculosis Patients and its Determinants in Andaman and Nicobar Islands.
- Author
-
RAO, SHIVANI, JAHNAVI, G., BURMA, S. P., and THATKAR, PANDURANG V.
- Subjects
TUBERCULOSIS patients ,TREATMENT effectiveness ,DIRECTLY observed therapy ,ISLANDS - Abstract
Introduction: Directly Observed Treatment Short (DOTS) course strategy is aimed at diagnosing 70% of infectious Tuberculosis (TB) and curing 85% of it. Aim: To analyse the trend in TB treatment outcomes in the newly diagnosed patients from 2010 to 2015 in Andaman and Nicobar Islands. Materials and Methods: A retrospective cohort study was conducted among TB patients registered from 2010 to 2015 at DOTS TB State Center. Patient particulars like age, sex, TB category at the beginning, and treatment outcomes were collected from the records that were available with DOTS. Results: Incidence of TB in Andaman and Nicobar islands was about 171 per 1,00,000 population during 2010 to 2015, a total of 3917 cases were registered. The total annual new case detection rate increased from 72% to 78%, treatment success rate increased from 84.9% to 89.3%, default rate decreased from 4.1% to 1.4%. Conclusion: The present study analyses the situation of newly diagnosed TB patients in Andaman and Nicobar Islands from 2010 to 2015. Over the years there was a significant increase in the cure rate and a significant decrease in failure and defaulter rates. Over the years there was a significant transition of the treatment outcomes in these islands. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
8. DNA double-strand breaks cooperate with loss of Ink4 and Arf tumor suppressors to generate glioblastomas with frequent Met amplification
- Author
-
Camacho, C V, primary, Todorova, P K, additional, Hardebeck, M C, additional, Tomimatsu, N, additional, Gil del Alcazar, C R, additional, Ilcheva, M, additional, Mukherjee, B, additional, McEllin, B, additional, Vemireddy, V, additional, Hatanpaa, K, additional, Story, M D, additional, Habib, A A, additional, Murty, V V, additional, Bachoo, R, additional, and Burma, S, additional
- Published
- 2014
- Full Text
- View/download PDF
9. EGFR wild type antagonizes EGFRvIII-mediated activation of Met in glioblastoma
- Author
-
Li, L, primary, Puliyappadamba, V T, additional, Chakraborty, S, additional, Rehman, A, additional, Vemireddy, V, additional, Saha, D, additional, Souza, R F, additional, Hatanpaa, K J, additional, Koduru, P, additional, Burma, S, additional, Boothman, D A, additional, and Habib, A A, additional
- Published
- 2013
- Full Text
- View/download PDF
10. An EGFR wild type–EGFRvIII–HB-EGF feed-forward loop regulates the activation of EGFRvIII
- Author
-
Li, L, primary, Chakraborty, S, additional, Yang, C-R, additional, Hatanpaa, K J, additional, Cipher, D J, additional, Puliyappadamba, V T, additional, Rehman, A, additional, Jiwani, A J, additional, Mickey, B, additional, Madden, C, additional, Raisanen, J, additional, Burma, S, additional, Saha, D, additional, Wang, Z, additional, Pingle, S C, additional, Kesari, S, additional, Boothman, D A, additional, and Habib, A A, additional
- Published
- 2013
- Full Text
- View/download PDF
11. AZD5438, an Inhibitor of CDK 1, 2, and 9, Enhances the Radiosensitivity of Non-small Cell Lung Carcinoma Cells
- Author
-
Tumati, V., primary, Raghavan, P., additional, Yu, L., additional, Chan, N., additional, Tomimatsu, N., additional, Burma, S., additional, Bristow, R.G., additional, and Saha, D., additional
- Published
- 2012
- Full Text
- View/download PDF
12. RADIOBIOLOGY
- Author
-
Gu, C., primary, Demir, H., additional, Joshi, K., additional, Nakamura, Y., additional, Yamada, R., additional, Gupta, S., additional, Kwon, C.-H., additional, Chaudhury, A. R., additional, Nakano, I., additional, Yong, R., additional, Yang, C., additional, Lonser, R., additional, Zhuang, Z., additional, Gwak, H.-S., additional, Jo, G. H., additional, Bogler, O., additional, Chwae, Y.-J., additional, Yoo, H., additional, Lee, S. H., additional, Park, J. B., additional, Burrell, K., additional, Jelveh, S., additional, Hill, R., additional, Zadeh, G., additional, Mcellin, B., additional, Sirasanagandla, S., additional, Mashimo, T., additional, Nannepaja, S., additional, Vemireddy, V., additional, Burma, S., additional, and Bachoo, R., additional
- Published
- 2011
- Full Text
- View/download PDF
13. DNA Double-Strand Break Repair: Implications for Glioblastoma Therapy
- Author
-
Amancherla, K., primary, Mukherjee, B., additional, McEllin, B., additional, Pichamoorthy, N., additional, Camacho, C., additional, Tomimatsu, N., additional, Bachoo, R., additional, and Burma, S., additional
- Published
- 2011
- Full Text
- View/download PDF
14. Modulation of the DNA-damage response to HZE particles by shielding
- Author
-
MUKHERJEE, B, primary, CAMACHO, C, additional, TOMIMATSU, N, additional, MILLER, J, additional, and BURMA, S, additional
- Published
- 2008
- Full Text
- View/download PDF
15. Nucleophosmin suppresses oncogene-induced apoptosis and senescence and enhances oncogenic cooperation in cells with genomic instability
- Author
-
Li, J., primary, Sejas, D. P., additional, Burma, S., additional, Chen, D. J., additional, and Pang, Q., additional
- Published
- 2007
- Full Text
- View/download PDF
16. An unusual 30-kDa protein binding to the polyhedrin gene promoter of Autographa californica nuclear polyhedrosis virus.
- Author
-
Burma, S., primary, Mukherjee, B., additional, Jain, A., additional, Habib, S., additional, and Hasnain, S.E., additional
- Published
- 1994
- Full Text
- View/download PDF
17. Synthetic resins. XII. Synthesis and characterization of the resins derived from resacetophenone–substituted benzoic acids–furfural
- Author
-
Senapati, M., primary, Burma, S. P. N. S., additional, and Nayak, P. L., additional
- Published
- 1992
- Full Text
- View/download PDF
18. Synthetic resins. XIII. Synthesis and characterization of the resins derived from quinacetophenone/substituted benzoic acid/furfuraldehyde
- Author
-
Burma, S. P. N. S., primary, Lenka, S., additional, and Nayak, P. L., additional
- Published
- 1992
- Full Text
- View/download PDF
19. DNA-dependent protein kinase-independent activation of p53 in response to DNA damage.
- Author
-
Burma, S, Kurimasa, A, Xie, G, Taya, Y, Araki, R, Abe, M, Crissman, H A, Ouyang, H, Li, G C, and Chen, D J
- Abstract
Phosphorylation at serine 15 of the human p53 tumor suppressor protein is induced by DNA damage and correlates with accumulation of p53 and its activation as a transcription factor. The DNA-dependent protein kinase (DNA-PK) can phosphorylate serine 15 of human p53 and the homologous serine 18 of murine p53 in vitro. Contradictory reports exist about the requirement for DNA-PK in vivo for p53 activation and cell cycle arrest in response to ionizing radiation. While primary SCID (severe combined immunodeficiency) cells, that have defective DNA-PK, show normal p53 activation and cell cycle arrest, a transcriptionally inert form of p53 is induced in the SCID cell line SCGR11. In order to unambiguously define the role of the DNA-PK catalytic subunit (DNA-PKcs) in p53 activation, we examined p53 phosphorylation in mouse embryonic fibroblasts (MEFs) from DNA-PKcs-null mice. We found a similar pattern of serine 18 phosphorylation and accumulation of p53 in response to irradiation in both control and DNA-PKcs-null MEFs. The induced p53 was capable of sequence-specific DNA binding even in the absence of DNA-PKcs. Transactivation of the cyclin-dependent-kinase inhibitor p21, a downstream target of p53, and the G1 cell cycle checkpoint were also found to be normal in the DNA-PKcs -/- MEFs. Our results demonstrate that DNA-PKcs, unlike the related ATM protein, is not essential for the activation of p53 and G1 cell cycle arrest in response to ionizing radiation.
- Published
- 1999
20. The 30-kDa protein binding to the "initiator" of the baculovirus polyhedrin promoter also binds specifically to the coding strand.
- Author
-
Mukherjee, B, Burma, S, and Hasnain, S E
- Abstract
We previously reported the purification and characterization of the polyhedrin promoter-binding protein (PPBP), an unusual DNA-binding protein that interacts with transcriptionally important motifs of the baculovirus polyhedrin gene promoter (S. Burma, B. Mukherjee, A. Jain, S. Habib, and S.E. Hasnain, J. Biol. Chem. (1994) 269, 2750-2757. PPBP also exhibits a sequence-specific single-stranded DNA-binding activity. Gel retardations and competition analyses with double- and single-stranded oligonucleotides indicated that PPBP binds the coding strand and not the noncoding strand of the promoter. This was further confirmed by UV cross-linking and Southwestern blotting experiments. Gel retardations with mutated oligonucleotides indicated that both dsDNA and ssDNA binding involve common AATA-AATAAGTATT motifs. However, ssDNA binding is dependent upon ionic interactions unlike dsDNA binding, which is mainly through nonionic interactions. The affinity of PPBP for the coding strand appears to be higher than that for duplex promoter DNA. Interestingly, the PPBP-coding strand complex has a longer half-life (approximately 60 min) than the PPBP-duplex promoter complex (approximately 15 min). PPBP represents a unique example of an "initiator" promoter-binding protein with dual dsDNA and ssDNA binding activities, and this reconciles very well with the unusual binding characteristics displayed by it. The formation of the PPBP-coding strand complex in vivo may be a crucial step for the exceptionally high and repeated rounds of transcriptional activity of the baculovirus polyhedrin gene promoter.
