319 results on '"George R Stark"'
Search Results
2. Multiple tumor suppressors regulate a HIF-dependent negative feedback loop via ISGF3 in human clear cell renal cancer
- Author
-
Lili Liao, Zongzhi Z Liu, Lauren Langbein, Weijia Cai, Eun-Ah Cho, Jie Na, Xiaohua Niu, Wei Jiang, Zhijiu Zhong, Wesley L Cai, Geetha Jagannathan, Essel Dulaimi, Joseph R Testa, Robert G Uzzo, Yuxin Wang, George R Stark, Jianxin Sun, Stephen Peiper, Yaomin Xu, Qin Yan, and Haifeng Yang
- Subjects
kidney cancer ,VHL ,PBRM1 ,KDM5C ,BAP1 ,ISGF3 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Whereas VHL inactivation is a primary event in clear cell renal cell carcinoma (ccRCC), the precise mechanism(s) of how this interacts with the secondary mutations in tumor suppressor genes, including PBRM1, KDM5C/JARID1C, SETD2, and/or BAP1, remains unclear. Gene expression analyses reveal that VHL, PBRM1, or KDM5C share a common regulation of interferon response expression signature. Loss of HIF2α, PBRM1, or KDM5C in VHL-/-cells reduces the expression of interferon stimulated gene factor 3 (ISGF3), a transcription factor that regulates the interferon signature. Moreover, loss of SETD2 or BAP1 also reduces the ISGF3 level. Finally, ISGF3 is strongly tumor-suppressive in a xenograft model as its loss significantly enhances tumor growth. Conversely, reactivation of ISGF3 retards tumor growth by PBRM1-deficient ccRCC cells. Thus after VHL inactivation, HIF induces ISGF3, which is reversed by the loss of secondary tumor suppressors, suggesting that this is a key negative feedback loop in ccRCC.
- Published
- 2018
- Full Text
- View/download PDF
3. Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma
- Author
-
Weiwei Tao, Chengwei Chu, Wenchao Zhou, Zhi Huang, Kui Zhai, Xiaoguang Fang, Qian Huang, Aili Zhang, Xiuxing Wang, Xingjiang Yu, Haidong Huang, Qiulian Wu, Andrew E. Sloan, Jennifer S. Yu, Xiaoxia Li, George R. Stark, Jeremy N. Rich, and Shideng Bao
- Subjects
Science - Abstract
The tumour microenvironment plays an important role in promoting glioblastoma. Here, the authors show that glioma stem cells secrete WISP1, which promotes both the survival of the stem cells and tumour-associated macrophages.
- Published
- 2020
- Full Text
- View/download PDF
4. Inflammation mobilizes copper metabolism to promote colon tumorigenesis via an IL-17-STEAP4-XIAP axis
- Author
-
Yun Liao, Junjie Zhao, Katarzyna Bulek, Fangqiang Tang, Xing Chen, Gang Cai, Shang Jia, Paul L. Fox, Emina Huang, Theresa T. Pizarro, Matthew F. Kalady, Mark W. Jackson, Shideng Bao, Ganes C. Sen, George R. Stark, Christopher J. Chang, and Xiaoxia Li
- Subjects
Science - Abstract
STEAP4 promotes the uptake of copper, and copper is known to be enhanced in cancer tissues. Here, the authors show that STEAP4 is induced by IL17, which is increased in inflamed tissues, consequently the increased copper levels activate NFκB signalling and suppression of apoptosis.
- Published
- 2020
- Full Text
- View/download PDF
5. Bazedoxifene inhibits sustained STAT3 activation and increases survival in GBM
- Author
-
Samantha M. Wightman, Tyler J. Alban, Xing Chen, Justin D. Lathia, Yuxin Wang, and George R. Stark
- Subjects
Glioblastoma ,IL6 ,STAT3 ,Bazedoxifene ,Glioma stem cells ,Cancer stem cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
An important factor correlated with poor survival in glioblastoma (GBM) is the aberrant and persistent activation of STAT3, a critical transcription factor that regulates multiple genes with key roles in cell survival, proliferation, resistance to chemotherapy, and stem cell maintenance. The Interleukin-6 (IL6)-STAT3 signaling axis has been studied extensively in inflammation and cancer. However, it is not completely understood how high levels of activated STAT3 are sustained in tumors. Previously, we identified a novel mechanism of biphasic activation of STAT3 in response to gp130-linked cytokines, including IL6, in which activation of STAT3 is prolonged by circumventing the negative regulatory mechanisms induced by its initial activationTo target prolonged STAT3 activation, we used the small molecule inhibitor bazedoxifene (BZA), which blocks formation of the IL6 receptor-gp130 complex. Glioma stem-like cells (GSCs) are more tumorigenic and more resistant to therapy. STAT3 is a key driver of the expression of stem cell transcription factors, making it a therapeutically important target in GBM. We show that treating GSCs with BZA decreases their self-renewal capacity and the expression of GSC markers in vitro. Additionally, BZA crosses the blood-brain barrier and confers a survival advantage in an orthotopic syngeneic mouse model of GBM. Although IL6-STAT3 signaling is important for GSC survival, a therapeutic agent that inhibits this pathway without toxicity has yet to be identified. Our findings reveal a mechanism of sustained STAT3 signaling in GBM and reveal its role in GSC maintenance, and we identify BZA as a novel candidate for treating GBM.
- Published
- 2021
- Full Text
- View/download PDF
6. The opposing effects of interferon-beta and oncostatin-M as regulators of cancer stem cell plasticity in triple-negative breast cancer
- Author
-
Mary R. Doherty, Jenny G. Parvani, Ilaria Tamagno, Damian J. Junk, Benjamin L. Bryson, Hyeon Joo Cheon, George R. Stark, and Mark W. Jackson
- Subjects
Interferon-beta ,Oncostatin-M ,SNAIL ,Triple-negative breast cancer ,Tumor microenvironment ,Cancer stem cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Highly aggressive, metastatic and therapeutically resistant triple-negative breast cancers (TNBCs) are often enriched for cancer stem cells (CSC). Cytokines within the breast tumor microenvironment (TME) influence the CSC state by regulating tumor cell differentiation programs. Two prevalent breast TME cytokines are oncostatin-M (OSM) and interferon-β (IFN-β). OSM is a member of the IL-6 family of cytokines and can drive the de-differentiation of TNBC cells to a highly aggressive CSC state. Conversely, IFN-β induces the differentiation of TNBC, resulting in the repression of CSC properties. Here, we assess how these breast TME cytokines influence CSC plasticity and clinical outcome. Methods Using transformed human mammary epithelial cell (HMEC) and TNBC cell models, we assessed the CSC markers and properties following exposure to OSM and/or IFN-β. CSC markers included CD24, CD44, and SNAIL; CSC properties included tumor sphere formation, migratory capacity, and tumor initiation. Results There are three major findings from our study. First, exposure of purified, non-CSC to IFN-β prevents OSM-mediated CD44 and SNAIL expression and represses tumor sphere formation and migratory capacity. Second, during OSM-induced de-differentiation, OSM represses endogenous IFN-β mRNA expression and autocrine/paracrine IFN-β signaling. Restoring IFN-β signaling to OSM-driven CSC re-engages IFN-β-mediated differentiation by repressing OSM/STAT3/SMAD3-mediated SNAIL expression, tumor initiation, and growth. Finally, the therapeutic use of IFN-β to treat OSM-driven tumors significantly suppresses tumor growth. Conclusions Our findings suggest that the levels of IFN-β and OSM in TNBC dictate the abundance of cells with a CSC phenotype. Indeed, TNBCs with elevated IFN-β signaling have repressed CSC properties and a better clinical outcome. Conversely, TNBCs with elevated OSM signaling have a worse clinical outcome. Likewise, since OSM suppresses IFN-β expression and signaling, our studies suggest that strategies to limit OSM signaling or activate IFN-β signaling will disengage the de-differentiation programs responsible for the aggressiveness of TNBCs.
- Published
- 2019
- Full Text
- View/download PDF
7. STAT2 hinders STING intracellular trafficking and reshapes its activation in response to DNA damage
- Author
-
Chenyao Wang, Jing Nan, Elise Holvey-Bates, Xing Chen, Samantha Wightman, Muhammad-Bilal Latif, Junjie Zhao, Xiaoxia Li, Ganes C. Sen, George R. Stark, and Yuxin Wang
- Subjects
Multidisciplinary - Abstract
In cancer cells, endogenous or therapy-induced DNA damage leads to the abnormal presence of DNA in the cytoplasm, which triggers the activation of cGAS (cyclic GMP–AMP synthase) and STING (stimulator of interferon genes). STAT2 suppresses the cGAMP-induced expression of IRF3-dependent genes by binding to STING, blocking its intracellular trafficking, which is essential for the full response to STING activation. STAT2 reshapes STING signaling by inhibiting the induction of IRF3-dependent, but not NF-κB–dependent genes. This noncanonical activity of STAT2 is regulated independently of its tyrosine phosphorylation but does depend on the phosphorylation of threonine 404, which promotes the formation of a STAT2:STING complex that keeps STING bound to the endoplasmic reticulum (ER) and increases resistance to DNA damage. We conclude that STAT2 is a key negative intracellular regulator of STING, a function that is quite distinct from its function as a transcription factor.
