99 results on '"Bal L"'
Search Results
2. Data from RAD51AP1 Loss Attenuates Colorectal Cancer Stem Cell Renewal and Sensitizes to Chemotherapy
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Vinata B. Lokeshwar, Puttur D. Prasad, Santhakumar Manicassamy, Nagendra Singh, Hasan Korkaya, Pamela M. Martin, Nikhil Patel, Ravirajsinh N. Jedeja, Pachiappan Arjunan, Sarrah L. Hasanali, Daley S. Morera, Pragya Rajpurohit, Adrienne Lester, Utkarsh Parwal, Kavin Tamizhmani, Sabarish Ramachandran, and Allison E. Bridges
- Abstract
DNA damage, induced by either chemical carcinogens or environmental pollutants, plays an important role in the initiation of colorectal cancer. DNA repair processes, however, are involved in both protecting against cancer formation, and also contributing to cancer development, by ensuring genomic integrity and promoting the efficient DNA repair in tumor cells, respectively. Although DNA repair pathways have been well exploited in the treatment of breast and ovarian cancers, the role of DNA repair processes and their therapeutic efficacy in colorectal cancer is yet to be appreciably explored. To understand the role of DNA repair, especially homologous recombination (HR), in chemical carcinogen-induced colorectal cancer growth, we unraveled the role of RAD51AP1 (RAD51-associated protein 1), a protein involved in HR, in genotoxic carcinogen (azoxymethane, AOM)–induced colorectal cancer. Although AOM treatment alone significantly increased RAD51AP1 expression, the combination of AOM and dextran sulfate sodium (DSS) treatment dramatically increased by several folds. RAD51AP1 expression is found in mouse colonic crypt and proliferating cells. RAD51AP1 expression is significantly increased in majority of human colorectal cancer tissues, including BRAF/KRAS mutant colorectal cancer, and associated with reduced treatment response and poor prognosis. Rad51ap1-deficient mice were protected against AOM/DSS-induced colorectal cancer. These observations were recapitulated in a genetically engineered mouse model of colorectal cancer (ApcMin/+). Furthermore, chemotherapy-resistant colorectal cancer is associated with increased RAD51AP1 expression. This phenomenon is associated with reduced cell proliferation and colorectal cancer stem cell (CRCSC) self-renewal. Overall, our studies provide evidence that RAD51AP1 could be a novel diagnostic marker for colorectal cancer and a potential therapeutic target for colorectal cancer prevention and treatment.Implications:This study provides first in vivo evidence that RAD51AP1 plays a critical role in colorectal cancer growth and drug resistance by regulating CRCSC self-renewal.
- Published
- 2023
3. Data from β-Arrestins Regulate Stem Cell-Like Phenotype and Response to Chemotherapy in Bladder Cancer
- Author
-
Bal L. Lokeshwar, Vinata B. Lokeshwar, Markus A. Kuczyk, Axel S. Merseburger, Jiemin Li, Isha R. Dabke, Richard F. Pearce, James J. Hoy, Martin J. Hennig, Jie Gao, Daley S. Morera, Diandra K. Smith, and Georgios Kallifatidis
- Abstract
β-Arrestins are classic attenuators of G-protein–coupled receptor signaling. However, they have multiple roles in cellular physiology, including carcinogenesis. This work shows for the first time that β-arrestins have prognostic significance for predicting metastasis and response to chemotherapy in bladder cancer. β-Arrestin-1 (ARRB1) and β-arrestin-2 (ARRB2) mRNA levels were measured by quantitative RT-PCR in two clinical specimen cohorts (n = 63 and 43). The role of ARRBs in regulating a stem cell-like phenotype and response to chemotherapy treatments was investigated. The consequence of forced expression of ARRBs on tumor growth and response to Gemcitabine in vivo were investigated using bladder tumor xenografts in nude mice. ARRB1 levels were significantly elevated and ARRB2 levels downregulated in cancer tissues compared with normal tissues. In multivariate analysis only ARRB2 was an independent predictor of metastasis, disease-specific-mortality, and failure to Gemcitabine + Cisplatin (G+C) chemotherapy; ∼80% sensitivity and specificity to predict clinical outcome. ARRBs were found to regulate stem cell characteristics in bladder cancer cells. Depletion of ARRB2 resulted in increased cancer stem cell markers but ARRB2 overexpression reduced expression of stem cell markers (CD44, ALDH2, and BMI-1), and increased sensitivity toward Gemcitabine. Overexpression of ARRB2 resulted in reduced tumor growth and increased response to Gemcitabine in tumor xenografts. CRISPR-Cas9–mediated gene-knockout of ARRB1 resulted in the reversal of this aggressive phenotype. ARRBs regulate cancer stem cell-like properties in bladder cancer and are potential prognostic indicators for tumor progression and chemotherapy response.
- Published
- 2023
4. Supplemental Data from β-Arrestin-2 Counters CXCR7-Mediated EGFR Transactivation and Proliferation
- Author
-
Bal L. Lokeshwar, James J. Hoy, Nicole Salazar, Rajendra Kumar Singh, Daniel Munoz, and Georgios Kallifatidis
- Abstract
Supplemental Data
- Published
- 2023
5. Supplementary Methods from RAD51AP1 Loss Attenuates Colorectal Cancer Stem Cell Renewal and Sensitizes to Chemotherapy
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Vinata B. Lokeshwar, Puttur D. Prasad, Santhakumar Manicassamy, Nagendra Singh, Hasan Korkaya, Pamela M. Martin, Nikhil Patel, Ravirajsinh N. Jedeja, Pachiappan Arjunan, Sarrah L. Hasanali, Daley S. Morera, Pragya Rajpurohit, Adrienne Lester, Utkarsh Parwal, Kavin Tamizhmani, Sabarish Ramachandran, and Allison E. Bridges
- Abstract
Supplementary Methods
- Published
- 2023
6. Supplemental Figure 1: from β-Arrestin-2 Counters CXCR7-Mediated EGFR Transactivation and Proliferation
- Author
-
Bal L. Lokeshwar, James J. Hoy, Nicole Salazar, Rajendra Kumar Singh, Daniel Munoz, and Georgios Kallifatidis
- Abstract
Quantification of Western blot analysis from Figure 2.