- Published
- 1995
21. Prevalence of female genital tuberculosis, its risk factors and associated clinical features among the women of Andaman Islands, India: a community-based study.
- Author
-
Parvez, R., Sugunan, A. P., Vijayachari, P., Burma, S. P., Mandal, A., Saha, M. K., and Shah, W. A.
- Abstract
Objective: There is scarcity of information on the prevalence of female genital tuberculosis (FGTB) in the community. The present study was carried out to estimate the prevalence of FGTB, its risk factors and associated clinical features. Study design: Community-based cross-sectional survey. Methods: This study was carried during October 2011 and May 2014 in the Andaman Islands. A total of 13,300 women aged 20-59 years were primarily screened using a structured questionnaire. About 721 (5.4%) were found initially eligible for screening for genital tuberculosis by clinical examination and specimen collection for laboratory tests but only 460 (63.8%) expressed their willingness. Endometrial specimens were collected from 405 (88%) subjects. The association of the potential risk factors with genital tuberculosis was tested by Chi-squared test. A similar analysis was performed to identify clinical features associated with genital tuberculosis. Results: The estimated prevalence of FGTB was 45.1 cases per 100,000 women (95% confidence interval [CI]: 16.6-98.1). Infertility and oligomenorrhoea were identified as clinical features associated with FGTB. Past history of tuberculosis and history of close contact with tuberculosis cases were identified as risk factors. Conclusions: This study shows the prevalence of FGTB among the female population of the Andaman Islands. Though the estimated prevalence was close to the expected prevalence, but as only 63.8% of the eligible women could be adequately screened, a much higher prevalence of FGTB could not be ruled out. Infertility, oligomenorrhoea, past history of tuberculosis and contact with tuberculosis case were identified as factors associated with genital tuberculosis. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
22. Promotion of DNA end resection by BRCA1-BARD1 in homologous recombination.
- Author
-
Salunkhe S, Daley JM, Kaur H, Tomimatsu N, Xue C, Raina VB, Jasper AM, Rogers CM, Li W, Zhou S, Mojidra R, Kwon Y, Fang Q, Ji JH, Badamchi Shabestari A, Fitzgerald O, Dinh H, Mukherjee B, Habib AA, Hromas R, Mazin AV, Wasmuth EV, Olsen SK, Libich DS, Zhou D, Zhao W, Greene EC, Burma S, and Sung P
- Subjects
- Humans, DNA metabolism, DNA genetics, DNA Helicases, DNA Repair, DNA Repair Enzymes, DNA, Single-Stranded metabolism, Protein Binding, Rad51 Recombinase metabolism, Recombinational DNA Repair, Single Molecule Imaging, Up-Regulation, Werner Syndrome Helicase metabolism, Werner Syndrome Helicase genetics, BRCA1 Protein metabolism, BRCA1 Protein genetics, DNA Breaks, Double-Stranded, Exodeoxyribonucleases metabolism, Homologous Recombination, RecQ Helicases metabolism, RecQ Helicases genetics, Tumor Suppressor Proteins metabolism, Tumor Suppressor Proteins genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
The licensing step of DNA double-strand break repair by homologous recombination entails resection of DNA ends to generate a single-stranded DNA template for assembly of the repair machinery consisting of the RAD51 recombinase and ancillary factors
1 . DNA end resection is mechanistically intricate and reliant on the tumour suppressor complex BRCA1-BARD1 (ref.2 ). Specifically, three distinct nuclease entities-the 5'-3' exonuclease EXO1 and heterodimeric complexes of the DNA endonuclease DNA2, with either the BLM or WRN helicase-act in synergy to execute the end resection process3 . A major question concerns whether BRCA1-BARD1 directly regulates end resection. Here, using highly purified protein factors, we provide evidence that BRCA1-BARD1 physically interacts with EXO1, BLM and WRN. Importantly, with reconstituted biochemical systems and a single-molecule analytical tool, we show that BRCA1-BARD1 upregulates the activity of all three resection pathways. We also demonstrate that BRCA1 and BARD1 harbour stand-alone modules that contribute to the overall functionality of BRCA1-BARD1. Moreover, analysis of a BARD1 mutant impaired in DNA binding shows the importance of this BARD1 attribute in end resection, both in vitro and in cells. Thus, BRCA1-BARD1 enhances the efficiency of all three long-range DNA end resection pathways during homologous recombination in human cells., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
23. Distinct roles of the two BRCA2 DNA binding domains in DNA damage repair and replication fork preservation.
- Author
-
Neal F, Li W, Uhrig ME, Sharma N, Syed S, Burma S, Hromas R, Mazin A, Dray E, Libich D, Olsen S, Wasmuth E, Zhao W, Sørensen CS, Wiese C, Kwon Y, and Sung P
- Abstract
Homologous recombination (HR) is a highly conserved tool for the removal of DNA double-strand breaks (DSBs) and the preservation of stalled and damaged DNA replication forks. Successful completion of HR requires the tumor suppressor BRCA2. Germline mutations in BRCA2 lead to familial breast, ovarian, and other cancers, underscoring the importance of this protein for maintaining genome stability. BRCA2 harbors two distinct DNA binding domains, one that possesses three oligonucleotide/oligosaccharide binding (OB) folds (known as the OB-DBD), and with the other residing in the C-terminal recombinase binding domain (termed the CTRB-DBD) encoded by the last gene exon. Here, we employ a combination of genetic, biochemical, and cellular approaches to delineate contributions of these two DNA binding domains toward HR and the maintenance of stressed DNA replication forks. We show that OB-DBD and CTRB-DBD confer ssDNA and dsDNA binding capabilities to BRCA2, respectively, and that BRCA2 variants mutated in either DNA binding domain are impaired in the ability to load the recombinase RAD51 onto ssDNA pre-occupied by RPA. While the CTRB-DBD mutant is modestly affected for HR, it exhibits a strong defect in the protection of stressed replication forks. In contrast, the OB-DBD is indispensable for both BRCA2 functions. Our study thus defines the unique contributions of the two BRCA2 DNA binding domains in genome maintenance.
- Published
- 2024
- Full Text
- View/download PDF
24. DSS1 restrains BRCA2's engagement with dsDNA for homologous recombination, replication fork protection, and R-loop homeostasis.
- Author
-
Huang Y, Li W, Foo T, Ji JH, Wu B, Tomimatsu N, Fang Q, Gao B, Long M, Xu J, Maqbool R, Mukherjee B, Ni T, Alejo S, He Y, Burma S, Lan L, Xia B, and Zhao W
- Subjects
- Humans, Homeostasis, Protein Binding, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Protein Domains, Cell Line, Tumor, DNA Damage, Proteasome Endopeptidase Complex, BRCA2 Protein metabolism, BRCA2 Protein genetics, BRCA2 Protein chemistry, DNA Replication, Homologous Recombination, DNA metabolism, Rad51 Recombinase metabolism, Rad51 Recombinase genetics, Mutation, DNA, Single-Stranded metabolism, DNA, Single-Stranded genetics
- Abstract
DSS1, essential for BRCA2-RAD51 dependent homologous recombination (HR), associates with the helical domain (HD) and OB fold 1 (OB1) of the BRCA2 DSS1/DNA-binding domain (DBD) which is frequently targeted by cancer-associated pathogenic variants. Herein, we reveal robust ss/dsDNA binding abilities in HD-OB1 subdomains and find that DSS1 shuts down HD-OB1's DNA binding to enable ssDNA targeting of the BRCA2-RAD51 complex. We show that C-terminal helix mutations of DSS1, including the cancer-associated R57Q mutation, disrupt this DSS1 regulation and permit dsDNA binding of HD-OB1/BRCA2-DBD. Importantly, these DSS1 mutations impair BRCA2/RAD51 ssDNA loading and focus formation and cause decreased HR efficiency, destabilization of stalled forks and R-loop accumulation, and hypersensitize cells to DNA-damaging agents. We propose that DSS1 restrains the intrinsic dsDNA binding of BRCA2-DBD to ensure BRCA2/RAD51 targeting to ssDNA, thereby promoting optimal execution of HR, and potentially replication fork protection and R-loop suppression., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