- Published
- 2023
- Full Text
- View/download PDF
8. Data Supplement from Quinacrine Overcomes Resistance to Erlotinib by Inhibiting FACT, NF-κB, and Cell-Cycle Progression in Non–Small Cell Lung Cancer
- Author
-
George R. Stark, Neelesh Sharma, Goutham Narla, Sarmishtha De, Afshin Dowlati, John Pink, Katerina Gurova, and Josephine Kam Tai Dermawan
- Abstract
Legends for Supplementary Tables 1,2 and Figures S1-S3
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Quinacrine Overcomes Resistance to Erlotinib by Inhibiting FACT, NF-κB, and Cell-Cycle Progression in Non–Small Cell Lung Cancer
- Author
-
George R. Stark, Neelesh Sharma, Goutham Narla, Sarmishtha De, Afshin Dowlati, John Pink, Katerina Gurova, and Josephine Kam Tai Dermawan
- Abstract
Erlotinib is a tyrosine kinase inhibitor approved for the treatment of patients with advanced non–small cell lung cancer (NSCLC). In these patients, erlotinib prolongs survival but its benefit remains modest because many tumors express wild-type (wt) EGFR or develop a second-site EGFR mutation. To test drug combinations that could improve the efficacy of erlotinib, we combined erlotinib with quinacrine, which inhibits the FACT (facilitates chromatin transcription) complex that is required for NF-κB transcriptional activity. In A549 (wtEGFR), H1975 (EGFR-L858R/T790M), and H1993 (MET amplification) NSCLC cells, this drug combination was highly synergistic, as quantified by Chou–Talalay combination indices, and slowed xenograft tumor growth. At a sub-IC50 but more clinically attainable concentration of erlotinib, quinacrine, alone or in combination with erlotinib, significantly inhibited colony formation and induced cell-cycle arrest and apoptosis. Quinacrine decreased the level of active FACT subunit SSRP1 and suppressed NF-κB–dependent luciferase activity. Knockdown of SSRP1 decreased cell growth and sensitized cells to erlotinib. Moreover, transcriptomic profiling showed that quinacrine or combination treatment significantly affected cell-cycle–related genes that contain binding sites for transcription factors that regulate SSRP1 target genes. As potential biomarkers of drug combination efficacy, we identified genes that were more strongly suppressed by the combination than by single treatment, and whose increased expression predicted poorer survival in patients with lung adenocarcinoma. This preclinical study shows that quinacrine overcomes erlotinib resistance by inhibiting FACT and cell-cycle progression, and supports a clinical trial testing erlotinib alone versus this combination in advanced NSCLC. Mol Cancer Ther; 13(9); 2203–14. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Tables from IFNL4-ΔG Allele Is Associated with an Interferon Signature in Tumors and Survival of African-American Men with Prostate Cancer
- Author
-
Stefan Ambs, Ludmila Prokunina-Olsson, Eric A. Klein, George R. Stark, Robert H. Silverman, Robert M. Stephens, Christopher A. Loffredo, Symone V. Jordan, Adeola Obajemu, Olusegun O. Onabajo, Tiffany H. Dorsey, Cristina Magi-Galluzzi, Ming Yi, Tiffany A. Wallace, and Wei Tang
- Abstract
Supplementary Table 1. Characteristics of the African-American prostate cancer patients in the Cleveland Clinic Cohort according to overall survival; Supplementary Table 2. IRDS Probesets (n=49) for the Affymetrix U133A 2.0 array
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures from IFNL4-ΔG Allele Is Associated with an Interferon Signature in Tumors and Survival of African-American Men with Prostate Cancer
- Author
-
Stefan Ambs, Ludmila Prokunina-Olsson, Eric A. Klein, George R. Stark, Robert H. Silverman, Robert M. Stephens, Christopher A. Loffredo, Symone V. Jordan, Adeola Obajemu, Olusegun O. Onabajo, Tiffany H. Dorsey, Cristina Magi-Galluzzi, Ming Yi, Tiffany A. Wallace, and Wei Tang
- Abstract
Supplementary Figure 1. Expression of IFNL4 after transfection of human prostate cancer cell lines with the IFNL4-Halo construct; Supplementary Figure 2. Expression of two interferon signatures, IRG and IRDS, in cultured prostate cancer epithelial cells from 14 African-American and 13 European-American men; Supplementary Figure 3. Association of IFNL4 SNP rs12979860-T allele with decreased overall survival among African-American prostate cancer patients in the Cleveland Clinic cohort (n = 197).
- Published
- 2023
- Full Text
- View/download PDF
12. Figure S4 from The FACT inhibitor CBL0137 Synergizes with Cisplatin in Small-Cell Lung Cancer by Increasing NOTCH1 Expression and Targeting Tumor-Initiating Cells
- Author
-
George R. Stark, Afshin Dowlati, Gary Wildey, Claire J. Coleman, Daniel J. Lindner, and Sarmishtha De
- Abstract
Supplementary Fig S4. FACT is involved in the expression of regulatory stem cell transcription factors in TICs. A and B, CD44high cells derived from H446 and CD133high cells derived from H526 were treated with 500 nM (H446) or 1µM (H526) CBL0137 for 24 h. The mRNA levels of SOX2 were analyzed by qPCR (A), and SOX2 protein levels were determined by Western analysis (B).
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Pharmacological Targeting of the Histone Chaperone Complex FACT Preferentially Eliminates Glioblastoma Stem Cells and Prolongs Survival in Preclinical Models
- Author
-
Monica Venere, George R. Stark, Justin D. Lathia, Jeremy N. Rich, Katerina V. Gurova, Andrei A. Purmal, Andrew E. Sloan, Poorva Sandlesh, Qiulian Wu, Daniel J. Silver, Masahiro Hitomi, and Josephine Kam Tai Dermawan
- Abstract
The nearly universal recurrence of glioblastoma (GBM) is driven in part by a treatment-resistant subpopulation of GBM stem cells (GSC). To identify improved therapeutic possibilities, we combined the EGFR/HER2 inhibitor lapatinib with a novel small molecule, CBL0137, which inhibits FACT (facilitates chromatin transcription), a histone chaperone complex predominantly expressed in undifferentiated cells. Lapatinib and CBL0137 synergistically inhibited the proliferation of patient-derived GBM cells. Compared with non–stem tumor cells (NSTC) enriched from the same specimens, the GSCs were extremely sensitive to CBL0137 monotherapy or FACT knockdown. FACT expression was elevated in GSCs compared with matched NSTCs and decreased in GSCs upon differentiation. Acute exposure of GSCs to CBL0137 increased asymmetric cell division, decreased GSC marker expression, and decreased the capacity of GSCs to form tumor spheres in vitro and to initiate tumors in vivo. Oral administration of CBL0137 to mice bearing orthotopic GBM prolonged their survival. Knockdown of FACT reduced the expression of genes encoding several core stem cell transcription factors (SOX2, OCT4, NANOG, and OLIG2), and FACT occupied the promoters of these genes. FACT expression was elevated in GBM tumors compared with non-neoplastic brain tissues, portended a worse prognosis, and positively correlated with GSC markers and stem cell gene expression signatures. Preferential targeting of GSCs by CBL0137 and synergy with EGFR inhibitors support the development of clinical trials combining these two agents in GBM. Cancer Res; 76(8); 2432–42. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure S1 from Pharmacological Targeting of the Histone Chaperone Complex FACT Preferentially Eliminates Glioblastoma Stem Cells and Prolongs Survival in Preclinical Models
- Author
-
Monica Venere, George R. Stark, Justin D. Lathia, Jeremy N. Rich, Katerina V. Gurova, Andrei A. Purmal, Andrew E. Sloan, Poorva Sandlesh, Qiulian Wu, Daniel J. Silver, Masahiro Hitomi, and Josephine Kam Tai Dermawan
- Abstract
CBL0137 accumulates in brain tissues.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplemental Materials & Methods from Pharmacological Targeting of the Histone Chaperone Complex FACT Preferentially Eliminates Glioblastoma Stem Cells and Prolongs Survival in Preclinical Models
- Author
-
Monica Venere, George R. Stark, Justin D. Lathia, Jeremy N. Rich, Katerina V. Gurova, Andrei A. Purmal, Andrew E. Sloan, Poorva Sandlesh, Qiulian Wu, Daniel J. Silver, Masahiro Hitomi, and Josephine Kam Tai Dermawan
- Abstract
This file contains detailed materials & methods in addition to the materials & methods section in the main text.
- Published
- 2023
- Full Text
- View/download PDF
16. Data from IFNL4-ΔG Allele Is Associated with an Interferon Signature in Tumors and Survival of African-American Men with Prostate Cancer
- Author
-
Stefan Ambs, Ludmila Prokunina-Olsson, Eric A. Klein, George R. Stark, Robert H. Silverman, Robert M. Stephens, Christopher A. Loffredo, Symone V. Jordan, Adeola Obajemu, Olusegun O. Onabajo, Tiffany H. Dorsey, Cristina Magi-Galluzzi, Ming Yi, Tiffany A. Wallace, and Wei Tang
- Abstract
Purpose: Men of African ancestry experience an excessive prostate cancer mortality that could be related to an aggressive tumor biology. We previously described an immune-inflammation signature in prostate tumors of African-American (AA) patients. Here, we further deconstructed this signature and investigated its relationships with tumor biology, survival, and a common germline variant in the IFNλ4 (IFNL4) gene.Experimental Design: We analyzed gene expression in prostate tissue datasets and performed genotype and survival analyses. We also overexpressed IFNL4 in human prostate cancer cells.Results: We found that a distinct interferon (IFN) signature that is analogous to the previously described “IFN-related DNA damage resistance signature” (IRDS) occurs in prostate tumors. Evaluation of two independent patient cohorts revealed that IRDS is detected about twice as often in prostate tumors of AA than European-American men. Furthermore, analysis in TCGA showed an association of increased IRDS in prostate tumors with decreased disease-free survival. To explain these observations, we assessed whether IRDS is associated with an IFNL4 germline variant (rs368234815-ΔG) that controls production of IFNλ4, a type III IFN, and is most common in individuals of African ancestry. We show that the IFNL4 rs368234815-ΔG allele was significantly associated with IRDS in prostate tumors and overall survival of AA patients. Moreover, IFNL4 overexpression induced IRDS in three human prostate cancer cell lines.Conclusions: Our study links a germline variant that controls production of IFNλ4 to the occurrence of a clinically relevant IFN signature in prostate tumors that may predominantly affect men of African ancestry. Clin Cancer Res; 24(21); 5471–81. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental Figure Legends from Pharmacological Targeting of the Histone Chaperone Complex FACT Preferentially Eliminates Glioblastoma Stem Cells and Prolongs Survival in Preclinical Models
- Author
-
Monica Venere, George R. Stark, Justin D. Lathia, Jeremy N. Rich, Katerina V. Gurova, Andrei A. Purmal, Andrew E. Sloan, Poorva Sandlesh, Qiulian Wu, Daniel J. Silver, Masahiro Hitomi, and Josephine Kam Tai Dermawan
- Abstract
This file contains figure legends to supplemental figures S1-S5.