- Published
- 2023
7. Supplementary Tables 1-4 and Supplementary Figures S1 to S7; and Supplementary Materials and Methods from β-Arrestins Regulate Stem Cell-Like Phenotype and Response to Chemotherapy in Bladder Cancer
- Author
-
Bal L. Lokeshwar, Vinata B. Lokeshwar, Markus A. Kuczyk, Axel S. Merseburger, Jiemin Li, Isha R. Dabke, Richard F. Pearce, James J. Hoy, Martin J. Hennig, Jie Gao, Daley S. Morera, Diandra K. Smith, and Georgios Kallifatidis
- Abstract
Supplementary Table S1: Specimen and patient characteristics; Supplementary Table S2: Determination of the association between adjuvant treatment failure and demographic/clinical parameters or marker levels; Supplementary Table S3: qPCR primers and ARRB1-specific CRISPR guide RNA sequences; Supplementary Table S4: Source of various antibodies used for immunofluorescence and Western blotting; Supplementary Figure S1: Kaplan Meier plot for stratification of patients into higher risk and lower risk for Disease-Specific Mortality (DSM) based on the ARRB2 arrestin level; Supplementary Figure S2: Expression of cytokeratins and Bmi-1 following modulation of ARRB2 levels in 5637 cells; Supplementary Figure S3: Expression of SOX2 following modulation of ARRB2 levels in bladder cancer cell lines; Supplementary Figure S4: Response of ARRB2 overexpressing transfectants to Cisplatin; Supplementary Figure S5: Expression ABC transporters in ARRB2-modulated bladder cancer cells; Supplementary Figure S6: Effect of ARRB2 overexpression on Cancer Stem Cell markers in vivo; Supplementary Figure S7: ARRB1 depletion affects clonogenic growth, spheroid formation and expression of CSC markers
- Published
- 2023
8. Supplemental Figure 3: from β-Arrestin-2 Counters CXCR7-Mediated EGFR Transactivation and Proliferation
- Author
-
Bal L. Lokeshwar, James J. Hoy, Nicole Salazar, Rajendra Kumar Singh, Daniel Munoz, and Georgios Kallifatidis
- Abstract
C4-2B cells were seeded in 8 well chamber slides in antibiotic-free media (30,000 cells per well). Twenty-four hours later, cells were transfected with ARR2-eGFP plasmid using Lipofectamine 2000. After overnight incubation, culture medium was replaced with low serum media (1% FBS). Twenty-four hours later, cells were washed with PBS-Tween and fixed with PFA, 4%.
- Published
- 2023
9. Supplementary table 1 from β-Arrestin-2 Counters CXCR7-Mediated EGFR Transactivation and Proliferation
- Author
-
Bal L. Lokeshwar, James J. Hoy, Nicole Salazar, Rajendra Kumar Singh, Daniel Munoz, and Georgios Kallifatidis
- Abstract
The primary antibodies used were: β-Actin (Cell Signaling, #5125s), β-arrestin-2 Antibody (SantaCruz Biotechnology, sc-13140), Akt Antibody (Cell Signaling, # 9272), phospho-Akt (pT308) (epitomics, #2214-1), phospho-Akt (pS473) (epitomics, #2118-1), Cyclin A (Cell Signaling, # 4656s), Cyclin D1 (Abcam, ab 134175), CXCR7 Antibody (Proteintech, 20423-1-AP, and customized Antibody from Abmart, GeneTech), EGFR Antibody (Cell Signaling, #2646), phospho-EGFR (pY1110) (Abcam, ab68470, ab47370), phospho-EGFR (pY1068) (Epitomics, 1138-1), Lamin A/C (epitomics, 2966-1), phospho-p44/42 MAPK (pERK1/2) (cell signaling, #9106s), p44/42 MAPK (cell signaling, #9102), Src Family (Tyr416) Antibody (Cell Signaling, #2108), phospho-Src Family (pY416) Antibody (Cell Signaling, #2101), ɑ-Tubulin Antibody (Cell Signaling, #2144). Blots were probed with a 1:10,000 dilution of an anti-rabbit or anti-mouse horseradish peroxidase-conjugated secondary antibody (Cell Signaling).
- Published
- 2023
10. Supplementary Data from RAD51AP1 Loss Attenuates Colorectal Cancer Stem Cell Renewal and Sensitizes to Chemotherapy
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Vinata B. Lokeshwar, Puttur D. Prasad, Santhakumar Manicassamy, Nagendra Singh, Hasan Korkaya, Pamela M. Martin, Nikhil Patel, Ravirajsinh N. Jedeja, Pachiappan Arjunan, Sarrah L. Hasanali, Daley S. Morera, Pragya Rajpurohit, Adrienne Lester, Utkarsh Parwal, Kavin Tamizhmani, Sabarish Ramachandran, and Allison E. Bridges
- Abstract
Supplementary Figures and Legends 1-3
- Published
- 2023
11. Data from A Novel Splice Variant of HYAL-4 Drives Malignant Transformation and Predicts Outcome in Patients with Bladder Cancer
- Author
-
Bal L. Lokeshwar, Santu Ghosh, Luis E. Lopez, Sravan Kavuri, Zachary Klaassen, Murugesan Manoharan, Ijeoma Azih, Kelly Hoye, Andre R. Jordan, Neetika Dhir, Ronny R. Racine, Diogo O. Escudero, Rohitha Baskar, Martin J.P. Hennig, Jiaojiao Wang, Soum D. Lokeshwar, Judith Knapp, Marie C. Hupe, Travis J. Yates, Sarrah L. Hasanali, Daley S. Morera, and Vinata B. Lokeshwar
- Abstract
Purpose:Poor prognosis of patients with muscle-invasive bladder cancer that often metastasizes drives the need for discovery of molecular determinants of bladder cancer progression. Chondroitin sulfate proteoglycans, including CD44, regulate cancer progression; however, the identity of a chondroitinase (Chase) that cleaves chondroitin sulfate from proteoglycans is unknown. HYAL-4 is an understudied gene suspected to encode a Chase, with no known biological function. We evaluated HYAL-4 expression and its role in bladder cancer.Experimental Design:In clinical specimens, HYAL-4 wild-type (Wt) and V1 expression was evaluated by RT-qPCR, IHC, and/or immunoblotting; a novel assay measured Chase activity. Wt and V1 were stably expressed or silenced in normal urothelial and three bladder cancer cell lines. Transfectants were analyzed for stem cell phenotype, invasive signature and tumorigenesis, and metastasis in four xenograft models, including orthotopic bladder.Results:HYAL-4 expression, specifically a novel splice variant (V1), was elevated in bladder tumors; Wt expression was barely detectable. V1 encoded a truncated 349 amino acid protein that was secreted. In bladder cancer tissues, V1 levels associated with metastasis and cancer-specific survival with high efficacy and encoded Chase activity. V1 cleaved chondroitin-6-sulfate from CD44, increasing CD44 secretion. V1 induced stem cell phenotype, motility/invasion, and an invasive signature. CD44 knockdown abrogated these phenotypes. V1-expressing urothelial cells developed angiogenic, muscle-invasive tumors. V1-expressing bladder cancer cells formed tumors at low density and formed metastatic bladder tumors when implanted orthotopically.Conclusions:Our study discovered the first naturally-occurring eukaryotic/human Chase and connected it to disease pathology, specifically cancer. V1-Chase is a driver of malignant bladder cancer and potential predictor of outcome in patients with bladder cancer.