25. An untapped window of opportunity for glioma: targeting therapy-induced senescence prior to recurrence.
- Author
-
Riviere-Cazaux C, Carlstrom LP, Neth BJ, Olson IE, Rajani K, Rahman M, Ikram S, Mansour MA, Mukherjee B, Warrington AE, Short SC, von Zglinicki T, Brown DA, Burma S, Tchkonia T, Schafer MJ, Baker DJ, Kizilbash SH, Kirkland JL, and Burns TC
- Abstract
High-grade gliomas are primary brain tumors that are incredibly refractory long-term to surgery and chemoradiation, with no proven durable salvage therapies for patients that have failed conventional treatments. Post-treatment, the latent glioma and its microenvironment are characterized by a senescent-like state of mitotic arrest and a senescence-associated secretory phenotype (SASP) induced by prior chemoradiation. Although senescence was once thought to be irreversible, recent evidence has demonstrated that cells may escape this state and re-enter the cell cycle, contributing to tumor recurrence. Moreover, senescent tumor cells could spur the growth of their non-senescent counterparts, thereby accelerating recurrence. In this review, we highlight emerging evidence supporting the use of senolytic agents to ablate latent, senescent-like cells that could contribute to tumor recurrence. We also discuss how senescent cell clearance can decrease the SASP within the tumor microenvironment thereby reducing tumor aggressiveness at recurrence. Finally, senolytics could improve the long-term sequelae of prior therapy on cognition and bone marrow function. We critically review the senolytic drugs currently under preclinical and clinical investigation and the potential challenges that may be associated with deploying senolytics against latent glioma. In conclusion, senescence in glioma and the microenvironment are critical and potential targets for delaying or preventing tumor recurrence and improving patient functional outcomes through senotherapeutics., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
26. Crucial roles of the BRCA1-BARD1 E3 ubiquitin ligase activity in homology-directed DNA repair.
- Author
-
Wang M, Li W, Tomimatsu N, Yu CH, Ji JH, Alejo S, Witus SR, Alimbetov D, Fitzgerald O, Wu B, Wang Q, Huang Y, Gan Y, Dong F, Kwon Y, Sareddy GR, Curiel TJ, Habib AA, Hromas R, Dos Santos Passos C, Yao T, Ivanov DN, Brzovic PS, Burma S, Klevit RE, and Zhao W
- Subjects
- Humans, BRCA1 Protein metabolism, Ubiquitination, Histones genetics, Histones metabolism, Ubiquitin-Protein Ligases metabolism, Recombinational DNA Repair, DNA, DNA Repair, Tumor Suppressor Proteins metabolism, Neoplasms
- Abstract
The tumor-suppressor breast cancer 1 (BRCA1) in complex with BRCA1-associated really interesting new gene (RING) domain 1 (BARD1) is a RING-type ubiquitin E3 ligase that modifies nucleosomal histone and other substrates. The importance of BRCA1-BARD1 E3 activity in tumor suppression remains highly controversial, mainly stemming from studying mutant ligase-deficient BRCA1-BARD1 species that we show here still retain significant ligase activity. Using full-length BRCA1-BARD1, we establish robust BRCA1-BARD1-mediated ubiquitylation with specificity, uncover multiple modes of activity modulation, and construct a truly ligase-null variant and a variant specifically impaired in targeting nucleosomal histones. Cells expressing either of these BRCA1-BARD1 separation-of-function alleles are hypersensitive to DNA-damaging agents. Furthermore, we demonstrate that BRCA1-BARD1 ligase is not only required for DNA resection during homology-directed repair (HDR) but also contributes to later stages for HDR completion. Altogether, our findings reveal crucial, previously unrecognized roles of BRCA1-BARD1 ligase activity in genome repair via HDR, settle prior controversies regarding BRCA1-BARD1 ligase functions, and catalyze new efforts to uncover substrates related to tumor suppression., Competing Interests: Declaration of interests D.N.I. is a co-founder and a shareholder of E3 Bioscience LLC, a commercial entity that manufactures FRET-active E2∼Ub conjugates used in this study., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
27. Structural insights into BCDX2 complex function in homologous recombination.
- Author
-
Rawal Y, Jia L, Meir A, Zhou S, Kaur H, Ruben EA, Kwon Y, Bernstein KA, Jasin M, Taylor AB, Burma S, Hromas R, Mazin AV, Zhao W, Zhou D, Wasmuth EV, Greene EC, Sung P, and Olsen SK
- Subjects
- Humans, Cryoelectron Microscopy, DNA Replication, DNA, Single-Stranded chemistry, DNA, Single-Stranded genetics, DNA, Single-Stranded metabolism, DNA, Single-Stranded ultrastructure, Neoplasms genetics, Nucleoproteins metabolism, Protein Subunits chemistry, Protein Subunits metabolism, Rad51 Recombinase chemistry, Rad51 Recombinase metabolism, Rad51 Recombinase ultrastructure, Substrate Specificity, DNA-Binding Proteins chemistry, DNA-Binding Proteins metabolism, DNA-Binding Proteins ultrastructure, Homologous Recombination, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism, Multiprotein Complexes ultrastructure
- Abstract
Homologous recombination (HR) fulfils a pivotal role in the repair of DNA double-strand breaks and collapsed replication forks
1 . HR depends on the products of several paralogues of RAD51, including the tetrameric complex of RAD51B, RAD51C, RAD51D and XRCC2 (BCDX2)2 . BCDX2 functions as a mediator of nucleoprotein filament assembly by RAD51 and single-stranded DNA (ssDNA) during HR, but its mechanism remains undefined. Here we report cryogenic electron microscopy reconstructions of human BCDX2 in apo and ssDNA-bound states. The structures reveal how the amino-terminal domains of RAD51B, RAD51C and RAD51D participate in inter-subunit interactions that underpin complex formation and ssDNA-binding specificity. Single-molecule DNA curtain analysis yields insights into how BCDX2 enhances RAD51-ssDNA nucleoprotein filament assembly. Moreover, our cryogenic electron microscopy and functional analyses explain how RAD51C alterations found in patients with cancer3-6 inactivate DNA binding and the HR mediator activity of BCDX2. Our findings shed light on the role of BCDX2 in HR and provide a foundation for understanding how pathogenic alterations in BCDX2 impact genome repair., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
28. DNA binding and RAD51 engagement by the BRCA2 C-terminus orchestrate DNA repair and replication fork preservation.
- Author
-
Kwon Y, Rösner H, Zhao W, Selemenakis P, He Z, Kawale AS, Katz JN, Rogers CM, Neal FE, Badamchi Shabestari A, Petrosius V, Singh AK, Joel MZ, Lu L, Holloway SP, Burma S, Mukherjee B, Hromas R, Mazin A, Wiese C, Sørensen CS, and Sung P
- Subjects
- DNA Repair, BRCA2 Protein metabolism, DNA, Homologous Recombination, DNA Replication, Rad51 Recombinase genetics, Rad51 Recombinase metabolism
- Abstract
The tumor suppressor BRCA2 participates in DNA double-strand break repair by RAD51-dependent homologous recombination and protects stressed DNA replication forks from nucleolytic attack. We demonstrate that the C-terminal Recombinase Binding (CTRB) region of BRCA2, encoded by gene exon 27, harbors a DNA binding activity. CTRB alone stimulates the DNA strand exchange activity of RAD51 and permits the utilization of RPA-coated ssDNA by RAD51 for strand exchange. Moreover, CTRB functionally synergizes with the Oligonucleotide Binding fold containing DNA binding domain and BRC4 repeat of BRCA2 in RPA-RAD51 exchange on ssDNA. Importantly, we show that the DNA binding and RAD51 interaction attributes of the CTRB are crucial for homologous recombination and protection of replication forks against MRE11-mediated attrition. Our findings shed light on the role of the CTRB region in genome repair, reveal remarkable functional plasticity of BRCA2, and help explain why deletion of Brca2 exon 27 impacts upon embryonic lethality., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
29. EEPD1 promotes repair of oxidatively-stressed replication forks.
- Author
-
Jaiswal AS, Kim HS, Schärer OD, Sharma N, Williamson EA, Srinivasan G, Phillips L, Kong K, Arya S, Misra A, Dutta A, Gupta Y, Walter CA, Burma S, Narayan S, Sung P, Nickoloff JA, and Hromas R
- Abstract
Unrepaired oxidatively-stressed replication forks can lead to chromosomal instability and neoplastic transformation or cell death. To meet these challenges cells have evolved a robust mechanism to repair oxidative genomic DNA damage through the base excision repair (BER) pathway, but less is known about repair of oxidative damage at replication forks. We found that depletion or genetic deletion of EEPD1 decreases clonogenic cell survival after oxidative DNA damage. We demonstrate that EEPD1 is recruited to replication forks stressed by oxidative damage induced by H
2 O2 and that EEPD1 promotes replication fork repair and restart and decreases chromosomal abnormalities after such damage. EEPD1 binds to abasic DNA structures and promotes resolution of genomic abasic sites after oxidative stress. We further observed that restoration of expression of EEPD1 via expression vector transfection restores cell survival and suppresses chromosomal abnormalities induced by oxidative stress in EEPD1-depleted cells. Consistent with this, we found that EEPD1 preserves replication fork integrity by preventing oxidatively-stressed unrepaired fork fusion, thereby decreasing chromosome instability and mitotic abnormalities. Our results indicate a novel role for EEPD1 in replication fork preservation and maintenance of chromosomal stability during oxidative stress., (© The Author(s) 2023. Published by Oxford University Press on behalf of NAR Cancer.)- Published
- 2023
- Full Text
- View/download PDF
30. Regulation of TORC1 by MAPK Signaling Determines Sensitivity and Acquired Resistance to Trametinib in Pediatric BRAFV600E Brain Tumor Models.