- Published
- 2023
- Full Text
- View/download PDF
18. Data from The FACT inhibitor CBL0137 Synergizes with Cisplatin in Small-Cell Lung Cancer by Increasing NOTCH1 Expression and Targeting Tumor-Initiating Cells
- Author
-
George R. Stark, Afshin Dowlati, Gary Wildey, Claire J. Coleman, Daniel J. Lindner, and Sarmishtha De
- Abstract
Traditional treatments of small-cell lung cancer (SCLC) with cisplatin, a standard-of-care therapy, spare the tumor-initiating cells (TIC) that mediate drug resistance. Here we report a novel therapeutic strategy that preferentially targets TICs in SCLC, in which cisplatin is combined with CBL0137, an inhibitor of the histone chaperone facilitates chromatin transcription (FACT), which is highly expressed in TICs. Combination of cisplatin and CBL0137 killed patient-derived and murine SCLC cell lines synergistically. In response to CBL0137 alone, TICs were more sensitive than non-TICs, in part, because CBL0137 increased expression of the tumor suppressor NOTCH1 by abrogating the binding of negative regulator SP3 to the NOTCH1 promoter, and in part because treatment decreased the high expression of stem cell transcription factors. The combination of cisplatin and CBL0137 greatly reduced the growth of a patient-derived xenograft in mice and also the growth of a syngeneic mouse SCLC tumor. Thus, CBL0137 can be a highly effective drug against SCLC, especially in combination with cisplatin.Significance: These findings reveal a novel therapeutic regimen for SCLC, combining cisplatin with an inhibitor that preferentially targets tumor-initiating cells. Cancer Res; 78(9); 2396–406. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Table 1 from NF-κB: Regulation by Methylation
- Author
-
George R. Stark and Tao Lu
- Abstract
Methylation of NF-κB, genetic alteration of PRMT5 and hypothetical model.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 4 from p53-Mediated Growth Suppression in Response to Nutlin-3 in Cyclin D1–Transformed Cells Occurs Independently of p21
- Author
-
Mark W. Jackson, George R. Stark, John T. Patton, and Charlene E. Kan
- Abstract
Supplementary Figure 4 from p53-Mediated Growth Suppression in Response to Nutlin-3 in Cyclin D1–Transformed Cells Occurs Independently of p21
- Published
- 2023
- Full Text
- View/download PDF
21. Data from p53-Mediated Growth Suppression in Response to Nutlin-3 in Cyclin D1–Transformed Cells Occurs Independently of p21
- Author
-
Mark W. Jackson, George R. Stark, John T. Patton, and Charlene E. Kan
- Abstract
Interaction of cyclin D1 with cyclin-dependent kinases (CDK) results in the hyperphosphorylation of the RB family of proteins, thereby inactivating the tumor-suppressive function of RB. Our previous findings suggest that constitutive cyclin D1/CDK activity inhibits p53-mediated gene repression by preventing the appropriate regulation of CDK activity by the CDK inhibitor p21, a transcriptional target of p53. To study the role of cyclin D1 in driving human mammary cell transformation, we expressed a constitutively active cyclin D1–CDK fusion protein (D1/CDK) in immortalized human mammary epithelial cells. D1/CDK-expressing human mammary epithelial cells grew anchorage-independently in the presence of wild-type p53, consistent with the idea that D1/CDK disrupts downstream p53 signaling. Using this transformation model, we examined the sensitivity of the D1/CDK-expressing cells to Nutlin-3, an HDM2 antagonist that activates p53. Surprisingly, treatment of D1/CDK-transformed cells with Nutlin-3 prevented their anchorage-independent growth. The Nutlin-3–induced growth arrest was enforced in D1/CDK-expressing cells despite the presence of hyperphosphorylated RB implicating a p53-dependent, RB-independent mechanism for growth suppression. Further analysis identified that CDC2 and cyclin B1, key cell cycle regulators, were stably down-regulated following p53 stabilization by Nutlin-3, consistent with direct interaction between p53 and the CDC2 and cyclin B1 promoters, leading to the repression of transcription by methylation. In contrast to D1/CDK expression, direct inactivation of p53 resulted in no repression of CDC2 and no cell cycle arrest. We conclude that induction of p53 by Nutlin-3 is a viable therapeutic strategy in cancers with constitutive CDK signaling due to the direct repression of specific p53 target genes. [Cancer Res 2007;67(20):9862–8]
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure Legends 1-4 from p53-Mediated Growth Suppression in Response to Nutlin-3 in Cyclin D1–Transformed Cells Occurs Independently of p21
- Author
-
Mark W. Jackson, George R. Stark, John T. Patton, and Charlene E. Kan
- Abstract
Supplementary Figure Legends 1-4 from p53-Mediated Growth Suppression in Response to Nutlin-3 in Cyclin D1–Transformed Cells Occurs Independently of p21
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure 3 from p53-Mediated Growth Suppression in Response to Nutlin-3 in Cyclin D1–Transformed Cells Occurs Independently of p21
- Author
-
Mark W. Jackson, George R. Stark, John T. Patton, and Charlene E. Kan
- Abstract
Supplementary Figure 3 from p53-Mediated Growth Suppression in Response to Nutlin-3 in Cyclin D1–Transformed Cells Occurs Independently of p21
- Published
- 2023
- Full Text
- View/download PDF
24. Pharmacological inhibition of BACE1 suppresses glioblastoma growth by stimulating macrophage phagocytosis of tumor cells
- Author
-
Aili Zhang, Weiwei Tao, George R. Stark, Qian Huang, Xiaoguang Fang, Zhi Huang, Shideng Bao, Kui Zhai, Thomas A. Hamilton, and Xiaoxia Li
- Subjects
Cancer Research ,biology ,Amyloid ,Activator (genetics) ,Chemistry ,Macrophages ,Phagocytosis ,medicine.disease ,Oncology ,Glioma ,medicine ,biology.protein ,Cancer research ,Aspartic Acid Endopeptidases ,Humans ,Macrophage ,Amyloid Precursor Protein Secretases ,Glioblastoma ,Receptor ,STAT3 ,Reprogramming - Abstract
Glioblastoma (GBM) contains abundant tumor-associated macrophages (TAMs). The majority of TAMs are tumor-promoting macrophages (pTAMs), while tumor-suppressive macrophages (sTAMs) are the minority. Thus, reprogramming pTAMs into sTAMs represents an attractive therapeutic strategy. By screening a collection of small-molecule compounds, we find that inhibiting β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) with MK-8931 potently reprograms pTAMs into sTAMs and promotes macrophage phagocytosis of glioma cells; moreover, low-dose radiation markedly enhances TAM infiltration and synergizes with MK-8931 treatment to suppress malignant growth. BACE1 is preferentially expressed by pTAMs in human GBMs and is required to maintain pTAM polarization through trans-interleukin 6 (IL-6)–soluble IL-6 receptor (sIL-6R)–signal transducer and activator of transcription 3 (STAT3) signaling. Because MK-8931 and other BACE1 inhibitors have been developed for Alzheimer’s disease and have been shown to be safe for humans in clinical trials, these inhibitors could potentially be streamlined for cancer therapy. Collectively, this study offers a promising therapeutic approach to enhance macrophage-based therapy for malignant tumors. Zhai et al. identify clinical brain-penetrant BACE1 inhibitors as regulators of macrophage-dependent phagocytosis in glioblastoma through IL-6–STAT3 signaling and demonstrate preclinical therapeutic efficacy in orthotopic mouse models and PDXs.
- Published
- 2021
- Full Text
- View/download PDF
25. Cooperative Transcriptional Activation of Antimicrobial Genes by STAT and NF-κB Pathways by Concerted Recruitment of the Mediator Complex
- Author
-
Sebastian Wienerroither, Priyank Shukla, Matthias Farlik, Andrea Majoros, Bernadette Stych, Claus Vogl, HyeonJoo Cheon, George R. Stark, Birgit Strobl, Mathias Müller, and Thomas Decker
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The transcriptional response to infection with the bacterium Listeria monocytogenes (Lm) requires cooperative signals of the type I interferon (IFN-I)-stimulated JAK-STAT and proinflammatory NF-κB pathways. Using ChIP-seq analysis, we define genes induced in Lm-infected macrophages through synergistic transcriptional activation by NF-κB and the IFN-I-activated transcription factor ISGF3. Using the Nos2 and IL6 genes as prime examples of this group, we show that NF-κB functions to recruit enzymes that establish histone marks of transcriptionally active genes. In addition, NF-κB regulates transcriptional elongation by employing the mediator kinase module for the recruitment of the pTEFb complex. ISGF3 has a major role in associating the core mediator with the transcription start as a prerequisite for TFIID and RNA polymerase II (Pol II) binding. Our data suggest that the functional cooperation between two major antimicrobial pathways is based on promoter priming by NF-κB and the engagement of the core mediator for Pol II binding by ISGF3.
- Published
- 2015
- Full Text
- View/download PDF
26. How cancer cells make and respond to interferon-I
- Author
-
HyeonJoo Cheon, Yuxin Wang, Samantha M. Wightman, Mark W. Jackson, and George R. Stark
- Subjects
Cancer Research ,Oncology ,Neoplasms ,Humans ,Tyrosine ,Interferons ,Interferon-Stimulated Gene Factor 3 ,Article ,Interferon-Stimulated Gene Factor 3, gamma Subunit ,Signal Transduction - Abstract
Acute exposure of cancer cells to high concentrations of type I interferon (IFN-I) drives growth arrest and apoptosis, whereas chronic exposure to low concentrations provides important prosurvival advantages. Tyrosine-phosphorylated IFN-stimulated gene (ISG) factor 3 (ISGF3) drives acute deleterious responses to IFN-I, whereas unphosphorylated (U-)ISGF3, lacking tyrosine phosphorylation, drives essential constitutive prosurvival mechanisms. Surprisingly, programmed cell death-ligand 1 (PD-L1), often expressed on the surfaces of tumor cells and well recognized for its importance in inactivating cytotoxic T cells, also has important cell-intrinsic protumor activities, including dampening acute responses to cytotoxic high levels of IFN-I and sustaining the expression of the low levels that benefit tumors. More thorough understanding of the newly recognized complex roles of IFN-I in cancer may lead to the identification of novel therapeutic strategies.
- Published
- 2022
27. Activated Protein Phosphatase 2A Disrupts Nutrient Sensing Balance Between Mechanistic Target of Rapamycin Complex 1 and Adenosine Monophosphate–Activated Protein Kinase, Causing Sarcopenia in Alcohol‐Associated Liver Disease
- Author
-
Gangarao Davuluri, Maradumane L. Mohan, George R. Stark, Rebecca L. McCullough, Samjhana Thapaliya, Srinivasan Dasarathy, Laura E. Nagy, Megan R. McMullen, Sathyamangla V. Naga Prasad, Jinendiran Sekar, Mahesha Gangadhariah, Nicole Welch, McKenzie Stine, Sashi Kant, Avinash Kumar, and Khaled Alsabbagh Alchirazi
- Subjects
Male ,0301 basic medicine ,Sarcopenia ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Myoblasts ,Mice ,03 medical and health sciences ,0302 clinical medicine ,AMP-Activated Protein Kinase Kinases ,Animals ,Homeostasis ,Humans ,Immunoprecipitation ,Protein Phosphatase 2 ,Protein kinase A ,Liver Diseases, Alcoholic ,Protein kinase B ,Mice, Knockout ,Hepatology ,Kinase ,Chemistry ,Autophagy ,AMPK ,Protein phosphatase 2 ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Phosphorylation ,Female ,030211 gastroenterology & hepatology - Abstract
BACKGROUND AND AIMS Despite the high clinical significance of sarcopenia in alcohol-associated cirrhosis, there are currently no effective therapies because the underlying mechanisms are poorly understood. We determined the mechanisms of ethanol-induced impaired phosphorylation of mechanistic target of rapamycin complex 1 (mTORC1) and adenosine monophosphate-activated protein kinase (AMPK) with consequent dysregulated skeletal muscle protein homeostasis (balance between protein synthesis and breakdown). APPROACH AND RESULTS Differentiated murine myotubes, gastrocnemius muscle from mice with loss and gain of function of regulatory genes following ethanol treatment, and skeletal muscle from patients with alcohol-associated cirrhosis were used. Ethanol increases skeletal muscle autophagy by dephosphorylating mTORC1, circumventing the classical kinase regulation by protein kinase B (Akt). Concurrently and paradoxically, ethanol exposure results in dephosphorylation and inhibition of AMPK, an activator of autophagy and inhibitor of mTORC1 signaling. However, AMPK remains inactive with ethanol exposure despite lower cellular and tissue adenosine triphosphate, indicating a "pseudofed" state. We identified protein phosphatase (PP) 2A as a key mediator of ethanol-induced signaling and functional perturbations using loss and gain of function studies. Ethanol impairs binding of endogenous inhibitor of PP2A to PP2A, resulting in methylation and targeting of PP2A to cause dephosphorylation of mTORC1 and AMPK. Activity of phosphoinositide 3-kinase-γ (PI3Kγ), a negative regulator of PP2A, was decreased in response to ethanol. Ethanol-induced molecular and phenotypic perturbations in wild-type mice were observed in PI3Kγ-/- mice even at baseline. Importantly, overexpressing kinase-active PI3Kγ but not the kinase-dead mutant reversed ethanol-induced molecular perturbations. CONCLUSIONS Our study describes the mechanistic underpinnings for ethanol-mediated dysregulation of protein homeostasis by PP2A that leads to sarcopenia with a potential for therapeutic approaches by targeting the PI3Kγ-PP2A axis.