- Published
- 2023
12. Supplementary Tables and Figures from A Novel Splice Variant of HYAL-4 Drives Malignant Transformation and Predicts Outcome in Patients with Bladder Cancer
- Author
-
Bal L. Lokeshwar, Santu Ghosh, Luis E. Lopez, Sravan Kavuri, Zachary Klaassen, Murugesan Manoharan, Ijeoma Azih, Kelly Hoye, Andre R. Jordan, Neetika Dhir, Ronny R. Racine, Diogo O. Escudero, Rohitha Baskar, Martin J.P. Hennig, Jiaojiao Wang, Soum D. Lokeshwar, Judith Knapp, Marie C. Hupe, Travis J. Yates, Sarrah L. Hasanali, Daley S. Morera, and Vinata B. Lokeshwar
- Abstract
Four Supplementary Figures and Seven Supplementary Tables
- Published
- 2023
13. Data from CXC Receptor-1 Silencing Inhibits Androgen-Independent Prostate Cancer
- Author
-
Bal L. Lokeshwar, Diogo O. Escudero, Dominic A. Lyn, and Nagarajarao Shamaladevi
- Abstract
The CXC receptor-1 (CXCR1) is a coreceptor for interleukin-8 (IL-8) and is expressed on both normal and tumor cells. The function of CXCR1 in prostate cancer was investigated by silencing its expression, using RNA interference. We established stable cell colonies of PC-3 cells, depleted of CXCR1, using lentiviral plasmids (pLK0.1puro) generating small hairpin RNA (shRNA) against CXCR1 mRNA. Stable shRNA transfectants (PLK1–PLK5) that express significantly reduced CXCR1 mRNA (≥90% down) and protein (≥43% down) or vector-only transfectants (PC-3V) were characterized. PLK cells showed reduced cell proliferation (down, ≥66%), due to cell cycle arrest at G1-S phase, decreases in Cyclin D1, CDK4, phosphorylated Rb, and extracellular signal-regulated kinase 1/2 levels compared with those in PC-3V cells. CXCR1 depletion lead to increases in spontaneous apoptosis by mitochondria-mediated intrinsic mechanism and increases in proapoptotic proteins (BAD, 40%; BAX, 12%), but decreases in antiapoptotic proteins (BCL2, down 38%; BCLxL, 20%). PLK2 cells grew as slow-growing tumors (decrease of 54%), compared with that of PC3V tumors in athymic mice. Ex vivo analyses of PLK2 tumor tissues showed reduced expression of Cyclin D1 and vascular endothelial growth factor, and increased apoptosis activity. Other IL-8–expressing prostate cancer cell lines also exhibited similar phenotypes when CXCR1 was depleted by CXCR1 shRNA transfection. In contrast to these cells, CXCR1 depletion had little effect on IL-8 ligand–deficient LNCaP cells. RNA interference rescue using mutated CXCR1 plasmids reversed the silencing effect of PLK2, thus demonstrating the specificity of phenotypic alteration by CXCR1 shRNA. These studies establish that CXCR1 promotes IL-8–mediated tumor growth. [Cancer Res 2009;69(21):8265–74]
- Published
- 2023
14. Supplementary Table 1 from Combined Inhibition of DNMT and HDAC Blocks the Tumorigenicity of Cancer Stem-like Cells and Attenuates Mammary Tumor Growth
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Suash Sharma, Puttur D. Prasad, Ravindra Kolhe, Santhakumar Manicassamy, Jeong-Hyeon Choi, Huidong Shi, Priyanka Thakur, Gurusamy Mariappan, Sabarish Ramachandran, and Rajneesh Pathania
- Abstract
Supplementary table 1 will provide a complete list of genes that are differentially regulated genes in RNA-seq analysis for control and 5-AzaC+Butyrate treated CSCs
- Published
- 2023
15. Supplementary Figure 5 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
Supplementary Figure 5 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Published
- 2023
16. Supplementary Table 2 from Combined Inhibition of DNMT and HDAC Blocks the Tumorigenicity of Cancer Stem-like Cells and Attenuates Mammary Tumor Growth
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Suash Sharma, Puttur D. Prasad, Ravindra Kolhe, Santhakumar Manicassamy, Jeong-Hyeon Choi, Huidong Shi, Priyanka Thakur, Gurusamy Mariappan, Sabarish Ramachandran, and Rajneesh Pathania
- Abstract
Supplementary Table S2 will provide information about the Pathway and gene network analyses
- Published
- 2023
17. Supplementary Figure Legend 5 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
Supplementary Figure Legend 5 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Published
- 2023
18. Supplementary Figure 3 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
Supplementary Figure 3 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Published
- 2023
19. Supplementary Figure 2 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
Supplementary Figure 2 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Published
- 2023
20. Data from Interleukin-8 Is a Molecular Determinant of Androgen Independence and Progression in Prostate Cancer
- Author
-
Bal L. Lokeshwar, Vinata B. Lokeshwar, Yekutiel Sandman, David M. Meinbach, Rajendra K. Singh, Dominic Lyn, Yohei Omori, and Shinako Araki
- Abstract
The proinflammatory chemokine interleukin-8 (IL-8) is undetectable in androgen-responsive prostate cancer cells (e.g., LNCaP and LAPC-4), but it is highly expressed in androgen-independent metastatic cells, such as PC-3. In this report, we show IL-8 functions in androgen independence, chemoresistance, tumor growth, and angiogenesis. We stably transfected LNCaP and LAPC-4 cells with IL-8 cDNA and selected IL-8–secreting (IL8-S) transfectants. The IL8-S transfectants that secreted IL-8 at levels similar to that secreted by PC-3 cells (100–170 ng/106 cells) were characterized. Continuous or transient exposure of LNCaP and LAPC-4 cells to IL-8 reduced their dependence on androgen for growth and decreased sensitivity (>3.5×) to an antiandrogen. IL-8–induced cell proliferation was mediated through CXCR1 and was independent of androgen receptor (AR). Quantitative PCR, immunoblotting, and transfection studies showed that IL8-S cells or IL-8–treated LAPC-4 cells exhibit a 2- to 3-fold reduction in PSA and AR levels, when compared with vector transfectants. IL8-S cells expressed 2- to 3-fold higher levels of phospho-EGFR, src, Akt, and nuclear factor κB (NF-κB) and showed increased survival when treated with docetaxel. This increase was blocked by NF-κB and src inhibitors, but not by an Akt inhibitor. IL8-S transfectants displayed a 3- to 5-fold increased motility, invasion, matrix metalloproteinase-9 and vascular endothelial growth factor production. LNCaP IL8-S cells grew rapidly as tumors, with increased microvessel density and abnormal tumor vasculature when compared with the tumors derived from their vector-transfected counterparts. Therefore, IL-8 is a molecular determinant of androgen-independent prostate cancer growth and progression. [Cancer Res 2007;67(14):6854–62]