- Author
-
Li F, Bondra KM, Ghilu S, Studebaker A, Liu Q, Michalek JE, Kogiso M, Li XN, Kalapurakal JA, James CD, Burma S, Kurmasheva RT, and Houghton PJ
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Humans, Mice, Mitogen-Activated Protein Kinase Kinases, Mutation, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf genetics, Sirolimus, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Glioma drug therapy, Glioma genetics, Glioma metabolism, Mechanistic Target of Rapamycin Complex 1 genetics, Pyridones therapeutic use, Pyrimidinones therapeutic use
- Abstract
Purpose: We investigated why three patient-derived xenograft (PDX) childhood BRAFV600E-mutant brain tumor models are highly sensitive to trametinib. Mechanisms of acquired resistance selected in situ, and approaches to prevent resistance were also examined, which may translate to both low-grade glioma (LGG) molecular subtypes., Experimental Design: Sensitivity to trametinib [MEK inhibitor (MEKi)] alone or in combination with rapamycin (TORC1 inhibitor), was evaluated in pediatric PDX models. The effect of combined treatment of trametinib with rapamycin on development of trametinib resistance in vivo was examined. PDX tissue and tumor cells from trametinib-resistant xenografts were characterized., Results: In pediatric models TORC1 is activated through ERK-mediated inactivation of the tuberous sclerosis complex (TSC): consequently inhibition of MEK also suppressed TORC1 signaling. Trametinib-induced tumor regression correlated with dual inhibition of MAPK/TORC1 signaling, and decoupling TORC1 regulation from BRAF/MAPK control conferred trametinib resistance. In mice, acquired resistance to trametinib developed within three cycles of therapy in all three PDX models. Resistance to trametinib developed in situ is tumor-cell-intrinsic and the mechanism was tumor line specific. Rapamycin retarded or blocked development of resistance., Conclusions: In these three pediatric BRAF-mutant brain tumors, TORC1 signaling is controlled by the MAPK cascade. Trametinib suppressed both MAPK/TORC1 pathways leading to tumor regression. While low-dose intermittent rapamycin to enhance inhibition of TORC1 only modestly enhanced the antitumor activity of trametinib, it prevented or retarded development of trametinib resistance, suggesting future therapeutic approaches using rapamycin analogs in combination with MEKis that may be therapeutically beneficial in both KIAA1549::BRAF- and BRAFV600E-driven gliomas., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
31. EGFR ligand shifts the role of EGFR from oncogene to tumour suppressor in EGFR-amplified glioblastoma by suppressing invasion through BIN3 upregulation.
- Author
-
Guo G, Gong K, Beckley N, Zhang Y, Yang X, Chkheidze R, Hatanpaa KJ, Garzon-Muvdi T, Koduru P, Nayab A, Jenks J, Sathe AA, Liu Y, Xing C, Wu SY, Chiang CM, Mukherjee B, Burma S, Wohlfeld B, Patel T, Mickey B, Abdullah K, Youssef M, Pan E, Gerber DE, Tian S, Sarkaria JN, McBrayer SK, Zhao D, and Habib AA
- Subjects
- Cell Line, Tumor, ErbB Receptors genetics, ErbB Receptors metabolism, Humans, Ligands, Oncogenes genetics, Up-Regulation, Glioblastoma metabolism, Microfilament Proteins metabolism
- Abstract
The epidermal growth factor receptor (EGFR) is a prime oncogene that is frequently amplified in glioblastomas. Here we demonstrate a new tumour-suppressive function of EGFR in EGFR-amplified glioblastomas regulated by EGFR ligands. Constitutive EGFR signalling promotes invasion via activation of a TAB1-TAK1-NF-κB-EMP1 pathway, resulting in large tumours and decreased survival in orthotopic models. Ligand-activated EGFR promotes proliferation and surprisingly suppresses invasion by upregulating BIN3, which inhibits a DOCK7-regulated Rho GTPase pathway, resulting in small hyperproliferating non-invasive tumours and improved survival. Data from The Cancer Genome Atlas reveal that in EGFR-amplified glioblastomas, a low level of EGFR ligands confers a worse prognosis, whereas a high level of EGFR ligands confers an improved prognosis. Thus, increased EGFR ligand levels shift the role of EGFR from oncogene to tumour suppressor in EGFR-amplified glioblastomas by suppressing invasion. The tumour-suppressive function of EGFR can be activated therapeutically using tofacitinib, which suppresses invasion by increasing EGFR ligand levels and upregulating BIN3., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
32. Selective Vulnerability of Senescent Glioblastoma Cells to BCL-XL Inhibition.
- Author
-
Rahman M, Olson I, Mansour M, Carlstrom LP, Sutiwisesak R, Saber R, Rajani K, Warrington AE, Howard A, Schroeder M, Chen S, Decker PA, Sananikone EF, Zhu Y, Tchkonia T, Parney IF, Burma S, Brown D, Rodriguez M, Sarkaria JN, Kirkland JL, and Burns TC
- Subjects
- Apoptosis, Cell Line, Tumor, Cellular Senescence, Humans, Proto-Oncogene Proteins c-bcl-2 metabolism, Senotherapeutics, Temozolomide pharmacology, Glioblastoma drug therapy, Glioblastoma genetics, Glioblastoma metabolism
- Abstract
Glioblastoma (GBM) is a rapidly fatal malignancy typically treated with radiation and temozolomide (TMZ), an alkylating chemotherapeutic. These cytotoxic therapies cause oxidative stress and DNA damage, yielding a senescent-like state of replicative arrest in surviving tumor cells. Unfortunately, recurrence is inevitable and may be driven by surviving tumor cells eventually escaping senescence. A growing number of so-called "senolytic" drugs have been recently identified that are defined by their ability to selectively eliminate senescent cells. A growing inventory of senolytic drugs is under consideration for several diseases associated with aging, inflammation, DNA damage, as well as cancer. Ablation of senescent tumor cells after radiation and chemotherapy could help mitigate recurrence by decreasing the burden of residual tumor cells at risk of recurrence. This strategy has not been previously explored for GBM. We evaluated a panel of 10 previously described senolytic drugs to determine whether any could exhibit selective activity against human GBM persisting after exposure to radiation or TMZ. Three of the 10 drugs have known activity against BCL-XL and preferentially induced apoptosis in radiated or TMZ-treated glioma. This senolytic activity was observed in 12 of 12 human GBM cell lines. Efficacy could not be replicated with BCL-2 inhibition or senolytic agents acting against other putative senolytic targets. Knockdown of BCL-XL decreased survival of radiated GBM cells, whereas knockdown of BCL-2 or BCL-W yielded no senolytic effect., Implications: These findings imply that molecularly heterogeneous GBM lines share selective senescence-induced BCL-XL dependency increase the significance and translational relevance of the senolytic therapy for latent glioma., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
33. Targeting radioresistance and replication fork stability in prostate cancer.
- Author
-
Li X, Baek G, Carreira S, Yuan W, Ma S, Hofstad M, Lee S, Gao Y, Bertan C, Fenor de la Maza MLD, Alluri PG, Burma S, Chen BP, Raj GV, de Bono J, Pommier Y, and Mani RS
- Subjects
- Cell Cycle Proteins genetics, Cell Line, Tumor, Histones metabolism, Humans, Male, Transcription Factors genetics, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant radiotherapy
- Abstract
The bromodomain and extraterminal (BET) family of chromatin reader proteins bind to acetylated histones and regulate gene expression. The development of BET inhibitors (BETi) has expanded our knowledge of BET protein function beyond transcriptional regulation and has ushered several prostate cancer (PCa) clinical trials. However, BETi as a single agent is not associated with antitumor activity in patients with castration-resistant prostate cancer (CRPC). We hypothesized novel combinatorial strategies are likely to enhance the efficacy of BETi. By using PCa patient-derived explants and xenograft models, we show that BETi treatment enhanced the efficacy of radiation therapy (RT) and overcame radioresistance. Mechanistically, BETi potentiated the activity of RT by blocking DNA repair. We also report a synergistic relationship between BETi and topoisomerase I (TOP1) inhibitors (TOP1i). We show that the BETi OTX015 synergized with the new class of synthetic noncamptothecin TOP1i, LMP400 (indotecan), to block tumor growth in aggressive CRPC xenograft models. Mechanistically, BETi potentiated the antitumor activity of TOP1i by disrupting replication fork stability. Longitudinal analysis of patient tumors indicated that TOP1 transcript abundance increased as patients progressed from hormone-sensitive prostate cancer to CRPC. TOP1 was highly expressed in metastatic CRPC, and its expression correlated with the expression of BET family genes. These studies open new avenues for the rational combinatorial treatment of aggressive PCa.