- Published
- 2021
- Full Text
- View/download PDF
28. The JAK-STAT pathway at 30: Much learned, much more to do
- Author
-
Rachael L. Philips, Yuxin Wang, HyeonJoo Cheon, Yuka Kanno, Massimo Gadina, Vittorio Sartorelli, Curt M. Horvath, James E. Darnell, George R. Stark, and John J. O’Shea
- Subjects
Mammals ,STAT Transcription Factors ,Animals ,COVID-19 ,Cytokines ,Humans ,Interferons ,General Biochemistry, Genetics and Molecular Biology ,Article ,Janus Kinases ,Signal Transduction - Abstract
The discovery of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway arose from investigations of how cells respond to interferons (IFNs), revealing a paradigm in cell signaling conserved from slime molds to mammals. These discoveries revealed mechanisms underlying rapid gene expression mediated by a wide variety of extracellular polypeptides including cytokines, interleukins, and related factors. This knowledge has provided numerous insights into human disease, from immune deficiencies to cancer, and was rapidly translated to new drugs for autoimmune, allergic, and infectious diseases, including COVID-19. Despite these advances, major challenges and opportunities remain.
- Published
- 2022
29. Type I interferon signaling facilitates resolution of acute liver injury by priming macrophage polarization
- Author
-
Qiaoling Song, Shyamasree Datta, Xue Liang, Xiaohan Xu, Paul Pavicic, Xiaonan Zhang, Yuanyuan Zhao, Shan Liu, Jun Zhao, Yuting Xu, Jing Xu, Lihong Wu, Zhihua Wu, Minghui Zhang, Zhan Zhao, Chunhua Lin, Yuxin Wang, Peng Han, Peng Jiang, Yating Qin, Wei Li, Yingying Zhang, Yonglun Luo, Ganes Sen, George R. Stark, Chenyang Zhao, Thomas Hamilton, and Jinbo Yang
- Subjects
Infectious Diseases ,Immunology ,Immunology and Allergy - Abstract
Due to their broad functional plasticity, myeloid cells contribute to both liver injury and recovery during acetaminophen overdose-induced acute liver injury (APAP-ALI). A comprehensive understanding of cellular diversity and intercellular crosstalk is essential to elucidate the mechanisms and to develop therapeutic strategies for APAP-ALI treatment. Here, we identified the function of IFN-I in the myeloid compartment during APAP-ALI. Utilizing single-cell RNA sequencing, we characterized the cellular atlas and dynamic progression of liver CD11b+ cells post APAP-ALI in WT and STAT2 T403A mice, which was further validated by immunofluorescence staining, bulk RNA-seq, and functional experiments in vitro and in vivo. We identified IFN-I-dependent transcriptional programs in a three-way communication pathway that involved IFN-I synthesis in intermediate restorative macrophages, leading to CSF-1 production in aging neutrophils that ultimately enabled Trem2+ restorative macrophage maturation, contributing to efficient liver repair. Overall, we uncovered the heterogeneity of hepatic myeloid cells in APAP-ALI at single-cell resolution and the therapeutic potential of IFN-I in the treatment of APAP-ALI.
- Published
- 2022
30. Germline PTEN mutations are associated with a skewed peripheral immune repertoire in humans and mice
- Author
-
Matthew G. Loya, Charis Eng, Alexander T. King, Nicholas Sarn, George R. Stark, Ritika Jaini, Stetson Thacker, and Qi Yu
- Subjects
Male ,0301 basic medicine ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,Biology ,010402 general chemistry ,medicine.disease_cause ,01 natural sciences ,Germline ,Immune tolerance ,Mice ,03 medical and health sciences ,Germline mutation ,Immune Tolerance ,Genetics ,medicine ,Animals ,Humans ,Tensin ,PTEN ,Gene Knock-In Techniques ,Molecular Biology ,Germ-Line Mutation ,Genetics (clinical) ,Cell Proliferation ,B-Lymphocytes ,Mutation ,Toll-Like Receptors ,PTEN Phosphohydrolase ,General Medicine ,Autoimmune regulator ,Phenotype ,0104 chemical sciences ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,biology.protein ,Female ,General Article ,Hamartoma Syndrome, Multiple ,Transcription Factors - Abstract
Individuals with germline mutations in the gene encoding phosphatase and tensin homolog on chromosome ten (PTEN) are diagnosed with PTEN hamartoma tumor syndrome (PHTS) and are at high risk for developing breast, thyroid and other cancers and/or autoimmunity or neurodevelopmental issues including autism spectrum disorders. Although well recognized as a tumor suppressor, involvement of PTEN mutations in mediating such a diverse range of phenotypes indicates a more central involvement for PTEN in immunity than previously recognized. To address this, sequencing of the T-cell receptor variable-region β-chain was performed on peripheral blood from PHTS patients. Based on patient findings, we performed mechanistic studies in two Pten knock-in murine models, distinct from each other in cell compartment-specific predominance of Pten. We found that PTEN mutations in humans and mice are associated with a skewed T- and B-cell gene repertoire, characterized by increased prevalence of high-frequency clones. Immunological characterization showed that Pten mutants have increased B-cell proliferation and a proclivity towards increased T-cell reactivity upon Toll-like-receptor stimulation. Furthermore, decreases in nuclear but not cytoplasmic Pten levels associated with a reduction in expression of the autoimmune regulator (Aire), a critical mediator of central immune tolerance. Mechanistically, we show that nuclear PTEN most likely regulates Aire expression via its emerging role in splicing regulation. We conclude that germline disruption of PTEN, both in human and mouse, results in compromised central immune tolerance processes that may significantly impact individual stress responses and therefore predisposition to autoimmunity and cancer.
- Published
- 2020
- Full Text
- View/download PDF
31. Loss of ZIP facilitates JAK2-STAT3 activation in tamoxifen-resistant breast cancer
- Author
-
Rui Zhao, Jing Zhang, Lei Shi, Ning Zhu, Aihong Mao, Yuxin Wang, Jing Nan, Yuping Du, Xing Chen, Zhao Zhang, Xinxin Zhang, Yuxi Lin, Jinbo Yang, George R. Stark, Wei Wei, Jingjie Sun, Ruidong Miao, Xin Li, and Xiaodong Qin
- Subjects
STAT3 Transcription Factor ,tamoxifen resistance ,Estrogen receptor ,Breast Neoplasms ,Mice, SCID ,Mice ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,STAT3 ,skin and connective tissue diseases ,Multidisciplinary ,biology ,VBIM ,JAK-STAT signaling pathway ,Cell Biology ,Biological Sciences ,Janus Kinase 2 ,medicine.disease ,Phenotype ,JAK/STAT ,Death-Associated Protein Kinases ,Tamoxifen ,Receptors, Estrogen ,Drug Resistance, Neoplasm ,biology.protein ,Cancer research ,Phosphorylation ,Female ,Histone deacetylase ,ZIP ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Significance Tamoxifen is beneficial in treating estrogen receptor–positive breast cancer, but resistance to this treatment eventually ensues. A method to identify mechanisms of tamoxifen resistance identified the histone deacetylase ZIP, leading to the finding that increased expression of the tyrosine kinase JAK2 is one important factor. As a result of this discovery, it may be possible to use an inhibitor of JAK2 to block the aberrant activation of STAT3 caused by ZIP deficiency to help overcome or prevent tamoxifen resistance., Tamoxifen, a widely used modulator of the estrogen receptor (ER), targets ER-positive breast cancer preferentially. We used a powerful validation-based insertion mutagenesis method to find that expression of a dominant-negative, truncated form of the histone deacetylase ZIP led to resistance to tamoxifen. Consistently, increased expression of full-length ZIP gives the opposite phenotype, inhibiting the expression of genes whose products mediate resistance. An important example is JAK2. By binding to two specific sequences in the promoter, ZIP suppresses JAK2 expression. Increased expression and activation of JAK2 when ZIP is inhibited lead to increased STAT3 phosphorylation and increased resistance to tamoxifen, both in cell culture experiments and in a mouse xenograft model. Furthermore, data from human tumors are consistent with the conclusion that decreased expression of ZIP leads to resistance to tamoxifen in ER-positive breast cancer.
- Published
- 2020
32. Validation-Based Insertional Mutagenesis (VBIM), a powerful forward genetic screening strategy
- Author
-
Sarmishtha De, Ilaria Tamagno, George R. Stark, and Mark W. Jackson
- Subjects
Medical Laboratory Technology ,Mutagenesis, Insertional ,Phenotype ,General Immunology and Microbiology ,General Neuroscience ,Lentivirus ,Mutation ,Health Informatics ,Genetic Testing ,General Pharmacology, Toxicology and Pharmaceutics ,General Biochemistry, Genetics and Molecular Biology ,Article - Abstract
Forward genetics begins with a biological phenotype and attempts to identify genetic changes that influence that phenotype. These changes can be induced in a selected group of genes, for instance, by using libraries of cDNAs, shRNAs, CRISPR guide RNAs, or genetic suppressor elements (GSEs), or randomly throughout the genome using chemical or insertional mutagens, with each approach creating distinct genetic changes. The Validation-Based Insertional Mutagenesis (VBIM) strategy utilizes modified lentiviruses as insertional mutagens, placing strong promoters throughout the genome. Generating libraries with millions of cells carrying one or a few VBIM promoter insertions is straightforward, allowing selection of cells in which overexpression of VBIM-driven RNAs or proteins promote the phenotype of interest. VBIM-driven RNAs may encode full-length proteins, truncated proteins (which may have wild-type, constitutive, or dominant-negative activity), or antisense RNAs that can disrupt gene expression. The diversity in VBIM-driven changes allows for the identification of both gain-of-function and loss-of-function mutations in a single screen. Additionally, VBIM can target any genomic locus, regardless of whether it is expressed in the cells under study or known to have a biological function, allowing for true whole-genome screens without the complication and cost of constructing, maintaining, and delivering a comprehensive library. Here, we review the VBIM strategy and discuss examples in which VBIM has been successfully used in diverse screens to identify novel genes or novel functions for known genes. In addition, we discuss considerations for transitioning the VBIM strategy to in vivo screens. We hope that other laboratories will be encouraged to use the VBIM strategy to identify genes that influence their phenotypes of interest. © 2022 Wiley Periodicals LLC.