- Published
- 2023
21. Data from Combined Inhibition of DNMT and HDAC Blocks the Tumorigenicity of Cancer Stem-like Cells and Attenuates Mammary Tumor Growth
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Suash Sharma, Puttur D. Prasad, Ravindra Kolhe, Santhakumar Manicassamy, Jeong-Hyeon Choi, Huidong Shi, Priyanka Thakur, Gurusamy Mariappan, Sabarish Ramachandran, and Rajneesh Pathania
- Abstract
Recently, impressive technical advancements have been made in the isolation and validation of mammary stem cells and cancer stem cells (CSC), but the signaling pathways that regulate stem cell self-renewal are largely unknown. Furthermore, CSCs are believed to contribute to chemo- and radioresistance. In this study, we used the MMTV-Neu-Tg mouse mammary tumor model to identify potential new strategies for eliminating CSCs. We found that both luminal progenitor and basal stem cells are susceptible to genetic and epigenetic modifications, which facilitate oncogenic transformation and tumorigenic potential. A combination of the DNMT inhibitor 5-azacytidine and the HDAC inhibitor butyrate markedly reduced CSC abundance and increased the overall survival in this mouse model. RNA-seq analysis of CSCs treated with 5-azacytidine plus butyrate provided evidence that inhibition of chromatin modifiers blocks growth-promoting signaling molecules such as RAD51AP1 and SPC25, which play key roles in DNA damage repair and kinetochore assembly. Moreover, RAD51AP1 and SPC25 were significantly overexpressed in human breast tumor tissues and were associated with reduced overall patient survival. In conclusion, our studies suggest that breast CSCs are intrinsically sensitive to genetic and epigenetic modifications and can therefore be significantly affected by epigenetic-based therapies, warranting further investigation of combined DNMT and HDAC inhibition in refractory or drug-resistant breast cancer. Cancer Res; 76(11); 3224–35. ©2016 AACR.
- Published
- 2023
22. Supplementary Table 1 from Interleukin-8 Is a Molecular Determinant of Androgen Independence and Progression in Prostate Cancer
- Author
-
Bal L. Lokeshwar, Vinata B. Lokeshwar, Yekutiel Sandman, David M. Meinbach, Rajendra K. Singh, Dominic Lyn, Yohei Omori, and Shinako Araki
- Abstract
Supplementary Table 1 from Interleukin-8 Is a Molecular Determinant of Androgen Independence and Progression in Prostate Cancer
- Published
- 2023
23. Supplementary Tables 1-2, Figures 1-3 from CXC Receptor-1 Silencing Inhibits Androgen-Independent Prostate Cancer
- Author
-
Bal L. Lokeshwar, Diogo O. Escudero, Dominic A. Lyn, and Nagarajarao Shamaladevi
- Abstract
Supplementary Tables 1-2, Figures 1-3 from CXC Receptor-1 Silencing Inhibits Androgen-Independent Prostate Cancer
- Published
- 2023
24. Supplementary Figure 1 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
Supplementary Figure 1 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Published
- 2023
25. Data from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
The proinflammatory chemokine receptor CXCR7 that binds the ligands CXCL11 and CXCL12 (SDF-1a) is elevated in a variety of human cancers, but its functions are not understood as it does not elicit classical chemokine receptor signaling. Here we report that the procancerous cytokine IL-8 (interleukin-8) upregulates CXCR7 expression along with ligand-independent functions of CXCR7 that promote the growth and proliferation of human prostate cancer cells (CaP cells). In cell culture, ectopic expression or addition of IL-8 selectively increased expression of CXCR7 at the level of mRNA and protein production. Conversely, suppressing IL-8 signaling abolished the ability of IL-8 to upregulate CXCR7. RNAi-mediated knockdown of CXCR7 in CaP cells caused multiple antitumor effects, including decreased cell proliferation, cell-cycle arrest in G1 phase, and decreased expression of proteins involved in G1 to S phase progression. In contrast, addition of the CXCR7 ligand SDF-1a and CXCL11 to CaP cells did not affect cell proliferation. Over expression of CXCR7 in normal prostate cells increased their proliferation in a manner associated with increased levels of phospho-EGFR (epidermal growth factor receptor; pY1110) and phospho-ERK1/2. Notably, coimmunoprecipitation studies established a physical association of CXCR7 with EGFR, linking CXCR7-mediated cell proliferation to EGFR activation. Consistent with these findings, CXCR7-depleted CaP tumors grew more slowly than control tumors, expressing decreased tumor-associated expression of VEGF, cyclin D1, and p-EGFR. Together, these results reveal a novel mechanism of ligand-independent growth promotion by CXCR7 and its coregulation by the proinflammatory factor IL-8 in prostate cancer. Cancer Res; 71(9); 3268–77. ©2011 AACR.
- Published
- 2023
26. Supplementary Figures Legends 1-4, Tables 1-2 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
Supplementary Figures Legends 1-4, Tables 1-2 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Published
- 2023
27. Supplemental Materials and Methods from Combined Inhibition of DNMT and HDAC Blocks the Tumorigenicity of Cancer Stem-like Cells and Attenuates Mammary Tumor Growth
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Suash Sharma, Puttur D. Prasad, Ravindra Kolhe, Santhakumar Manicassamy, Jeong-Hyeon Choi, Huidong Shi, Priyanka Thakur, Gurusamy Mariappan, Sabarish Ramachandran, and Rajneesh Pathania
- Abstract
The Supplemental materials and method file has methods for the Generation of mammospheres and tumorospheres, Preparation of single cell suspension from mammary gland, methods for RNA Isolation and Real-time PCR, and Clonogenic assay.