- Published
- 2022
- Full Text
- View/download PDF
34. Bloom helicase mediates formation of large single-stranded DNA loops during DNA end processing.
- Author
-
Xue C, Salunkhe SJ, Tomimatsu N, Kawale AS, Kwon Y, Burma S, Sung P, and Greene EC
- Subjects
- DNA genetics, Homologous Recombination genetics, DNA, Single-Stranded genetics, RecQ Helicases genetics, RecQ Helicases metabolism
- Abstract
Bloom syndrome (BS) is associated with a profoundly increased cancer risk and is caused by mutations in the Bloom helicase (BLM). BLM is involved in the nucleolytic processing of the ends of DNA double-strand breaks (DSBs), to yield long 3' ssDNA tails that serve as the substrate for break repair by homologous recombination (HR). Here, we use single-molecule imaging to demonstrate that BLM mediates formation of large ssDNA loops during DNA end processing. A BLM mutant lacking the N-terminal domain (NTD) retains vigorous in vitro end processing activity but fails to generate ssDNA loops. This same mutant supports DSB end processing in cells, however, these cells do not form RAD51 DNA repair foci and the processed DSBs are channeled into synthesis-dependent strand annealing (SSA) instead of HR-mediated repair, consistent with a defect in RAD51 filament formation. Together, our results provide insights into BLM functions during homologous recombination., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
35. Elimination of Radiation-Induced Senescence in the Brain Tumor Microenvironment Attenuates Glioblastoma Recurrence.
- Author
-
Fletcher-Sananikone E, Kanji S, Tomimatsu N, Di Cristofaro LFM, Kollipara RK, Saha D, Floyd JR, Sung P, Hromas R, Burns TC, Kittler R, Habib AA, Mukherjee B, and Burma S
- Subjects
- Aniline Compounds pharmacology, Animals, Antineoplastic Agents pharmacology, Astrocytes drug effects, Astrocytes metabolism, Astrocytes pathology, Brain drug effects, Brain metabolism, Glioblastoma drug therapy, Glioblastoma etiology, Glioblastoma metabolism, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local etiology, Neoplasm Recurrence, Local metabolism, Sulfonamides pharmacology, Brain pathology, Cellular Senescence, Gamma Rays adverse effects, Glioblastoma pathology, Neoplasm Recurrence, Local pathology, Senescence-Associated Secretory Phenotype, Tumor Microenvironment
- Abstract
Glioblastomas (GBM) are routinely treated with ionizing radiation (IR) but inevitably recur and develop therapy resistance. During treatment, the tissue surrounding tumors is also irradiated. IR potently induces senescence, and senescent stromal cells can promote the growth of neighboring tumor cells by secreting factors that create a senescence-associated secretory phenotype (SASP). Here, we carried out transcriptomic and tumorigenicity analyses in irradiated mouse brains to elucidate how radiotherapy-induced senescence of non-neoplastic brain cells promotes tumor growth. Following cranial irradiation, widespread senescence in the brain occurred, with the astrocytic population being particularly susceptible. Irradiated brains showed an altered transcriptomic profile characterized by upregulation of CDKN1A (p21), a key enforcer of senescence, and several SASP factors, including HGF, the ligand of the receptor tyrosine kinase (RTK) Met. Preirradiation of mouse brains increased Met-driven growth and invasiveness of orthotopically implanted glioma cells. Importantly, irradiated p21
-/- mouse brains did not exhibit senescence and consequently failed to promote tumor growth. Senescent astrocytes secreted HGF to activate Met in glioma cells and to promote their migration and invasion in vitro , which could be blocked by HGF-neutralizing antibodies or the Met inhibitor crizotinib. Crizotinib also slowed the growth of glioma cells implanted in preirradiated brains. Treatment with the senolytic drug ABT-263 (navitoclax) selectively killed senescent astrocytes in vivo , significantly attenuating growth of glioma cells implanted in preirradiated brains. These results indicate that SASP factors in the irradiated tumor microenvironment drive GBM growth via RTK activation, underscoring the potential utility of adjuvant senolytic therapy for preventing GBM recurrence after radiotherapy. SIGNIFICANCE: This study uncovers mechanisms by which radiotherapy can promote GBM recurrence by inducing senescence in non-neoplastic brain cells, suggesting that senolytic therapy can blunt recurrent GBM growth and aggressiveness., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
36. Comprehensive targeting of resistance to inhibition of RTK signaling pathways by using glucocorticoids.
- Author
-
Gong K, Guo G, Beckley NA, Yang X, Zhang Y, Gerber DE, Minna JD, Burma S, Zhao D, Akbay EA, and Habib AA
- Subjects
- A549 Cells, Animals, Carcinoma, Non-Small-Cell Lung, Cytokines metabolism, Disease Models, Animal, ErbB Receptors drug effects, Female, Humans, Lung Neoplasms, Mice, Mice, Inbred NOD, Mice, SCID, Mice, Transgenic, Prednisone, STAT3 Transcription Factor metabolism, Thalidomide, Tumor Necrosis Factor Inhibitors, Up-Regulation, Drug Resistance, Neoplasm drug effects, Glucocorticoids pharmacology, Protein Kinase Inhibitors pharmacology, Signal Transduction drug effects
- Abstract
Inhibition of RTK pathways in cancer triggers an adaptive response that promotes therapeutic resistance. Because the adaptive response is multifaceted, the optimal approach to blunting it remains undetermined. TNF upregulation is a biologically significant response to EGFR inhibition in NSCLC. Here, we compared a specific TNF inhibitor (etanercept) to thalidomide and prednisone, two drugs that block TNF and also other inflammatory pathways. Prednisone is significantly more effective in suppressing EGFR inhibition-induced inflammatory signals. Remarkably, prednisone induces a shutdown of bypass RTK signaling and inhibits key resistance signals such as STAT3, YAP and TNF-NF-κB. Combined with EGFR inhibition, prednisone is significantly superior to etanercept or thalidomide in durably suppressing tumor growth in multiple mouse models, indicating that a broad suppression of adaptive signals is more effective than blocking a single component. We identify prednisone as a drug that can effectively inhibit adaptive resistance with acceptable toxicity in NSCLC and other cancers., (© 2021. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2021
- Full Text
- View/download PDF
37. Therapy-Induced Senescence: Opportunities to Improve Anticancer Therapy.
- Author
-
Prasanna PG, Citrin DE, Hildesheim J, Ahmed MM, Venkatachalam S, Riscuta G, Xi D, Zheng G, Deursen JV, Goronzy J, Kron SJ, Anscher MS, Sharpless NE, Campisi J, Brown SL, Niedernhofer LJ, O'Loghlen A, Georgakilas AG, Paris F, Gius D, Gewirtz DA, Schmitt CA, Abazeed ME, Kirkland JL, Richmond A, Romesser PB, Lowe SW, Gil J, Mendonca MS, Burma S, Zhou D, and Coleman CN
- Subjects
- Biomarkers, Humans, Senescence-Associated Secretory Phenotype, Cellular Senescence, Neoplasms drug therapy, Neoplasms pathology
- Abstract
Cellular senescence is an essential tumor suppressive mechanism that prevents the propagation of oncogenically activated, genetically unstable, and/or damaged cells. Induction of tumor cell senescence is also one of the underlying mechanisms by which cancer therapies exert antitumor activity. However, an increasing body of evidence from preclinical studies demonstrates that radiation and chemotherapy cause accumulation of senescent cells (SnCs) both in tumor and normal tissue. SnCs in tumors can, paradoxically, promote tumor relapse, metastasis, and resistance to therapy, in part, through expression of the senescence-associated secretory phenotype. In addition, SnCs in normal tissue can contribute to certain radiation- and chemotherapy-induced side effects. Because of its multiple roles, cellular senescence could serve as an important target in the fight against cancer. This commentary provides a summary of the discussion at the National Cancer Institute Workshop on Radiation, Senescence, and Cancer (August 10-11, 2020, National Cancer Institute, Bethesda, MD) regarding the current status of senescence research, heterogeneity of therapy-induced senescence, current status of senotherapeutics and molecular biomarkers, a concept of "one-two punch" cancer therapy (consisting of therapeutics to induce tumor cell senescence followed by selective clearance of SnCs), and its integration with personalized adaptive tumor therapy. It also identifies key knowledge gaps and outlines future directions in this emerging field to improve treatment outcomes for cancer patients., (© The Author(s) 2021. Published by Oxford University Press.)
- Published
- 2021
- Full Text
- View/download PDF
38. Inhibition of the de novo pyrimidine biosynthesis pathway limits ribosomal RNA transcription causing nucleolar stress in glioblastoma cells.
- Author
-
Lafita-Navarro MC, Venkateswaran N, Kilgore JA, Kanji S, Han J, Barnes S, Williams NS, Buszczak M, Burma S, and Conacci-Sorrell M
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Biphenyl Compounds pharmacology, Biphenyl Compounds therapeutic use, Brain Neoplasms pathology, Cell Line, Tumor, Cell Nucleolus metabolism, Dihydroorotate Dehydrogenase, Drug Screening Assays, Antitumor, Female, Glioblastoma pathology, Humans, Mice, Multienzyme Complexes antagonists & inhibitors, Multienzyme Complexes metabolism, Nucleophosmin, Orotate Phosphoribosyltransferase antagonists & inhibitors, Orotate Phosphoribosyltransferase metabolism, Orotidine-5'-Phosphate Decarboxylase antagonists & inhibitors, Orotidine-5'-Phosphate Decarboxylase metabolism, Oxidoreductases Acting on CH-CH Group Donors antagonists & inhibitors, Oxidoreductases Acting on CH-CH Group Donors metabolism, RNA, Ribosomal biosynthesis, Ribosomes drug effects, Ribosomes metabolism, Stress, Physiological drug effects, Transcription, Genetic drug effects, Tumor Suppressor Protein p53 metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Brain Neoplasms drug therapy, Cell Nucleolus drug effects, Glioblastoma drug therapy, Pyrimidines biosynthesis
- Abstract
Glioblastoma is the most common and aggressive type of cancer in the brain; its poor prognosis is often marked by reoccurrence due to resistance to the chemotherapeutic agent temozolomide, which is triggered by an increase in the expression of DNA repair enzymes such as MGMT. The poor prognosis and limited therapeutic options led to studies targeted at understanding specific vulnerabilities of glioblastoma cells. Metabolic adaptations leading to increased synthesis of nucleotides by de novo biosynthesis pathways are emerging as key alterations driving glioblastoma growth. In this study, we show that enzymes necessary for the de novo biosynthesis of pyrimidines, DHODH and UMPS, are elevated in high grade gliomas and in glioblastoma cell lines. We demonstrate that DHODH's activity is necessary to maintain ribosomal DNA transcription (rDNA). Pharmacological inhibition of DHODH with the specific inhibitors brequinar or ML390 effectively depleted the pool of pyrimidines in glioblastoma cells grown in vitro and in vivo and impaired rDNA transcription, leading to nucleolar stress. Nucleolar stress was visualized by the aberrant redistribution of the transcription factor UBF and the nucleolar organizer nucleophosmin 1 (NPM1), as well as the stabilization of the transcription factor p53. Moreover, DHODH inhibition decreased the proliferation of glioblastoma cells, including temozolomide-resistant cells. Importantly, the addition of exogenous uridine, which reconstitutes the cellular pool of pyrimidine by the salvage pathway, to the culture media recovered the impaired rDNA transcription, nucleolar morphology, p53 levels, and proliferation of glioblastoma cells caused by the DHODH inhibitors. Our in vivo data indicate that while inhibition of DHODH caused a dramatic reduction in pyrimidines in tumor cells, it did not affect the overall pyrimidine levels in normal brain and liver tissues, suggesting that pyrimidine production by the salvage pathway may play an important role in maintaining these nucleotides in normal cells. Our study demonstrates that glioblastoma cells heavily rely on the de novo pyrimidine biosynthesis pathway to generate ribosomal RNA (rRNA) and thus, we identified an approach to inhibit ribosome production and consequently the proliferation of glioblastoma cells through the specific inhibition of the de novo pyrimidine biosynthesis pathway., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