- Published
- 2022
33. Bazedoxifene inhibits sustained STAT3 activation and increases survival in GBM
- Author
-
Justin D. Lathia, Xing Chen, Samantha M. Wightman, Yuxin Wang, Tyler J. Alban, and George R. Stark
- Subjects
eLDA, extreme limiting dilution assay ,Cancer Research ,BZA, Bazedoxifene ,BBB, blood-brain barrier ,STAT3, signal transducer and activator of transcription 3 ,Inflammation ,Bazedoxifene ,CNS, central nervous system ,JAK1, Janus Kinase 1 ,STAT3 ,Cancer stem cell ,Glioma ,IL6, interleukin-6 ,EGFR, Epidermal growth factor receptor ,medicine ,Glioma stem cells ,Transcription factor ,RC254-282 ,Original Research ,PDX, Patient-derived xenograph ,GSC, glioma cancer stem-like cell ,biology ,Chemistry ,Cancer stem cells ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,In vitro ,IL6 ,SOCS3, suppressor of cytokine signaling -3 ,Oncology ,Cancer research ,biology.protein ,GBM, glioblastoma ,Stem cell ,medicine.symptom ,IP, intraperitoneal ,Glioblastoma ,IL6Rα, Interleukin-6 receptor chain alpha - Abstract
Highlights • High STAT3 is correlated with poor prognosis in GBM. • Bazedoxifene targets IL-6-mediated sustained STAT3 activation. • STAT3 drives glioma stem cell maintenance, bazedoxifene targets this pathway. • Bazedoxifene crosses the BBB and prolongs survival in GBM bearing mice. • Bazedoxifene-treated tumors have less activated STAT3., An important factor correlated with poor survival in glioblastoma (GBM) is the aberrant and persistent activation of STAT3, a critical transcription factor that regulates multiple genes with key roles in cell survival, proliferation, resistance to chemotherapy, and stem cell maintenance. The Interleukin-6 (IL6)-STAT3 signaling axis has been studied extensively in inflammation and cancer. However, it is not completely understood how high levels of activated STAT3 are sustained in tumors. Previously, we identified a novel mechanism of biphasic activation of STAT3 in response to gp130-linked cytokines, including IL6, in which activation of STAT3 is prolonged by circumventing the negative regulatory mechanisms induced by its initial activationTo target prolonged STAT3 activation, we used the small molecule inhibitor bazedoxifene (BZA), which blocks formation of the IL6 receptor-gp130 complex. Glioma stem-like cells (GSCs) are more tumorigenic and more resistant to therapy. STAT3 is a key driver of the expression of stem cell transcription factors, making it a therapeutically important target in GBM. We show that treating GSCs with BZA decreases their self-renewal capacity and the expression of GSC markers in vitro. Additionally, BZA crosses the blood-brain barrier and confers a survival advantage in an orthotopic syngeneic mouse model of GBM. Although IL6-STAT3 signaling is important for GSC survival, a therapeutic agent that inhibits this pathway without toxicity has yet to be identified. Our findings reveal a mechanism of sustained STAT3 signaling in GBM and reveal its role in GSC maintenance, and we identify BZA as a novel candidate for treating GBM.
- Published
- 2021
34. The ubiquitin E3 ligase FBXO22 degrades PD-L1 and sensitizes cancer cells to DNA damage
- Author
-
Sarmishtha De, Kala Mahen, George R. Stark, Belinda Willard, and Elise G. Holvey-Bates
- Subjects
Lung Neoplasms ,DNA damage ,Ubiquitin-Protein Ligases ,Receptors, Cytoplasmic and Nuclear ,B7-H1 Antigen ,Ubiquitin ,PD-L1 ,Carcinoma, Non-Small-Cell Lung ,medicine ,Humans ,Phosphorylation ,Ubiquitins ,Cisplatin ,Multidisciplinary ,biology ,Chemistry ,Kinase ,F-Box Proteins ,Ubiquitination ,Cyclin-Dependent Kinase 5 ,Biological Sciences ,Immune checkpoint ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,A549 Cells ,Cancer cell ,biology.protein ,Cancer research ,medicine.drug ,DNA Damage ,Medulloblastoma - Abstract
High expression of programmed death-ligand 1 (PD-L1) in cancer cells drives immune-independent, cell-intrinsic functions, leading to resistance to DNA-damaging therapies. We find that high expression of the ubiquitin E3 ligase FBXO22 sensitizes nonsmall cell lung cancer (NSCLC) cells to ionizing radiation (IR) and cisplatin, and that activation of FBXO22 by phosphorylation is necessary for this function. Importantly, FBXO22 activates PD-L1 ubiquitination and degradation, which in turn increases the sensitivity of NSCLC cells to DNA damage. Cyclin-dependent kinase 5 (CDK5), aberrantly active in cancer cells, plays a crucial role in increasing the expression of PD-L1 in medulloblastoma [R. D. Dorand et al., Science 353, 399–403 (2016)]. We show in NSCLC cells that inhibiting CDK5 or reducing its expression increases the level of FBXO22, decreases that of PD-L1, and increases the sensitivity of the cells to DNA damage. We conclude that FBXO22 is a substrate of CDK5, and that inhibiting CDK5 reduces PD-L1 indirectly by increasing FBXO22. Pairing inhibitors of CDK5 with immune checkpoint inhibitors may increase the efficacy of immune checkpoint blockade alone or in combination with DNA-damaging therapies.
- Published
- 2021
35. PD-L1 sustains chronic, cancer cell-intrinsic responses to type I interferon, enhancing resistance to DNA damage
- Author
-
Elise G. Holvey-Bates, HyeonJoo Cheon, Daniel J. McGrail, and George R. Stark
- Subjects
Lung Neoplasms ,DNA damage ,B7-H1 Antigen ,Interferon-gamma ,Immune system ,Interferon ,PD-L1 ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,Cytotoxic T cell ,Humans ,Gene ,Multidisciplinary ,biology ,Chemistry ,Interferon-beta ,Biological Sciences ,Ligand (biochemistry) ,Nucleotidyltransferases ,Gene Expression Regulation, Neoplastic ,Cancer cell ,Interferon Type I ,Cancer research ,biology.protein ,medicine.drug ,DNA Damage ,Signal Transduction - Abstract
Programmed death ligand 1 (PD-L1), an immune-checkpoint protein expressed on cancer cells, also functions independently of the immune system. We found that PD-L1 inhibits the killing of cancer cells in response to DNA damage in an immune-independent manner by suppressing their acute response to type I interferon (IFN; IFN-I). In addition, PD-L1 plays a critical role in sustaining high levels of constitutive expression in cancer cells of a subset of IFN-induced genes, the IFN-related DNA damage resistance signature (IRDS) which, paradoxically, protects cancer cells. The cyclic GMP-AMP synthase-stimulator of the IFN genes (cGAS-STING) pathway is constitutively activated in a subset of cancer cells in the presence of high levels of PD-L1, thus leading to a constitutive, low level of IFN-β expression, which in turn increases IRDS expression. The constitutive low level of IFN-β expression is critical for the survival of cancer cells addicted to self-produced IFN-β. Our study reveals immune-independent functions of PD-L1 that inhibit cytotoxic acute responses to IFN-I and promote protective IRDS expression by supporting protective chronic IFN-I responses, both of which enhance the resistance of cancer cells to DNA damage.
- Published
- 2021
36. CBL0137 increases the targeting efficacy of Rovalpituzumab tesirine against tumour-initiating cells in small cell lung cancer
- Author
-
Yvonne Parker, Gary Wildey, George R. Stark, Daniel J. Lindner, Sarmishtha De, and Afshin Dowlati
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Immunoconjugates ,Lung Neoplasms ,Tics ,Carbazoles ,Apoptosis ,Mice, SCID ,Drug resistance ,Brief Communication ,Antibodies, Monoclonal, Humanized ,Small-cell lung cancer ,Mice ,03 medical and health sciences ,Targeted therapies ,0302 clinical medicine ,Mice, Inbred NOD ,Transcription (biology) ,Antineoplastic Combined Chemotherapy Protocols ,mental disorders ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Transcription factor ,Cell Proliferation ,Benzodiazepinones ,Experimental drug ,biology ,business.industry ,Rovalpituzumab tesirine ,Prognosis ,medicine.disease ,Small Cell Lung Carcinoma ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,Chromatin ,Histone ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business - Abstract
Small cell lung cancer (SCLC) is characterised by high relapse rates. Tumour-initiating cells (TICs) are responsible for drug resistance and recurrence of cancer. Rovalpituzumab tesirine (Rova-T), a potent humanised antibody–drug conjugate, selectively targets delta-like protein 3, which is highly expressed in SCLC TICs. The experimental drug CBL0137 (CBL) inhibits the histone chaperone FACT (facilitates chromatin transcription), which is required for the expression of transcription factors that are essential for TIC maintenance. Rova-T and CBL each target SCLC TICs as single agents. However, acquired or intrinsic resistance to single agents is a major problem in cancer. Therefore, we investigated the potential effect of combining Rova-T and CBL in SCLC to eradicate TICs more effectively. Our preclinical studies report a novel and highly translatable therapeutic strategy of dual targeting TICs using Rova-T in combination with CBL to potentially increase survival of SCLC patients.