- Published
- 2023
28. Supplementary Figure legends from Combined Inhibition of DNMT and HDAC Blocks the Tumorigenicity of Cancer Stem-like Cells and Attenuates Mammary Tumor Growth
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Suash Sharma, Puttur D. Prasad, Ravindra Kolhe, Santhakumar Manicassamy, Jeong-Hyeon Choi, Huidong Shi, Priyanka Thakur, Gurusamy Mariappan, Sabarish Ramachandran, and Rajneesh Pathania
- Abstract
Supplementary Figure legends provide clear description for the Supplementary Figures S1-S7.
- Published
- 2023
29. Supplemental Figures from Combined Inhibition of DNMT and HDAC Blocks the Tumorigenicity of Cancer Stem-like Cells and Attenuates Mammary Tumor Growth
- Author
-
Muthusamy Thangaraju, Vadivel Ganapathy, Bal L. Lokeshwar, Suash Sharma, Puttur D. Prasad, Ravindra Kolhe, Santhakumar Manicassamy, Jeong-Hyeon Choi, Huidong Shi, Priyanka Thakur, Gurusamy Mariappan, Sabarish Ramachandran, and Rajneesh Pathania
- Abstract
The Supplemental figure file contains severn additional figures. Supplementary figure 1 shows that the Lin-CD49f+CD24+ cells are tumorigenic, Supplementary Figure 2 provide additional evidence for myoepithelial stem and luminal progenitor cells are the targets of genetic mutations/epigenetic modifications that lead to tumor cell of origin, Supplementary Figure 3, shows that the treatment with DNMT and HDAC inhibitors reduces colony formation, Supplementary Figure 4l provide a Flow chart for RNA sequencing analysis, Supplementary Figure 5 shows a heat map for differential expression of genes in normal and in breast cancer, Supplementary Figure 6 shows RAD51AP1 expression in basal breast cancer, and Supplementary Figure 7 shows SPC25 expression in basal breast cancer
- Published
- 2023
30. Supplementary Figure 4 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Author
-
Bal L. Lokeshwar and Rajendra Kumar Singh
- Abstract
Supplementary Figure 4 from The IL-8–Regulated Chemokine Receptor CXCR7 Stimulates EGFR Signaling to Promote Prostate Cancer Growth
- Published
- 2023
31. Promotion of epithelial hyperplasia by interleukin‐8—CXCR axis in human prostate
- Author
-
Daley S. Morera, Diandra K. Smith, Martha K. Terris, Zachary Klaassen, Georgios Kallifatidis, Jiaojiao Wang, Bal L. Lokeshwar, Vinata B. Lokeshwar, Andre R. Jordan, Sarrah L. Hasanali, and Roni J. Bollag
- Subjects
Male ,0301 basic medicine ,Chemokine ,Stromal cell ,Urology ,Prostatic Hyperplasia ,Cell Growth Processes ,urologic and male genital diseases ,Article ,Epithelium ,Cell Line ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Prostate ,medicine ,Humans ,RNA, Messenger ,Interleukin 8 ,Oleanolic Acid ,Receptor ,Cells, Cultured ,Receptors, CXCR ,biology ,Chemistry ,Cell growth ,Interleukin-8 ,Hyperplasia ,medicine.disease ,Triterpenes ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Signal Transduction - Abstract
Background The clinical manifestation of benign prostatic hyperplasia (BPH) is causally linked to the inflammatory microenvironment and proliferation of epithelial and stromal cells in the prostate transitional zone. The CXC-chemokine interleukin-8 (IL-8) contributes to inflammation. We evaluated the expression of inflammatory cytokines in clinical specimens, primary cultures, and prostatic lineage cell lines. We investigated whether IL-8 via its receptor system (IL-8 axis) promotes BPH. Methods The messenger RNA and protein expression of chemokines, including components of the IL-8 axis, were measured in normal prostate (NP; n = 7) and BPH (n = 21), urine (n = 24) specimens, primary cultures, prostatic lineage epithelial cell lines (NHPrE1, BHPrE1, BPH-1), and normal prostate cells (RWPE-1). The functional role of the IL-8 axis in prostate epithelial cell growth was evaluated by CRISPR/Cas9 gene editing. The effect of a combination with two natural compounds, oleanolic acid (OA) and ursolic acid (UA), was evaluated on the expression of the IL-8 axis and epithelial cell growth. Results Among the 19 inflammatory chemokines and chemokine receptors we analyzed, levels of IL-8 and its receptors (CXCR1, CXCR2), as well as, of CXCR7, a receptor for CXCL12, were 5- to 25-fold elevated in BPH tissues when compared to NP tissues (P ≤ .001). Urinary IL-8 levels were threefold to sixfold elevated in BPH patients, but not in asymptomatic males and females with lower urinary tract symptoms (P ≤ .004). The expression of the IL-8 axis components was confined to the prostate luminal epithelial cells in both normal and BPH tissues. However, these components were elevated in BPH-1 and primary explant cultures as compared to RWPE-1, NHPrE1, and BHPrE1 cells. Knockout of CXCR7 reduced IL-8, and CXCR1 expression by 4- to 10-fold and caused greater than or equal to 50% growth inhibition in BPH-1 cells. Low-dose OA + UA combination synergistically inhibited the growth of BPH-1 and BPH primary cultures. In the combination, the drug reduction indices for UA and OA were 16.4 and 7852, respectively, demonstrating that the combination was effective in inhibiting BPH-1 growth at significantly reduced doses of UA or OA alone. Conclusion The IL-8 axis is a promotor of BPH pathogenesis. Low-dose OA + UA combination inhibits BPH cell growth by inducing autophagy and reducing IL-8 axis expression in BPH-epithelial cells.
- Published
- 2020
32. Ursolic Acid Analogs as Potential Therapeutics for Cancer
- Author
-
Siva S. Panda, Muthusamy Thangaraju, and Bal L. Lokeshwar
- Subjects
Chemistry (miscellaneous) ,Neoplasms ,Organic Chemistry ,Drug Discovery ,Anti-Inflammatory Agents ,Humans ,Molecular Medicine ,Pharmaceutical Science ,Antineoplastic Agents ,Physical and Theoretical Chemistry ,Antioxidants ,Triterpenes ,Analytical Chemistry - Abstract
Ursolic acid (UA) is a pentacyclic triterpene isolated from a large variety of vegetables, fruits and many traditional medicinal plants. It is a structural isomer of Oleanolic Acid. The medicinal application of UA has been explored extensively over the last two decades. The diverse pharmacological properties of UA include anti-inflammatory, antimicrobial, antiviral, antioxidant, anti-proliferative, etc. Especially, UA holds a promising position, potentially, as a cancer preventive and therapeutic agent due to its relatively non-toxic properties against normal cells but its antioxidant and antiproliferative activities against cancer cells. Cell culture studies have shown interference of UA with multiple pharmacological and molecular targets that play a critical role in many cells signaling pathways. Although UA is considered a privileged natural product, its clinical applications are limited due to its low absorption through the gastro-intestinal track and rapid elimination. The low bioavailability of UA limits its use as a therapeutic drug. To overcome these drawbacks and utilize the importance of the scaffold, many researchers have been engaged in designing and developing synthetic analogs of UA via structural modifications. This present review summarizes the synthetic UA analogs and their cytotoxic antiproliferative properties reported in the last two decades.