39. Specificity of end resection pathways for double-strand break regions containing ribonucleotides and base lesions.
- Author
-
Daley JM, Tomimatsu N, Hooks G, Wang W, Miller AS, Xue X, Nguyen KA, Kaur H, Williamson E, Mukherjee B, Hromas R, Burma S, and Sung P
- Subjects
- Blotting, Western, Cell Line, Tumor, DNA Glycosylases genetics, DNA Repair Enzymes genetics, DNA-(Apurinic or Apyrimidinic Site) Lyase genetics, Exodeoxyribonucleases genetics, Fluorescent Antibody Technique, Homologous Recombination genetics, Humans, RecQ Helicases genetics, Recombination, Genetic genetics, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, DNA Breaks, Double-Stranded, Ribonucleotides genetics, Ribonucleotides metabolism
- Abstract
DNA double-strand break repair by homologous recombination begins with nucleolytic resection of the 5' DNA strand at the break ends. Long-range resection is catalyzed by EXO1 and BLM-DNA2, which likely have to navigate through ribonucleotides and damaged bases. Here, we show that a short stretch of ribonucleotides at the 5' terminus stimulates resection by EXO1. Ribonucleotides within a 5' flap are resistant to cleavage by DNA2, and extended RNA:DNA hybrids inhibit both strand separation by BLM and resection by EXO1. Moreover, 8-oxo-guanine impedes EXO1 but enhances resection by BLM-DNA2, and an apurinic/apyrimidinic site stimulates resection by BLM-DNA2 and DNA strand unwinding by BLM. Accordingly, depletion of OGG1 or APE1 leads to greater dependence of DNA resection on DNA2. Importantly, RNase H2A deficiency impairs resection overall, which we attribute to the accumulation of long RNA:DNA hybrids at DNA ends. Our results help explain why eukaryotic cells possess multiple resection nucleases.
- Published
- 2020
- Full Text
- View/download PDF
40. EGFR inhibition triggers an adaptive response by co-opting antiviral signaling pathways in lung cancer.
- Author
-
Gong K, Guo G, Panchani N, Bender ME, Gerber DE, Minna JD, Fattah F, Gao B, Peyton M, Kernstine K, Mukherjee B, Burma S, Chiang CM, Zhang S, Amod Sathe A, Xing C, Dao KH, Zhao D, Akbay EA, and Habib AA
- Subjects
- Animals, Cell Proliferation, Humans, Mice, Protein Kinase Inhibitors pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Lung Neoplasms drug therapy, Signal Transduction
- Abstract
EGFR inhibition is an effective treatment in the minority of non-small cell lung cancer (NSCLC) cases harboring EGFR-activating mutations, but not in EGFR wild type (EGFRwt) tumors. Here, we demonstrate that EGFR inhibition triggers an antiviral defense pathway in NSCLC. Inhibiting mutant EGFR triggers Type I IFN-I upregulation via a RIG-I-TBK1-IRF3 pathway. The ubiquitin ligase TRIM32 associates with TBK1 upon EGFR inhibition, and is required for K63-linked ubiquitination and TBK1 activation. Inhibiting EGFRwt upregulates interferons via an NF-κB-dependent pathway. Inhibition of IFN signaling enhances EGFR-TKI sensitivity in EGFR mutant NSCLC and renders EGFRwt/KRAS mutant NSCLC sensitive to EGFR inhibition in xenograft and immunocompetent mouse models. Furthermore, NSCLC tumors with decreased IFN-I expression are more responsive to EGFR TKI treatment. We propose that IFN-I signaling is a major determinant of EGFR-TKI sensitivity in NSCLC and that a combination of EGFR TKI plus IFN-neutralizing antibody could be useful in most NSCLC patients., Competing Interests: Competing Interests Statement The authors declare no competing interests.
- Published
- 2020
- Full Text
- View/download PDF
41. Efficacy of EGFR plus TNF inhibition in a preclinical model of temozolomide-resistant glioblastoma.
- Author
-
Guo G, Gong K, Puliyappadamba VT, Panchani N, Pan E, Mukherjee B, Damanwalla Z, Bharia S, Hatanpaa KJ, Gerber DE, Mickey BE, Patel TR, Sarkaria JN, Zhao D, Burma S, and Habib AA
- Subjects
- Afatinib administration & dosage, Animals, Apoptosis, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Proliferation, ErbB Receptors antagonists & inhibitors, Female, Glioblastoma metabolism, Glioblastoma pathology, Humans, Mice, Mice, Inbred C57BL, Mice, Nude, Temozolomide administration & dosage, Thalidomide administration & dosage, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Brain Neoplasms drug therapy, Drug Resistance, Neoplasm drug effects, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma drug therapy, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Background: Glioblastoma (GBM) is the most common primary malignant adult brain tumor. Temozolomide (TMZ) is the standard of care and is most effective in GBMs that lack the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). Moreover, even initially responsive tumors develop a secondary resistance to TMZ and become untreatable. Since aberrant epidermal growth factor receptor (EGFR) signaling is widespread in GBM, EGFR inhibition has been tried in multiple clinical trials without success. We recently reported that inhibiting EGFR leads to increased secretion of tumor necrosis factor (TNF) and activation of a survival pathway in GBM. Here, we compare the efficacy of TMZ versus EGFR plus TNF inhibition in an orthotopic mouse model of GBM., Methods: We use an orthotopic model to examine the efficacy of TMZ versus EGFR plus TNF inhibition in multiple subsets of GBMs, including MGMT methylated and unmethylated primary GBMs, recurrent GBMs, and GBMs rendered experimentally resistant to TMZ., Results: The efficacy of the 2 treatments was similar in MGMT methylated GBMs. However, in MGMT unmethylated GBMs, a combination of EGFR plus TNF inhibition was more effective. We demonstrate that the 2 treatment approaches target distinct and non-overlapping pathways. Thus, importantly, EGFR plus TNF inhibition remains effective in TMZ-resistant recurrent GBMs and in GBMs rendered experimentally resistant to TMZ., Conclusion: EGFR inhibition combined with a blunting of the accompanying TNF-driven adaptive response could be a viable therapeutic approach in MGMT unmethylated and recurrent EGFR-expressing GBMs., (Published by Oxford University Press on behalf of the Society for Neuro-Oncology 2019.)
- Published
- 2019
- Full Text
- View/download PDF
42. MiR223-3p promotes synthetic lethality in BRCA1-deficient cancers.
- Author
-
Srinivasan G, Williamson EA, Kong K, Jaiswal AS, Huang G, Kim HS, Schärer O, Zhao W, Burma S, Sung P, and Hromas R
- Subjects
- 3' Untranslated Regions, Cell Line, Tumor, DNA Repair, DNA Replication, Gene Expression Regulation, Neoplastic, Genetic Association Studies, Genomic Instability, Humans, Recombinational DNA Repair, Translocation, Genetic, BRCA1 Protein deficiency, Genetic Predisposition to Disease, MicroRNAs genetics, Neoplasms genetics, Synthetic Lethal Mutations
- Abstract
Defects in DNA repair give rise to genomic instability, leading to neoplasia. Cancer cells defective in one DNA repair pathway can become reliant on remaining repair pathways for survival and proliferation. This attribute of cancer cells can be exploited therapeutically, by inhibiting the remaining repair pathway, a process termed synthetic lethality. This process underlies the mechanism of the Poly-ADP ribose polymerase-1 (PARP1) inhibitors in clinical use, which target BRCA1 deficient cancers, which is indispensable for homologous recombination (HR) DNA repair. HR is the major repair pathway for stressed replication forks, but when BRCA1 is deficient, stressed forks are repaired by back-up pathways such as alternative nonhomologous end-joining (aNHEJ). Unlike HR, aNHEJ is nonconservative, and can mediate chromosomal translocations. In this study we have found that miR223-3p decreases expression of PARP1, CtIP, and Pso4, each of which are aNHEJ components. In most cells, high levels of microRNA (miR) 223-3p repress aNHEJ, decreasing the risk of chromosomal translocations. Deletion of the miR223 locus in mice increases PARP1 levels in hematopoietic cells and enhances their risk of unprovoked chromosomal translocations. We also discovered that cancer cells deficient in BRCA1 or its obligate partner BRCA1-Associated Protein-1 (BAP1) routinely repress miR223-3p to permit repair of stressed replication forks via aNHEJ. Reconstituting the expression of miR223-3p in BRCA1- and BAP1-deficient cancer cells results in reduced repair of stressed replication forks and synthetic lethality. Thus, miR223-3p is a negative regulator of the aNHEJ DNA repair and represents a therapeutic pathway for BRCA1- or BAP1-deficient cancers., Competing Interests: Conflict of interest statement: R.H. has equity in Dialectic Therapeutics, which holds a licensing agreement for use of miR223-3p as a cancer therapeutic agent., (Copyright © 2019 the Author(s). Published by PNAS.)