- Published
- 2020
- Full Text
- View/download PDF
37. OAS-RNase L innate immune pathway mediates the cytotoxicity of a DNA-demethylating drug
- Author
-
Robert H. Silverman, George R. Stark, Yogen Saunthararajah, Beihua Dong, Yize Li, Elise G. Holvey-Bates, Susan R. Weiss, Shuvojit Banerjee, Elona Gusho, Frank Sicheri, Manisha Talukdar, Christina Gaughan, and Xiaorong Gu
- Subjects
0301 basic medicine ,Programmed cell death ,Medical Sciences ,RNase P ,DNA Methyltransferase Inhibitor ,DNA methyltransferase inhibitor ,RNase L ,Small Molecule Libraries ,03 medical and health sciences ,0302 clinical medicine ,5-azacytidine ,Radiation, Ionizing ,Endoribonucleases ,Gene expression ,2',5'-Oligoadenylate Synthetase ,Humans ,innate immunity ,Adaptor Proteins, Signal Transducing ,Multidisciplinary ,Cell Death ,Phosphoric Diester Hydrolases ,Chemistry ,Kinase ,Activator (genetics) ,JNK Mitogen-Activated Protein Kinases ,Transfection ,Biological Sciences ,Immunity, Innate ,3. Good health ,DNA Demethylation ,Isoenzymes ,030104 developmental biology ,A549 Cells ,Drug Resistance, Neoplasm ,Apoptosis ,030220 oncology & carcinogenesis ,Azacitidine ,Cancer research ,OAS - Abstract
Significance Drugs that cause epigenetic modification of DNA, such as 5-azacytidine (AZA), are used clinically to treat myelodysplastic syndromes and acute myeloid leukemia. In addition, AZA is being investigated for use against a range of different types of solid tumors, including lung and colorectal cancers. Treatment with AZA causes demethylation of DNA, thus increasing RNA synthesis, including the synthesis of double-stranded RNA, which is otherwise produced in virus-infected cells. We determined that cell death in response to AZA requires the antiviral enzyme RNase L. The results identify a drug target for enhancing the anticancer activity and reducing the toxicity of AZA and related drugs., Drugs that reverse epigenetic silencing, such as the DNA methyltransferase inhibitor (DNMTi) 5-azacytidine (AZA), have profound effects on transcription and tumor cell survival. AZA is an approved drug for myelodysplastic syndromes and acute myeloid leukemia, and is under investigation for different solid malignant tumors. AZA treatment generates self, double-stranded RNA (dsRNA), transcribed from hypomethylated repetitive elements. Self dsRNA accumulation in DNMTi-treated cells leads to type I IFN production and IFN-stimulated gene expression. Here we report that cell death in response to AZA treatment occurs through the 2′,5′-oligoadenylate synthetase (OAS)-RNase L pathway. OASs are IFN-induced enzymes that synthesize the RNase L activator 2-5A in response to dsRNA. Cells deficient in RNase L or OAS1 to 3 are highly resistant to AZA, as are wild-type cells treated with a small-molecule inhibitor of RNase L. A small-molecule inhibitor of c-Jun NH2-terminal kinases (JNKs) also antagonizes RNase L-dependent cell death in response to AZA, consistent with a role for JNK in RNase L-induced apoptosis. In contrast, the rates of AZA-induced and RNase L-dependent cell death were increased by transfection of 2-5A, by deficiencies in ADAR1 (which edits and destabilizes dsRNA), PDE12 or AKAP7 (which degrade 2-5A), or by ionizing radiation (which induces IFN-dependent signaling). Finally, OAS1 expression correlates with AZA sensitivity in the NCI-60 set of tumor cell lines, suggesting that the level of OAS1 can be a biomarker for predicting AZA sensitivity of tumor cells. These studies may eventually lead to pharmacologic strategies for regulating the antitumor activity and toxicity of AZA and related drugs.
- Published
- 2019
- Full Text
- View/download PDF
38. Inhibiting DNA-PK induces glioma stem cell differentiation and sensitizes glioblastoma to radiation in mice
- Author
-
Shideng Bao, Kui Zhai, Xiaoguang Fang, Jeremy N. Rich, Zhi Huang, Leo J.Y. Kim, Jennifer S. Yu, Alexandru Almasan, Xiaoxia Li, Weiwei Tao, Qiulian Wu, Qian Huang, and George R. Stark
- Subjects
endocrine system ,DNA damage ,Cellular differentiation ,DNA-Activated Protein Kinase ,Mice ,SOX2 ,Cell Line, Tumor ,Glioma ,medicine ,Animals ,Protein kinase A ,Transcription factor ,Brain Neoplasms ,Chemistry ,SOXB1 Transcription Factors ,fungi ,Cell Differentiation ,General Medicine ,medicine.disease ,DNA-Binding Proteins ,Cell culture ,embryonic structures ,Neoplastic Stem Cells ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Glioblastoma - Abstract
Glioblastoma (GBM), a lethal primary brain tumor, contains glioma stem cells (GSCs) that promote malignant progression and therapeutic resistance. SOX2 is a core transcription factor that maintains the properties of stem cells, including GSCs, but mechanisms associated with posttranslational SOX2 regulation in GSCs remain elusive. Here, we report that DNA-dependent protein kinase (DNA-PK) governs SOX2 stability through phosphorylation, resulting in GSC maintenance. Mass spectrometric analyses of SOX2-binding proteins showed that DNA-PK interacted with SOX2 in GSCs. The DNA-PK catalytic subunit (DNA-PKcs) was preferentially expressed in GSCs compared to matched non-stem cell tumor cells (NSTCs) isolated from patient-derived GBM xenografts. DNA-PKcs phosphorylated human SOX2 at S251, which stabilized SOX2 by preventing WWP2-mediated ubiquitination, thus promoting GSC maintenance. We then demonstrated that when the nuclear DNA of GSCs either in vitro or in GBM xenografts in mice was damaged by irradiation or treatment with etoposide, the DNA-PK complex dissociated from SOX2, which then interacted with WWP2, leading to SOX2 degradation and GSC differentiation. These results suggest that DNA-PKcs-mediated phosphorylation of S251 was critical for SOX2 stabilization and GSC maintenance. Pharmacological inhibition of DNA-PKcs with the DNA-PKcs inhibitor NU7441 reduced GSC tumorsphere formation in vitro and impaired growth of intracranial human GBM xenografts in mice as well as sensitized the GBM xenografts to radiotherapy. Our findings suggest that DNA-PK maintains GSCs in a stem cell state and that DNA damage triggers GSC differentiation through precise regulation of SOX2 stability, highlighting that DNA-PKcs has potential as a therapeutic target in glioblastoma.
- Published
- 2021
- Full Text
- View/download PDF
39. Correction: Multiple tumor suppressors regulate a HIF-dependent negative feedback loop via ISGF3 in human clear cell renal cancer
- Author
-
Geetha Jagannathan, Haifeng Yang, Robert G. Uzzo, Zongzhi Liu, Jie Na, Xiaohua Niu, Zhijiu Zhong, Stephen C. Peiper, Yaomin Xu, Essel Dulaimi, Qin Yan, George R. Stark, Wei Jiang, Eun-Ah Cho, Joseph R. Testa, Wesley L. Cai, Weijia Cai, Yuxin Wang, Jianxin Sun, Lili Liao, and Lauren Langbein
- Subjects
QH301-705.5 ,Science ,Mice, Nude ,Biology ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,law ,Negative feedback ,Cell Line, Tumor ,Gene expression ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Genes, Tumor Suppressor ,Cancer biology ,Multiple tumors ,Biology (General) ,Carcinoma, Renal Cell ,Cancer Biology ,Feedback, Physiological ,General Immunology and Microbiology ,General Neuroscience ,Gene Expression Profiling ,Cancer ,Correction ,General Medicine ,medicine.disease ,Chromosomes and Gene Expression ,Interferon-Stimulated Gene Factor 3, gamma Subunit ,Kidney Neoplasms ,Disease Models, Animal ,Gene Expression Regulation ,Von Hippel-Lindau Tumor Suppressor Protein ,Cancer research ,Suppressor ,Heterografts ,Medicine ,Clear cell ,Neoplasm Transplantation - Abstract
Whereas
- Published
- 2021
40. Suppressing <scp>PAR</scp> ylation by 2′,5′‐oligoadenylate synthetase 1 inhibits <scp>DNA</scp> damage‐induced cell death
- Author
-
Anna Kondratova, HyeonJoo Cheon, Beihua Dong, Elise G Holvey‐Bates, Metis Hasipek, Irina Taran, Christina Gaughan, Babal K Jha, Robert H Silverman, and George R Stark
- Subjects
General Immunology and Microbiology ,General Neuroscience ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2020
- Full Text
- View/download PDF
41. Amplification of CAD Genes
- Author
-
George R. Stark
- Published
- 2020
- Full Text
- View/download PDF
42. Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma
- Author
-
Andrew E. Sloan, Xiaoguang Fang, Xiuxing Wang, George R. Stark, Zhi Huang, Haidong Huang, Xiaoxia Li, Wenchao Zhou, Chengwei Chu, Qian Huang, Jeremy N. Rich, Weiwei Tao, Xingjiang Yu, Aili Zhang, Kui Zhai, Qiulian Wu, Shideng Bao, and Jennifer S. Yu
- Subjects
0301 basic medicine ,General Physics and Astronomy ,Kaplan-Meier Estimate ,Mice, SCID ,0302 clinical medicine ,RNA interference ,Mice, Inbred NOD ,Tumor Microenvironment ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Brain Neoplasms ,Wnt signaling pathway ,Glioma ,U937 Cells ,030220 oncology & carcinogenesis ,Doxycycline ,Neoplastic Stem Cells ,RNA Interference ,Stem cell ,Signal Transduction ,Cancer microenvironment ,endocrine system ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,CCN Intercellular Signaling Proteins ,03 medical and health sciences ,Paracrine signalling ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Gene silencing ,Animals ,Humans ,Autocrine signalling ,neoplasms ,Cell Proliferation ,Tumor microenvironment ,Macrophages ,fungi ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,nervous system diseases ,CNS cancer ,030104 developmental biology ,Cancer research ,lcsh:Q ,Proto-Oncogene Proteins c-akt - Abstract
The interplay between glioma stem cells (GSCs) and the tumor microenvironment plays crucial roles in promoting malignant growth of glioblastoma (GBM), the most lethal brain tumor. However, the molecular mechanisms underlying this crosstalk are incompletely understood. Here, we show that GSCs secrete the Wnt‐induced signaling protein 1 (WISP1) to facilitate a pro-tumor microenvironment by promoting the survival of both GSCs and tumor-associated macrophages (TAMs). WISP1 is preferentially expressed and secreted by GSCs. Silencing WISP1 markedly disrupts GSC maintenance, reduces tumor-supportive TAMs (M2), and potently inhibits GBM growth. WISP1 signals through Integrin α6β1-Akt to maintain GSCs by an autocrine mechanism and M2 TAMs through a paracrine manner. Importantly, inhibition of Wnt/β-catenin-WISP1 signaling by carnosic acid (CA) suppresses GBM tumor growth. Collectively, these data demonstrate that WISP1 plays critical roles in maintaining GSCs and tumor-supportive TAMs in GBM, indicating that targeting Wnt/β-catenin-WISP1 signaling may effectively improve GBM treatment and the patient survival., The tumour microenvironment plays an important role in promoting glioblastoma. Here, the authors show that glioma stem cells secrete WISP1, which promotes both the survival of the stem cells and tumour-associated macrophages.