- Published
- 2022
33. Targeting Mitochondrial Metabolism in Prostate Cancer with Triterpenoids
- Author
-
Georgios Kallifatidis, Bal L. Lokeshwar, and Kenza Mamouni
- Subjects
Male ,mitochondrial metabolism ,Oxidative phosphorylation ,Review ,Mitochondrion ,Biology ,Catalysis ,Oxidative Phosphorylation ,Inorganic Chemistry ,lcsh:Chemistry ,Prostate cancer ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Glycolysis ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,Organic Chemistry ,Prostatic Neoplasms ,General Medicine ,triterpenoids ,medicine.disease ,prostate cancer ,Warburg effect ,Triterpenes ,Computer Science Applications ,Mitochondria ,Oxidative Stress ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer cell ,Lipogenesis ,Cancer research - Abstract
Metabolic reprogramming is a hallmark of malignancy. It implements profound metabolic changes to sustain cancer cell survival and proliferation. Although the Warburg effect is a common feature of metabolic reprogramming, recent studies have revealed that tumor cells also depend on mitochondrial metabolism. Due to the essential role of mitochondria in metabolism and cell survival, targeting mitochondria in cancer cells is an attractive therapeutic strategy. However, the metabolic flexibility of cancer cells may enable the upregulation of compensatory pathways, such as glycolysis, to support cancer cell survival when mitochondrial metabolism is inhibited. Thus, compounds capable of targeting both mitochondrial metabolism and glycolysis may help overcome such resistance mechanisms. Normal prostate epithelial cells have a distinct metabolism as they use glucose to sustain physiological citrate secretion. During the transformation process, prostate cancer cells consume citrate to mainly power oxidative phosphorylation and fuel lipogenesis. A growing number of studies have assessed the impact of triterpenoids on prostate cancer metabolism, underlining their ability to hit different metabolic targets. In this review, we critically assess the metabolic transformations occurring in prostate cancer cells. We will then address the opportunities and challenges in using triterpenoids as modulators of prostate cancer cell metabolism.
- Published
- 2021
34. Contributors
- Author
-
S.J. Aditya Rao, Vijai V. Alex, Nasreen Amin, Ruby John Anto, Megha Das, Jie Gao, C.S. Joshi, Vinod K. Kannaujiya, Sanjay Kumar, Bal L. Lokeshwar, Kenza Mamouni, Tarun Minocha, Amit Kumar Mishra, Nitesh Kumar Mishra, Pragyan Mishra, Soumya Ranjan Mohanty, Haritha H. Nair, Sukanya Patra, Richa Raghuwanshi, Amit Ranjan, Rachana Shalini, Diksha Sharma, Vinamra Sharma, Ankita Shrivastava, Divya Singh, Monika Singh, Rajesh Kumar Singh, V.P. Singh, Akanksha Srivastava, Akhileshwar Kumar Srivastava, Ruchita Tripathi, Sanjeev Kumar Yadav, Arpana Yadava, and Lei Zhang
- Published
- 2021
35. Spice up your food for cancer prevention: Cancer chemo-prevention by natural compounds from common dietary spices
- Author
-
Bal L. Lokeshwar, Jie Gao, Kenza Mamouni, and Lei Zhang
- Subjects
chemistry.chemical_classification ,Cancer prevention ,Traditional medicine ,business.industry ,Clinical effectiveness ,food and beverages ,Cancer ,medicine.disease ,Flavones ,Bioavailability ,Clinical trial ,Immune system ,chemistry ,Polyphenol ,medicine ,business - Abstract
Spices play a unique role in food preparation and provide ethnic identity to cuisines. Spices are appreciated not only for their flavor but also known for their healing effects on both chronic or lethal diseases such as diabetes and cancer. A large corpus of scientific studies supports the cancer-preventive properties of spices, such as garlic, ginger, chili pepper, turmeric, cloves, cinnamon, saffron, and cardamom, the most common flavoring food items used globally. The focus of this review is on these spices that contain multiple bioactive compounds, some of which, in experimental models and human clinical studies, have shown significant anticancer and cancer preventive activities. The presence of high amounts of bioactive compounds such as alkaloids, flavones, polyphenols, triterpenes, and saponins in common spices have the potential to function as medicines that can prevent chronic metabolic diseases and even cancer. As anti-oxidants, anti-inflammatory, immune function modulators, and inducers of apoptosis in tumor cells, the bioactive compounds may be useful as single agents or as adjuvants. The molecular mechanisms of their biological action are their abilities to induce epigenetic modifications such as methylation, acetylation, ubiquitination, and suppression of lipid peroxidation. The low toxicity of these spices makes them favorable as chemopreventive agents. However, the significant therapeutic efficacy of these compounds in scientific clinical trials is still equivocal. Rigorous studies on human subjects are required to improve their bioavailability and standardize their clinical effectiveness. Such studies could define appropriate intervention strategies for maximum benefits from the dietary spices.
- Published
- 2021
36. Design, Synthesis, and Molecular Docking Studies of Curcumin Hybrid Conjugates as Potential Therapeutics for Breast Cancer
- Author
-
Siva S. Panda, Queen L. Tran, Pragya Rajpurohit, Girinath G. Pillai, Sean J. Thomas, Allison E. Bridges, Jason E. Capito, Muthusamy Thangaraju, and Bal L. Lokeshwar
- Subjects
curcumin ,DCA ,amino acid ,molecular hybridization ,molecular docking ,breast cancer ,Drug Discovery ,Pharmaceutical Science ,Molecular Medicine - Abstract
Curcumin (CUR) has received great attention over the past two decades due to its anticancer, anti-inflammatory, and antioxidant properties. Similarly, Dichloroacetate (DCA), an pyruvate dehydrogenase kinase 1 (PKD1) inhibitor, has gained huge attention as a potential anticancer drug. However, the clinical utility of these two agents is very limited because of the poor bioavailability and unsolicited side effects, respectively. We have synthesized fusion conjugates of CUR and DCA with an amino acids linker to overcome these limitations by utilizing the molecular hybridization approach. The molecular docking studies showed the potential targets of Curcumin-Modified Conjugates (CMCs) in breast cancer cells. We synthesized six hybrid conjugates named CMC1-6. These six CMC conjugates do not show any significant toxicity in a human normal immortalized mammary epithelial cell line (MCF10A) in vitro and C57BL/6 mice in vivo. However, treatment with CMC1 and CMC2 significantly reduced the growth and clonogenic survival by colony-formation assays in several human breast cancer cells (BC). Treatment by oral gavage of a transgenic mouse BC and metastatic BC tumor-bearing mice with CMC2 significantly reduced tumor growth and metastasis. Overall, our study provides strong evidence that CUR and DCA conjugates have a significant anticancer properties at a sub-micromolar concentration and overcome the clinical limitation of using CUR and DCA as potential anticancer drugs.