- Published
- 2019
- Full Text
- View/download PDF
43. Radiation-Induced DNA Damage Cooperates with Heterozygosity of TP53 and PTEN to Generate High-Grade Gliomas.
- Author
-
Todorova PK, Fletcher-Sananikone E, Mukherjee B, Kollipara R, Vemireddy V, Xie XJ, Guida PM, Story MD, Hatanpaa K, Habib AA, Kittler R, Bachoo R, Hromas R, Floyd JR, and Burma S
- Subjects
- Animals, Brain Neoplasms pathology, Female, Glioblastoma pathology, Glioma pathology, Humans, Linear Energy Transfer, Loss of Heterozygosity, Male, Mice, Mice, Inbred C57BL, Neoplasm Grading, Neoplastic Stem Cells pathology, Neoplastic Stem Cells radiation effects, Brain Neoplasms genetics, DNA Breaks, Double-Stranded, Glioblastoma genetics, Glioma genetics, Neoplasms, Radiation-Induced genetics, PTEN Phosphohydrolase genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Glioblastomas are lethal brain tumors that are treated with conventional radiation (X-rays and gamma rays) or particle radiation (protons and carbon ions). Paradoxically, radiation is also a risk factor for GBM development, raising the possibility that radiotherapy of brain tumors could promote tumor recurrence or trigger secondary gliomas. In this study, we determined whether tumor suppressor losses commonly displayed by patients with GBM confer susceptibility to radiation-induced glioma. Mice with Nestin-Cre-driven deletions of Trp53 and Pten alleles were intracranially irradiated with X-rays or charged particles of increasing atomic number and linear energy transfer (LET). Mice with loss of one allele each of Trp53 and Pten did not develop spontaneous gliomas, but were highly susceptible to radiation-induced gliomagenesis. Tumor development frequency after exposure to high-LET particle radiation was significantly higher compared with X-rays, in accordance with the irreparability of DNA double-strand breaks (DSB) induced by high-LET radiation. All resultant gliomas, regardless of radiation quality, presented histopathologic features of grade IV lesions and harbored populations of cancer stem-like cells with tumor-propagating properties. Furthermore, all tumors displayed concomitant loss of heterozygosity of Trp53 and Pten along with frequent amplification of the Met receptor tyrosine kinase, which conferred a stem cell phenotype to tumor cells. Our results demonstrate that radiation-induced DSBs cooperate with preexisting tumor suppressor losses to generate high-grade gliomas. Moreover, our mouse model can be used for studies on radiation-induced development of GBM and therapeutic strategies. SIGNIFICANCE: This study uncovers mechanisms by which ionizing radiation, especially particle radiation, promote GBM development or recurrence., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
44. TNF-driven adaptive response mediates resistance to EGFR inhibition in lung cancer.
- Author
-
Gong K, Guo G, Gerber DE, Gao B, Peyton M, Huang C, Minna JD, Hatanpaa KJ, Kernstine K, Cai L, Xie Y, Zhu H, Fattah FJ, Zhang S, Takahashi M, Mukherjee B, Burma S, Dowell J, Dao K, Papadimitrakopoulou VA, Olivas V, Bivona TG, Zhao D, and Habib AA
- Subjects
- A549 Cells, Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung therapy, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, ErbB Receptors metabolism, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Lung Neoplasms therapy, Mice, Mice, Nude, MicroRNAs genetics, MicroRNAs metabolism, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neoplasms, Experimental therapy, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Neoplasm genetics, RNA, Neoplasm metabolism, Drug Resistance, Neoplasm, Lung Neoplasms metabolism, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Neoplasms, Experimental metabolism, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism
- Abstract
Although aberrant EGFR signaling is widespread in cancer, EGFR inhibition is effective only in a subset of non-small cell lung cancer (NSCLC) with EGFR activating mutations. A majority of NSCLCs express EGFR wild type (EGFRwt) and do not respond to EGFR inhibition. TNF is a major mediator of inflammation-induced cancer. We find that a rapid increase in TNF level is a universal adaptive response to EGFR inhibition in NSCLC, regardless of EGFR status. EGFR signaling actively suppresses TNF mRNA levels by inducing expression of miR-21, resulting in decreased TNF mRNA stability. Conversely, EGFR inhibition results in loss of miR-21 and increased TNF mRNA stability. In addition, TNF-induced NF-κB activation leads to increased TNF transcription in a feed-forward loop. Inhibition of TNF signaling renders EGFRwt-expressing NSCLC cell lines and an EGFRwt patient-derived xenograft (PDX) model highly sensitive to EGFR inhibition. In EGFR-mutant oncogene-addicted cells, blocking TNF enhances the effectiveness of EGFR inhibition. EGFR plus TNF inhibition is also effective in NSCLC with acquired resistance to EGFR inhibition. We suggest concomitant EGFR and TNF inhibition as a potentially new treatment approach that could be beneficial for a majority of lung cancer patients.
- Published
- 2018
- Full Text
- View/download PDF
45. BRD4 Promotes DNA Repair and Mediates the Formation of TMPRSS2-ERG Gene Rearrangements in Prostate Cancer.
- Author
-
Li X, Baek G, Ramanand SG, Sharp A, Gao Y, Yuan W, Welti J, Rodrigues DN, Dolling D, Figueiredo I, Sumanasuriya S, Crespo M, Aslam A, Li R, Yin Y, Mukherjee B, Kanchwala M, Hughes AM, Halsey WS, Chiang CM, Xing C, Raj GV, Burma S, de Bono J, and Mani RS
- Subjects
- Acetylation, Cell Cycle Proteins, Cell Line, Tumor, Chromatin genetics, Chromatin metabolism, DNA Damage, Gene Fusion, Gene Rearrangement, Histones genetics, Histones metabolism, Humans, Male, Nuclear Proteins metabolism, Oncogene Proteins, Fusion metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Transcription Factors metabolism, DNA End-Joining Repair, Nuclear Proteins genetics, Oncogene Proteins, Fusion genetics, Prostatic Neoplasms genetics, Transcription Factors genetics
- Abstract
BRD4 belongs to the bromodomain and extraterminal (BET) family of chromatin reader proteins that bind acetylated histones and regulate gene expression. Pharmacological inhibition of BRD4 by BET inhibitors (BETi) has indicated antitumor activity against multiple cancer types. We show that BRD4 is essential for the repair of DNA double-strand breaks (DSBs) and mediates the formation of oncogenic gene rearrangements by engaging the non-homologous end joining (NHEJ) pathway. Mechanistically, genome-wide DNA breaks are associated with enhanced acetylation of histone H4, leading to BRD4 recruitment, and stable establishment of the DNA repair complex. In support of this, we also show that, in clinical tumor samples, BRD4 protein levels are negatively associated with outcome after prostate cancer (PCa) radiation therapy. Thus, in addition to regulating gene expression, BRD4 is also a central player in the repair of DNA DSBs, with significant implications for cancer therapy., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
46. EGFR Mutations Compromise Hypoxia-Associated Radiation Resistance through Impaired Replication Fork-Associated DNA Damage Repair.
- Author
-
Saki M, Makino H, Javvadi P, Tomimatsu N, Ding LH, Clark JE, Gavin E, Takeda K, Andrews J, Saha D, Story MD, Burma S, and Nirodi CS
- Subjects
- A549 Cells, Acid Anhydride Hydrolases, Carcinoma, Non-Small-Cell Lung drug therapy, Cell Hypoxia, Cell Line, Tumor, Cell Survival drug effects, Cell Survival radiation effects, Cetuximab pharmacology, DNA Damage, DNA Repair, DNA Repair Enzymes genetics, DNA-Binding Proteins genetics, Humans, Lung Neoplasms drug therapy, Mutation, Radiation Tolerance drug effects, Radiation-Sensitizing Agents pharmacology, Carcinoma, Non-Small-Cell Lung genetics, DNA metabolism, DNA Replication, ErbB Receptors genetics, Lung Neoplasms genetics
- Abstract
EGFR signaling has been implicated in hypoxia-associated resistance to radiation or chemotherapy. Non-small cell lung carcinomas (NSCLC) with activating L858R or ΔE746-E750 EGFR mutations exhibit elevated EGFR activity and downstream signaling. Here, relative to wild-type (WT) EGFR, mutant (MT) EGFR expression significantly increases radiosensitivity in hypoxic cells. Gene expression profiling in human bronchial epithelial cells (HBEC) revealed that MT-EGFR expression elevated transcripts related to cell cycle and replication in aerobic and hypoxic conditions and downregulated RAD50, a critical component of nonhomologous end joining and homologous recombination DNA repair pathways. NSCLCs and HBEC with MT-EGFR revealed elevated basal and hypoxia-induced γ-H2AX-associated DNA lesions that were coincident with replication protein A in the S-phase nuclei. DNA fiber analysis showed that, relative to WT-EGFR, MT-EGFR NSCLCs harbored significantly higher levels of stalled replication forks and decreased fork velocities in aerobic and hypoxic conditions. EGFR blockade by cetuximab significantly increased radiosensitivity in hypoxic cells, recapitulating MT-EGFR expression and closely resembling synthetic lethality of PARP inhibition. Implications: This study demonstrates that within an altered DNA damage response of hypoxic NSCLC cells, mutant EGFR expression, or EGFR blockade by cetuximab exerts a synthetic lethality effect and significantly compromises radiation resistance in hypoxic tumor cells. Mol Cancer Res; 15(11); 1503-16. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