- Published
- 2020
43. A virus-induced conformational switch of STAT1-STAT2 dimers boosts antiviral defenses
- Author
-
Zhuoya Wang, George R. Stark, Derek J. Taylor, Xin Wang, Wei Huang, Chenyang Zhao, Qiaoling Song, Jinbo Yang, Chenyao Wang, Yuxi Lin, Jing Nan, Belinda Willard, Elise G. Holvey-Bates, and Yuxin Wang
- Subjects
Protein Conformation ,Mice, Transgenic ,Virus ,Article ,Vesicular stomatitis Indiana virus ,chemistry.chemical_compound ,Mice ,Rhabdoviridae Infections ,Gene expression ,Chlorocebus aethiops ,Animals ,Humans ,Simplexvirus ,STAT1 ,STAT2 ,Phosphorylation ,Molecular Biology ,Vero Cells ,Innate immunity ,Innate immune system ,biology ,Tyrosine phosphorylation ,Herpes Simplex ,STAT2 Transcription Factor ,Cell Biology ,Fibroblasts ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,STAT1 Transcription Factor ,chemistry ,STAT protein ,biology.protein ,RNA Interference ,Protein Multimerization ,HeLa Cells ,Signal Transduction - Abstract
Type I interferons (IFN-I) protect us from viral infections. Signal transducer and activator of transcription 2 (STAT2) is a key component of interferon-stimulated gene factor 3 (ISGF3), which drives gene expression in response to IFN-I. Using electron microscopy, we found that, in naive cells, U-STAT2, lacking the activating tyrosine phosphorylation, forms a heterodimer with U-STAT1 in an inactive, anti-parallel conformation. A novel phosphorylation of STAT2 on T404 promotes IFN-I signaling by disrupting the U-STAT1-U-STAT2 dimer, facilitating the tyrosine phosphorylation of STATs 1 and 2 and enhancing the DNA-binding ability of ISGF3. IKK-ε, activated by virus infection, phosphorylates T404 directly. Mice with a T-A mutation at the corresponding residue (T403) are highly susceptible to virus infections. We conclude that T404 phosphorylation drives a critical conformational switch that, by boosting the response to IFN-I in infected cells, enables a swift and efficient antiviral defense.
- Published
- 2020
44. Inflammation mobilizes copper metabolism to promote colon tumorigenesis via an IL-17-STEAP4-XIAP axis
- Author
-
Shideng Bao, Emina Huang, Christopher J. Chang, Yun Liao, Ganes C. Sen, Matthew F. Kalady, George R. Stark, Fangqiang Tang, Mark W. Jackson, Xiaoxia Li, Katarzyna Bulek, Paul L. Fox, Shang Jia, Xing Chen, Junjie Zhao, Gang Cai, and Theresa T. Pizarro
- Subjects
0301 basic medicine ,Carcinogenesis ,General Physics and Astronomy ,medicine.disease_cause ,Inbred C57BL ,Inhibitor of Apoptosis Proteins ,Mice ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Aetiology ,lcsh:Science ,Cancer ,Mice, Knockout ,Multidisciplinary ,Interleukin-17 ,Chronic inflammation ,Colitis ,Colo-Rectal Cancer ,3. Good health ,XIAP ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Interleukin 17 ,medicine.symptom ,Intracellular ,Colon ,Science ,Knockout ,chemistry.chemical_element ,Inflammation ,General Biochemistry, Genetics and Molecular Biology ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,medicine ,Animals ,Humans ,Prevention ,Membrane Proteins ,General Chemistry ,Copper ,Colorectal cancer ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Apoptosis ,Cancer research ,lcsh:Q ,Digestive Diseases - Abstract
Copper levels are known to be elevated in inflamed and malignant tissues. But the mechanism underlying this selective enrichment has been elusive. In this study, we report a axis by which inflammatory cytokines, such as IL-17, drive cellular copper uptake via the induction of a metalloreductase, STEAP4. IL-17-induced elevated intracellular copper level leads to the activation of an E3-ligase, XIAP, which potentiates IL-17-induced NFκB activation and suppresses the caspase 3 activity. Importantly, this IL-17-induced STEAP4-dependent cellular copper uptake is critical for colon tumor formation in a murine model of colitis-associated tumorigenesis and STEAP4 expression correlates with IL-17 level and XIAP activation in human colon cancer. In summary, this study reveals a IL-17-STEAP4-XIAP axis through which the inflammatory response induces copper uptake, promoting colon tumorigenesis., STEAP4 promotes the uptake of copper, and copper is known to be enhanced in cancer tissues. Here, the authors show that STEAP4 is induced by IL17, which is increased in inflamed tissues, consequently the increased copper levels activate NFκB signalling and suppression of apoptosis.
- Published
- 2020
45. IFNL4-ΔG Allele Is Associated with an Interferon Signature in Tumors and Survival of African-American Men with Prostate Cancer
- Author
-
Ming Yi, Tiffany H. Dorsey, Wei Tang, Eric A. Klein, Olusegun O. Onabajo, Cristina Magi-Galluzzi, Robert M. Stephens, George R. Stark, Robert H. Silverman, Stefan Ambs, Christopher A. Loffredo, Symone V. Jordan, Ludmila Prokunina-Olsson, Tiffany A. Wallace, and Adeola Obajemu
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease ,Article ,Germline ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Prostate ,Interferon ,030220 oncology & carcinogenesis ,Internal medicine ,Genotype ,Cancer cell ,Medicine ,Allele ,business ,Gene ,medicine.drug - Abstract
Purpose: Men of African ancestry experience an excessive prostate cancer mortality that could be related to an aggressive tumor biology. We previously described an immune-inflammation signature in prostate tumors of African-American (AA) patients. Here, we further deconstructed this signature and investigated its relationships with tumor biology, survival, and a common germline variant in the IFNλ4 (IFNL4) gene. Experimental Design: We analyzed gene expression in prostate tissue datasets and performed genotype and survival analyses. We also overexpressed IFNL4 in human prostate cancer cells. Results: We found that a distinct interferon (IFN) signature that is analogous to the previously described “IFN-related DNA damage resistance signature” (IRDS) occurs in prostate tumors. Evaluation of two independent patient cohorts revealed that IRDS is detected about twice as often in prostate tumors of AA than European-American men. Furthermore, analysis in TCGA showed an association of increased IRDS in prostate tumors with decreased disease-free survival. To explain these observations, we assessed whether IRDS is associated with an IFNL4 germline variant (rs368234815-ΔG) that controls production of IFNλ4, a type III IFN, and is most common in individuals of African ancestry. We show that the IFNL4 rs368234815-ΔG allele was significantly associated with IRDS in prostate tumors and overall survival of AA patients. Moreover, IFNL4 overexpression induced IRDS in three human prostate cancer cell lines. Conclusions: Our study links a germline variant that controls production of IFNλ4 to the occurrence of a clinically relevant IFN signature in prostate tumors that may predominantly affect men of African ancestry. Clin Cancer Res; 24(21); 5471–81. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
46. CDK8/19 Mediator kinases potentiate induction of transcription by NFκB
- Author
-
Igor B. Roninson, Bing Hu, Eugenia V. Broude, Martina McDermott, Hao Ji, Gary P. Schools, Serena Altilia, Jiaxin Liang, Mengqian Chen, George R. Stark, Tao Lu, Donald C. Porter, David Oliver, Adam Schronce, Chang Uk Lim, Zhengguan Yang, and Michael Shtutman
- Subjects
0301 basic medicine ,Multidisciplinary ,biology ,General transcription factor ,NF-kappa B ,Promoter ,RNA polymerase II ,Biological Sciences ,Cyclin-Dependent Kinase 8 ,Molecular biology ,Cyclin-Dependent Kinases ,Cell biology ,03 medical and health sciences ,HEK293 Cells ,030104 developmental biology ,Mediator ,Gene Expression Regulation ,Cyclin-dependent kinase ,biology.protein ,Cytokines ,Humans ,Cyclin-dependent kinase 8 ,Kinase activity ,Transcription factor - Abstract
The nuclear factor-κB (NFκB) family of transcription factors has been implicated in inflammatory disorders, viral infections, and cancer. Most of the drugs that inhibit NFκB show significant side effects, possibly due to sustained NFκB suppression. Drugs affecting induced, but not basal, NFκB activity may have the potential to provide therapeutic benefit without associated toxicity. NFκB activation by stress-inducible cell cycle inhibitor p21 was shown to be mediated by a p21-stimulated transcription-regulating kinase CDK8. CDK8 and its paralog CDK19, associated with the transcriptional Mediator complex, act as coregulators of several transcription factors implicated in cancer; CDK8/19 inhibitors are entering clinical development. Here we show that CDK8/19 inhibition by different small-molecule kinase inhibitors or shRNAs suppresses the elongation of NFκB-induced transcription when such transcription is activated by p21-independent canonical inducers, such as TNFα. On NFκB activation, CDK8/19 are corecruited with NFκB to the promoters of the responsive genes. Inhibition of CDK8/19 kinase activity suppresses the RNA polymerase II C-terminal domain phosphorylation required for transcriptional elongation, in a gene-specific manner. Genes coregulated by CDK8/19 and NFκB include IL8, CXCL1, and CXCL2, which encode tumor-promoting proinflammatory cytokines. Although it suppressed newly induced NFκB-driven transcription, CDK8/19 inhibition in most cases had no effect on the basal expression of NFκB-regulated genes or promoters; the same selective regulation of newly induced transcription was observed with other transcription signals potentiated by CDK8/19. This selective role of CDK8/19 identifies these kinases as mediators of transcriptional reprogramming, a key aspect of development and differentiation as well as pathological processes.