- Published
- 2022
37. Atypical chemokine receptors in tumor cell growth and metastasis
- Author
-
Bal L, Lokeshwar, Georgios, Kallifatidis, and James J, Hoy
- Subjects
Neoplasms ,Animals ,Humans ,Receptors, Chemokine ,Chemokines ,Neoplasm Metastasis ,Cell Proliferation ,Signal Transduction - Abstract
Atypical chemokine receptors (ACKRs) are seven-transmembrane cell surface protein receptors expressed in immune cells, normal mesenchymal cells, and several tumor cells. As of this writing, six ACKRs have been characterized by diverse activities. They bind both cysteine-cysteine (CC) type and cysteine-X-cysteine (CXC)-type chemokines, either alone, or together with a ligand bound-functional G-protein coupled (typical) chemokine receptor. The major structural difference between ACKRs and typical chemokine receptors is the substituted DRYLAIV amino acid motif in the second intracellular loop of the ACKR. Due to this substitution, these receptors cannot bind Gαi-type G-proteins responsible for intracellular calcium mobilization and cellular chemotaxis. Although initially characterized as non-signaling transmembrane receptors (decoy receptors) that attenuate ligand-induced signaling by GPCRs, studies of all ACKRs have shown ligand-independent and ligand-dependent transmembrane signaling in both non-tumor and tumor cells. The precise function and mechanism of the differential expression of ACKRs in many tumors are not understood well. The use of antagonists of ACKRs ligands has shown limited antitumor potential; however, depleting ACKR expression resulted in a reduction in experimental tumor growth and metastasis. The ACKRs represent a unique class of transmembrane signaling proteins that regulate growth, survival, and metastatic processes in tumor cells, affecting multiple pathways of tumor growth. Therefore, closer investigations of ACKRs have a high potential for identifying therapeutics which affect the intracellular signaling, preferentially via the ligand-independent mechanism.
- Published
- 2020
38. Atypical chemokine receptors in tumor cell growth and metastasis
- Author
-
Bal L. Lokeshwar, Georgios Kallifatidis, and James J. Hoy
- Subjects
Chemokine ,biology ,Chemistry ,Chemotaxis ,Cell biology ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,Cell surface receptor ,030220 oncology & carcinogenesis ,biology.protein ,Decoy receptors ,Receptor ,Intracellular ,G protein-coupled receptor - Abstract
Atypical chemokine receptors (ACKRs) are seven-transmembrane cell surface protein receptors expressed in immune cells, normal mesenchymal cells, and several tumor cells. As of this writing, six ACKRs have been characterized by diverse activities. They bind both cysteine-cysteine (CC) type and cysteine-X-cysteine (CXC)-type chemokines, either alone, or together with a ligand bound-functional G-protein coupled (typical) chemokine receptor. The major structural difference between ACKRs and typical chemokine receptors is the substituted DRYLAIV amino acid motif in the second intracellular loop of the ACKR. Due to this substitution, these receptors cannot bind Gαi-type G-proteins responsible for intracellular calcium mobilization and cellular chemotaxis. Although initially characterized as non-signaling transmembrane receptors (decoy receptors) that attenuate ligand-induced signaling by GPCRs, studies of all ACKRs have shown ligand-independent and ligand-dependent transmembrane signaling in both non-tumor and tumor cells. The precise function and mechanism of the differential expression of ACKRs in many tumors are not understood well. The use of antagonists of ACKRs ligands has shown limited antitumor potential; however, depleting ACKR expression resulted in a reduction in experimental tumor growth and metastasis. The ACKRs represent a unique class of transmembrane signaling proteins that regulate growth, survival, and metastatic processes in tumor cells, affecting multiple pathways of tumor growth. Therefore, closer investigations of ACKRs have a high potential for identifying therapeutics which affect the intracellular signaling, preferentially via the ligand-independent mechanism.
- Published
- 2020
39. Sténose carotidienne post-irradiation cervicale : une approche personnalisée
- Author
-
J.M Bartoli, M. Gaudry, Omnes, Bal L, De Masi M, P. Piquet, and Baptiste David
- Subjects
medicine.medical_specialty ,business.industry ,Carotid arteries ,Technical success ,Radiation induced ,030204 cardiovascular system & hematology ,medicine.disease ,Surgery ,03 medical and health sciences ,Stenosis ,0302 clinical medicine ,Restenosis ,Cranial Nerve Injury ,Medicine ,Myocardial infarction ,Cardiology and Cardiovascular Medicine ,business ,Stroke ,030217 neurology & neurosurgery - Abstract
Introduction Surgical treatment of radio-induced carotid stenosis (RICS) is challenging and burdened by an elevated risk of local complications. Carotid artery stenting (CAS) may be a suitable alternative. The best approach is yet to be defined. We reviewed the results of both techniques following selection based on better-suitability characteristics (anatomic and clinical). Methods We retrospectively reviewed 38 patients treated for 43 RICS from a group of 1230 patients who had carotid interventions between 2008 and 2015 (5 bilateral). Primary endpoints were morbidity and mortality at 30 days (transient ischemic attack, stroke, myocardial infarction, or death). Secondary endpoints were technical success, wound complications, cranial nerve injury (CNI), restenosis (≥50%) and recurrent symptoms. Results RICS was symptomatic in 6 patients in the OR group and 3 in the CAS group. Lesions in the OR group were longer (P=0.02) and more calcified (P=0.08). Technical success rate was 100%. Cranial nerve injury rate was 14.2% (3/21). All injuries were completely resolved within several weeks. In the CAS group, technical success rate was 95% (21/22) with the one failure due to a residual stenosis exceeding 30%. Periprocedural stroke rates were 0% and 4.5% in the OR and CAS groups respectively (0/21 vs 1/22, P=0.32). There were no early deaths. Mean follow-up was 19.1 months (3-75). The restenosis rate was 9.5% (2/21) in the OR group and 9% (2/22) in the CAS group. Conclusion Our results do not support a preferred treatment strategy. The choice of treatment should be considered on an individual basis.