47. Androgen Receptor Variants Mediate DNA Repair after Prostate Cancer Irradiation.
- Author
-
Yin Y, Li R, Xu K, Ding S, Li J, Baek G, Ramanand SG, Ding S, Liu Z, Gao Y, Kanchwala MS, Li X, Hutchinson R, Liu X, Woldu SL, Xing C, Desai NB, Feng FY, Burma S, de Bono JS, Dehm SM, Mani RS, Chen BPC, and Raj GV
- Subjects
- Animals, Antineoplastic Agents pharmacology, Benzamides, DNA Repair radiation effects, DNA-Activated Protein Kinase metabolism, Humans, Male, Mice, Mice, Nude, Nitriles, Phenylthiohydantoin analogs & derivatives, Phenylthiohydantoin pharmacology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms radiotherapy, Radiation, Ionizing, Receptors, Androgen chemistry, Receptors, Androgen metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Androgen Antagonists pharmacology, DNA Repair genetics, DNA-Activated Protein Kinase antagonists & inhibitors, Prostatic Neoplasms genetics, Protein Kinase Inhibitors pharmacology, Receptors, Androgen genetics
- Abstract
In prostate cancer, androgen deprivation therapy (ADT) enhances the cytotoxic effects of radiotherapy. This effect is associated with weakening of the DNA damage response (DDR) normally supported by the androgen receptor. As a significant number of patients will fail combined ADT and radiotherapy, we hypothesized that DDR may be driven by androgen receptor splice variants (ARV) induced by ADT. Investigating this hypothesis, we found that ARVs increase the clonogenic survival of prostate cancer cells after irradiation in an ADT-independent manner. Notably, prostate cancer cell irradiation triggers binding of ARV to the catalytic subunit of the critical DNA repair kinase DNA-PK. Pharmacologic inhibition of DNA-PKc blocked this interaction, increased DNA damage, and elevated prostate cancer cell death after irradiation. Our findings provide a mechanistic rationale for therapeutic targeting of DNA-PK in the context of combined ADT and radiotherapy as a strategy to radiosensitize clinically localized prostate cancer. Cancer Res; 77(18); 4745-54. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
48. A TNF-JNK-Axl-ERK signaling axis mediates primary resistance to EGFR inhibition in glioblastoma.
- Author
-
Guo G, Gong K, Ali S, Ali N, Shallwani S, Hatanpaa KJ, Pan E, Mickey B, Burma S, Wang DH, Kesari S, Sarkaria JN, Zhao D, and Habib AA
- Subjects
- Antineoplastic Agents pharmacology, Brain Neoplasms drug therapy, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Extracellular Signal-Regulated MAP Kinases drug effects, Glioblastoma drug therapy, Humans, Brain Neoplasms metabolism, ErbB Receptors metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Glioblastoma metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Signal Transduction drug effects
- Abstract
Aberrant epidermal growth factor receptor (EGFR) signaling is widespread in cancer, making the EGFR an important target for therapy. EGFR gene amplification and mutation are common in glioblastoma (GBM), but EGFR inhibition has not been effective in treating this tumor. Here we propose that primary resistance to EGFR inhibition in glioma cells results from a rapid compensatory response to EGFR inhibition that mediates cell survival. We show that in glioma cells expressing either EGFR wild type or the mutant EGFRvIII, EGFR inhibition triggers a rapid adaptive response driven by increased tumor necrosis factor (TNF) secretion, which leads to activation in turn of c-Jun N-terminal kinase (JNK), the Axl receptor tyrosine kinase and extracellular signal-regulated kinases (ERK). Inhibition of this adaptive axis at multiple nodes rendered glioma cells with primary resistance sensitive to EGFR inhibition. Our findings provide a possible explanation for the failures of anti-EGFR therapy in GBM and suggest a new approach to the treatment of EGFR-expressing GBM using a combination of EGFR and TNF inhibition.
- Published
- 2017
- Full Text
- View/download PDF
49. DNA-damage-induced degradation of EXO1 exonuclease limits DNA end resection to ensure accurate DNA repair.
- Author
-
Tomimatsu N, Mukherjee B, Harris JL, Boffo FL, Hardebeck MC, Potts PR, Khanna KK, and Burma S
- Subjects
- Amino Acid Motifs, Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, DNA Repair Enzymes genetics, Enzyme Activation physiology, Exodeoxyribonucleases genetics, HEK293 Cells, HeLa Cells, Humans, Phosphorylation physiology, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Chromosomal Instability physiology, DNA Damage, DNA Repair physiology, DNA Repair Enzymes metabolism, Exodeoxyribonucleases metabolism, Proteolysis, Ubiquitination physiology
- Abstract
End resection of DNA double-strand breaks (DSBs) to generate 3'-single-stranded DNA facilitates DSB repair via error-free homologous recombination (HR) while stymieing repair by the error-prone non-homologous end joining (NHEJ) pathway. Activation of DNA end resection involves phosphorylation of the 5' to 3' exonuclease EXO1 by the phosphoinositide 3-kinase-like kinases ATM (ataxia telangiectasia-mutated) and ATR (ATM and Rad3-related) and by the cyclin-dependent kinases 1 and 2. After activation, EXO1 must also be restrained to prevent over-resection that is known to hamper optimal HR and trigger global genomic instability. However, mechanisms by which EXO1 is restrained are still unclear. Here, we report that EXO1 is rapidly degraded by the ubiquitin-proteasome system soon after DSB induction in human cells. ATR inhibition attenuated DNA-damage-induced EXO1 degradation, indicating that ATR-mediated phosphorylation of EXO1 targets it for degradation. In accord with these results, EXO1 became resistant to degradation when its SQ motifs required for ATR-mediated phosphorylation were mutated. We show that upon the induction of DNA damage, EXO1 is ubiquitinated by a member of the Skp1-Cullin1-F-box (SCF) family of ubiquitin ligases in a phosphorylation-dependent manner. Importantly, expression of degradation-resistant EXO1 resulted in hyper-resection, which attenuated both NHEJ and HR and severely compromised DSB repair resulting in chromosomal instability. These findings indicate that the coupling of EXO1 activation with its eventual degradation is a timing mechanism that limits the extent of DNA end resection for accurate DNA repair., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
50. Enhanced dependency of KRAS-mutant colorectal cancer cells on RAD51-dependent homologous recombination repair identified from genetic interactions in Saccharomyces cerevisiae.
- Author
-
Kalimutho M, Bain AL, Mukherjee B, Nag P, Nanayakkara DM, Harten SK, Harris JL, Subramanian GN, Sinha D, Shirasawa S, Srihari S, Burma S, and Khanna KK
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, DNA Breaks, Double-Stranded, DNA Damage, DNA-Binding Proteins genetics, Dose-Response Relationship, Drug, ErbB Receptors genetics, HCT116 Cells, Humans, Mutation, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, RNA, Small Interfering genetics, Rad51 Recombinase metabolism, Transcription Factors genetics, Colorectal Neoplasms genetics, Homologous Recombination, Proto-Oncogene Proteins p21(ras) genetics, Rad51 Recombinase genetics, Saccharomyces cerevisiae genetics
- Abstract
Activating KRAS mutations drive colorectal cancer tumorigenesis and influence response to anti-EGFR-targeted therapy. Despite recent advances in understanding Ras signaling biology and the revolution in therapies for melanoma using BRAF inhibitors, no targeted agents have been effective in KRAS-mutant cancers, mainly due to activation of compensatory pathways. Here, by leveraging the largest synthetic lethal genetic interactome in yeast, we identify that KRAS-mutated colorectal cancer cells have augmented homologous recombination repair (HRR) signaling. We found that KRAS mutation resulted in slowing and stalling of the replication fork and accumulation of DNA damage. Moreover, we found that KRAS-mutant HCT116 cells have an increase in MYC-mediated RAD51 expression with a corresponding increase in RAD51 recruitment to irradiation-induced DNA double-strand breaks (DSBs) compared to genetically complemented isogenic cells. MYC depletion using RNA interference significantly reduced IR-induced RAD51 foci formation and HRR. On the contrary, overexpression of either HA-tagged wild-type (WT) MYC or phospho-mutant S62A increased RAD51 protein levels and hence IR-induced RAD51 foci. Likewise, depletion of RAD51 selectively induced apoptosis in HCT116-mutant cells by increasing DSBs. Pharmacological inhibition targeting HRR signaling combined with PARP inhibition selectivity killed KRAS-mutant cells. Interestingly, these differences were not seen in a second isogenic pair of KRAS WT and mutant cells (DLD-1), likely due to their nondependency on the KRAS mutation for survival. Our data thus highlight a possible mechanism by which KRAS-mutant-dependent cells drive HRR in vitro by upregulating MYC-RAD51 expression. These data may offer a promising therapeutic vulnerability in colorectal cancer cells harboring otherwise nondruggable KRAS mutations, which warrants further investigation in vivo., (© 2017 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.