- Published
- 2017
- Full Text
- View/download PDF
47. Role of Oligoadenylate Synthetases in Myeloid Neoplasia
- Author
-
Yihong Guan, Dale Grabowski, Babal K. Jha, Robert H. Silverman, George R. Stark, Yogenthiran Saunthararajah, Xiaorong Gu, Metis Hasipek, and Jaroslaw P. Maciejewski
- Subjects
Genome instability ,Myeloid ,DNA repair ,DNA damage ,Poly ADP ribose polymerase ,Immunology ,Cell Biology ,Hematology ,Base excision repair ,Biology ,medicine.disease ,Biochemistry ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Cancer research - Abstract
Acute myeloid leukemias (AML), the most lethal forms of blood cancer, are genomic instability disorders primarily driven by somatic mutations that greatly impact proliferation and survival of mutant clones1. The multistep mechanism of disease progression can be attributed, in part, to the defects in one or more pathways involving responses to, or repair of, damaged DNA (DD) and base excision repair (BER). The basal levels of DD and BER are higher in myeloid leukemias due to higher levels of reactive oxygen species (ROS)-mediated accumulation of 8-oxyguanine (8-OG)2. Poly(ADP) Ribosylation (PARylation) by PAR polymerases (PARPs) is one of the early key steps in sensing and repairing ROS-induced DNA damage response (DDR) 3. Consistent with recent findings4,5 here we report that 2'-5'oligoadenylate synthases (OASs), a family of latent 2'-5'-adenylyl transferases, otherwise involved in cellular antiviral responses6, are also involved in PAR remodeling of the DDR in MDS and AML cells. The 2'-hydroxyl group of PAR ribose acts as an acceptor for 2'AMP in the OAS enzymatic reaction and terminates chain elongation (Fig. A). OAS1 over expression can increase genomic instability by increasing the flux of PARP-mediated DNA repair, promoting cellular proliferation (Fig. B)5. Expression analysis of OASs in AML patients (n=451, Beat AML) showed that OAS1 is upregulated (2-fold) in AML patients compared to normal bone marrow-derived CD34+ hematopoietic stem and progenitor cells (HSPC)(Fig. C). Knockout of OAS1 and OAS2 using Crispr-Cas9 in MDS-L cells (Fig. D), a cell line derived from an MDS patient, confers sensitivity to H2O2-induced DNA damage-mediated cell death; however, OAS3 has no effect. Conversely, ectopic over expression of OAS1 or OAS2 but not OAS3 in HEK293 cells provides protection against H2O2-induced cell death (Fig. E-F). Proteomic analysis of the OAS1 and OAS2 interactome using LCMS/MS suggests that over expression of OAS1 and OAS2 perturbs the differentiation program of HSPCs that may result in neoplastic evolution. Thus, OASs modify PAR chains, promoting speedy DNA repair and cell survival along with the induction of a differentiation block in HSPCs that leads to clonal expansion. In summary, an overburdened DDR may contribute to AML pathogenesis. Therefore, inhibiting this stimulator of BER/DDR can provide a novel therapeutic avenue in myeloid neoplasms. The current study points to the probable utility of a novel therapeutic approach of targeting OAS in combination with DNA damaging agents to prevent relapse and resistance in the treatment of leukemias. References 1. Abelson S, Collord G, Ng SWK, et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature. 2018;559(7714):400-404. doi:10.1038/s41586-018-0317-6 2. Jankowska AM, Gondek LP, Szpurka H, Nearman ZP, Tiu RV, Maciejewski JP. Base excision repair dysfunction in a subgroup of patients with myelodysplastic syndrome. Leukemia. 2008;22(3):551-558. doi:10.1038/sj.leu.2405055 3. Rogge RA, Gibson BA, Kraus WL. Identifying Genomic Sites of ADP-Ribosylation Mediated by Specific Nuclear PARP Enzymes Using Click-ChIP. Methods Mol Biol. 2018;1813:371-387. doi:10.1007/978-1-4939-8588-3_25 4. Khodarev NN, Minn AJ, Efimova EV, et al. Signal transducer and activator of transcription 1 regulates both cytotoxic and prosurvival functions in tumor cells. Cancer Res. 2007;67(19):9214-9220. doi:10.1158/0008-5472.CAN-07-1019 5. Kondratova AA, Cheon H, Dong B, et al. Suppressing PARylation by 2',5'-oligoadenylate synthetase 1 inhibits DNA damage-induced cell death. EMBO J. 2020;39(11):e101573. doi:10.15252/embj.2019101573 6. Chakrabarti A, Jha BK, Silverman RH. New insights into the role of RNase L in innate immunity. J Interferon Cytokine Res. 2011;31(1):49-57. doi:10.1089/jir.2010.0120 Disclosures Saunthararajah: EpiDestiny: Consultancy, Current equity holder in private company, Patents & Royalties: University of Illinois at Chicago.
- Published
- 2020
- Full Text
- View/download PDF
48. Negative regulation of type I <scp>IFN</scp> signaling by phosphorylation of <scp>STAT</scp> 2 on T387
- Author
-
Jinbo Yang, Yuxin Wang, Xin Wang, Jing Nan, George R. Stark, and Belinda Willard
- Subjects
0301 basic medicine ,Response element ,Mutant ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Cyclin-dependent kinase ,Humans ,Phosphorylation ,STAT2 ,Molecular Biology ,Gene ,General Immunology and Microbiology ,Kinase ,General Neuroscience ,STAT2 Transcription Factor ,Interferon-Stimulated Gene Factor 3 ,Interferon-beta ,Articles ,030104 developmental biology ,biology.protein ,Cancer research ,Protein Processing, Post-Translational ,Signal Transduction - Abstract
The transcription factor ISGF3, comprised of IRF9 and tyrosine‐phosphorylated STATs 1 and 2, transmits the signal from the type I interferon receptor to the genome. We have discovered a novel phosphorylation of STAT2 on T387 that negatively regulates this response. In most untreated cell types, the majority of STAT2 is phosphorylated on T387 constitutively. In response to interferon‐β, the T387A mutant of STAT2 is much more effective than wild‐type STAT2 in mediating the expression of many interferon‐stimulated genes, in protecting cells against virus infection, and in inhibiting cell growth. Interferon‐β‐treated cells expressing wild‐type STAT2 contain much less ISGF3 capable of binding to an interferon‐stimulated response element than do cells expressing T387A STAT2. T387 lies in a cyclin‐dependent kinase (CDK) consensus sequence, and CDK inhibitors decrease T387 phosphorylation. Using CDK inhibitors to reverse the constitutive inhibitory phosphorylation of T387 of U‐STAT2 might enhance the efficacy of type I interferons in many different clinical settings.
- Published
- 2016
- Full Text
- View/download PDF
49. Abstract B051: IFNL4-deltaG allele is associated with an interferon signature in tumors and survival of African-American men with prostate cancer
- Author
-
Eric A. Klein, Symone V. Jordan, Cristina Magi-Galluzzi, Olusegun O. Onabajo, Tiffany H. Dorsey, George R. Stark, Stefan Ambs, Wei Tang, Robert H. Silverman, Robert L. Stephens, Ming Yi, Tiffany A. Wallace, Adeola Obajemu, Christopher A. Loffredo, and Ludmila Prokunina-Olsson
- Subjects
Oncology ,medicine.medical_specialty ,Epidemiology ,business.industry ,medicine.disease ,Prostate cancer ,Interferon ,Internal medicine ,medicine ,African american men ,Allele ,business ,medicine.drug - Abstract
Purpose: Men of African ancestry experience an excessive prostate cancer mortality that could be related to an aggressive tumor biology. We previously described an immune-inflammation signature in prostate tumors of African-American patients. Here, we further deconstructed this signature and investigated its relationships with tumor biology, survival, and a common germline variant in the interferon λ4 (IFNL4) gene. Experimental Design: We analyzed gene expression in prostate tissue datasets and performed IFNL4 genotype and survival analyses. We also overexpressed IFNL4 in human prostate cancer cells. Results: We found that a distinct interferon signature that is analogous to the previously described “Interferon-related DNA Damage Resistance Signature” (IRDS) occurs in prostate tumors. Evaluation of two independent patient cohorts revealed that IRDS is detected about twice as often in prostate tumors of African-American than European-American men. Furthermore, analysis in The Cancer Genome Atlas (TCGA) showed an association of increased IRDS in prostate tumors with decreased disease-free survival. To explain these observations, we assessed whether IRDS is associated with an IFNL4 germline variant (rs368234815-ΔG) that controls production of IFN-λ4 protein, a type-III interferon, and is most common in individuals of African ancestry. We show that the IFNL4 rs368234815-ΔG allele was significantly associated with IRDS in prostate tumors and overall survival of African-American patients. Moreover, IFNL4 overexpression induced IRDS-like signatures in three human prostate cancer cell lines. Conclusions: Tumor interferon signaling has recently been shown to modulate response and resistance to immune checkpoint blockade. Here, we describe a distinct and biologically relevant interferon signature, IRDS, in prostate tumors that has a high prevalence in African-American patients. Our observations indicate that IRDS and IFNL4 rs368234815-ΔG may have a function in the tumor biology and survival of African-American patients, and influence immune therapy outcomes, which should be examined in future studies. Citation Format: Wei Tang, Tiffany Wallace, Ming Yi, Cristina Magi-Galluzzi, Tiffany Dorsey, Olusegun Onabajo, Adeola Obajemu, Symone Jordan, Christopher Loffredo, Robert Stephens, Robert Silverman, George Stark, Eric Klein, Ludmila Prokunina-Olsson, Stefan Ambs. IFNL4-deltaG allele is associated with an interferon signature in tumors and survival of African-American men with prostate cancer [abstract]. In: Proceedings of the Eleventh AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2018 Nov 2-5; New Orleans, LA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2020;29(6 Suppl):Abstract nr B051.
- Published
- 2020
- Full Text
- View/download PDF
50. Multiple tumor suppressors regulate a HIF-dependent negative feedback loop via ISGF3 in human clear cell renal cancer
- Author
-
Qin Yan, Stephen C. Peiper, Zhijiu Zhong, Wesley L. Cai, Lili Liao, Jie Na, Essel Dulaimi, Yaomin Xu, George R. Stark, Weijia Cai, Robert G. Uzzo, Geetha Jagannathan, Wei Jiang, Joseph R. Testa, Haifeng Yang, Xiaohua Niu, Yuxin Wang, Jianxin Sun, Zongzhi Liu, Lauren Langbein, and Eun-Ah Cho
- Subjects
0301 basic medicine ,QH301-705.5 ,Science ,General Biochemistry, Genetics and Molecular Biology ,PBRM1 ,law.invention ,03 medical and health sciences ,law ,Interferon ,VHL ,medicine ,Humans ,BAP1 ,Biology (General) ,Carcinoma, Renal Cell ,Transcription factor ,Cancer Biology ,Cell Proliferation ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,kidney cancer ,General Medicine ,Chromosomes and Gene Expression ,medicine.disease ,Kidney Neoplasms ,Clear cell renal cell carcinoma ,030104 developmental biology ,KDM5C ,Interferon-Stimulated Gene Factor 3 ,Cancer research ,ISGF3 ,Medicine ,Suppressor ,Clear cell ,Research Article ,Human ,medicine.drug - Abstract
WhereasVHLinactivation is a primary event in clear cell renal cell carcinoma (ccRCC), the precise mechanism(s) of how this interacts with the secondary mutations in tumor suppressor genes, includingPBRM1,KDM5C/JARID1C,SETD2, and/orBAP1, remains unclear. Gene expression analyses reveal that VHL, PBRM1, or KDM5C share a common regulation of interferon response expression signature. Loss of HIF2α, PBRM1, or KDM5C inVHL-/-cells reduces the expression of interferon stimulated gene factor 3 (ISGF3), a transcription factor that regulates the interferon signature. Moreover, loss of SETD2 or BAP1 also reduces the ISGF3 level. Finally, ISGF3 is strongly tumor-suppressive in a xenograft model as its loss significantly enhances tumor growth. Conversely, reactivation of ISGF3 retards tumor growth by PBRM1-deficient ccRCC cells. Thus afterVHLinactivation, HIF induces ISGF3, which is reversed by the loss of secondary tumor suppressors, suggesting that this is a key negative feedback loop in ccRCC.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.