- Published
- 2017
40. A Novel Splice Variant of HYAL-4 Drives Malignant Transformation and Predicts Outcome in Patients with Bladder Cancer
- Author
-
Andre R. Jordan, Sravan Kavuri, Travis Yates, Murugesan Manoharan, Marie C. Hupe, Rohitha Baskar, Zachary Klaassen, Martin J.P. Hennig, Bal L. Lokeshwar, Jiaojiao Wang, Luis E. Lopez, Ronny R. Racine, Sarrah L. Hasanali, Vinata B. Lokeshwar, Santu Ghosh, Diogo O. Escudero, Neetika Dhir, Daley S. Morera, Kelly Hoye, Soum D. Lokeshwar, Judith Knapp, and Ijeoma Azih
- Subjects
0301 basic medicine ,Cancer Research ,Hyaluronoglucosaminidase ,Apoptosis ,medicine.disease_cause ,Article ,Metastasis ,Malignant transformation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,Neoplasm Invasiveness ,Bladder cancer ,biology ,CD44 ,Cancer ,medicine.disease ,Prognosis ,Phenotype ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Alternative Splicing ,Disease Models, Animal ,030104 developmental biology ,Cell Transformation, Neoplastic ,Oncology ,Urinary Bladder Neoplasms ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Disease Progression ,Heterografts ,Carcinogenesis - Abstract
Purpose: Poor prognosis of patients with muscle-invasive bladder cancer that often metastasizes drives the need for discovery of molecular determinants of bladder cancer progression. Chondroitin sulfate proteoglycans, including CD44, regulate cancer progression; however, the identity of a chondroitinase (Chase) that cleaves chondroitin sulfate from proteoglycans is unknown. HYAL-4 is an understudied gene suspected to encode a Chase, with no known biological function. We evaluated HYAL-4 expression and its role in bladder cancer. Experimental Design: In clinical specimens, HYAL-4 wild-type (Wt) and V1 expression was evaluated by RT-qPCR, IHC, and/or immunoblotting; a novel assay measured Chase activity. Wt and V1 were stably expressed or silenced in normal urothelial and three bladder cancer cell lines. Transfectants were analyzed for stem cell phenotype, invasive signature and tumorigenesis, and metastasis in four xenograft models, including orthotopic bladder. Results: HYAL-4 expression, specifically a novel splice variant (V1), was elevated in bladder tumors; Wt expression was barely detectable. V1 encoded a truncated 349 amino acid protein that was secreted. In bladder cancer tissues, V1 levels associated with metastasis and cancer-specific survival with high efficacy and encoded Chase activity. V1 cleaved chondroitin-6-sulfate from CD44, increasing CD44 secretion. V1 induced stem cell phenotype, motility/invasion, and an invasive signature. CD44 knockdown abrogated these phenotypes. V1-expressing urothelial cells developed angiogenic, muscle-invasive tumors. V1-expressing bladder cancer cells formed tumors at low density and formed metastatic bladder tumors when implanted orthotopically. Conclusions: Our study discovered the first naturally-occurring eukaryotic/human Chase and connected it to disease pathology, specifically cancer. V1-Chase is a driver of malignant bladder cancer and potential predictor of outcome in patients with bladder cancer.
- Published
- 2019
41. G protein βγ translocation to the Golgi activates the mitogen‐activated protein kinases via phosphoinositide 3‐kinase γ
- Author
-
Nevin A. Lambert, Zhe Wei, Wei Huang, Bal L. Lokeshwar, Mostafa Khater, and Guangyu Wu
- Subjects
Phosphoinositide 3-kinase ,biology ,G protein ,Chemistry ,Kinase ,Mitogen-activated protein ,Chromosomal translocation ,Golgi apparatus ,Biochemistry ,Cell biology ,symbols.namesake ,Genetics ,symbols ,biology.protein ,Molecular Biology ,Biotechnology - Published
- 2020
42. Inhibition of androgen receptor promotes CXC-chemokine receptor 7-mediated prostate cancer cell survival
- Author
-
Diandra K. Smith, Georgios Kallifatidis, James J. Hoy, and Bal L. Lokeshwar
- Subjects
Male ,0301 basic medicine ,Cell Survival ,Science ,Article ,Androgen deprivation therapy ,03 medical and health sciences ,Chemokine receptor ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,LNCaP ,Androgen Receptor Antagonists ,medicine ,Humans ,CXC chemokine receptors ,Epidermal growth factor receptor ,Cell Proliferation ,Receptors, CXCR ,Multidisciplinary ,biology ,Prostatic Neoplasms ,medicine.disease ,3. Good health ,Androgen receptor ,030104 developmental biology ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Medicine - Abstract
The atypical C-X-C chemokine receptor 7 (CXCR7) has been implicated in supporting aggressive cancer phenotypes in several cancers including prostate cancer. However, the mechanisms driving overexpression of this receptor in cancer are poorly understood. This study investigates the role of androgen receptor (AR) in regulating CXCR7. Androgen deprivation or AR inhibition significantly increased CXCR7 expression in androgen-responsive prostate cancer cell lines, which was accompanied by enhanced epidermal growth factor receptor (EGFR)-mediated mitogenic signaling, promoting tumor cell survival through an androgen-independent signaling program. Using multiple approaches we demonstrate that AR directly binds to the CXCR7 promoter, suppressing transcription. Clustered regularly interspaced short palindromic repeats (CRISPR) directed Cas9 nuclease-mediated gene editing of CXCR7 revealed that prostate cancer cells depend on CXCR7 for proliferation, survival and clonogenic potential. Loss of CXCR7 expression by CRISPR-Cas9 gene editing resulted in a halt of cell proliferation, severely impaired EGFR signaling and the onset of cellular senescence. Characterization of a mutated CXCR7-expressing LNCaP cell clone showed altered intracellular signaling and reduced spheroid formation potential. Our results demonstrate that CXCR7 is a potential target for adjuvant therapy in combination with androgen deprivation therapy (ADT) to prevent androgen-independent tumor cell survival.
- Published
- 2017
43. Abstract 5069: Breast cancer prevention by triterpenoids from allspice
- Author
-
Jie Gao, Kenza Mamouni, Georgios Kallifatidis, Siva Panda, Muthusamy Thangaraju, and Bal L. Lokeshwar
- Subjects
Cancer Research ,Oncology - Abstract
Breast cancer ranks second as a lethal cancer in women. Although survival following initial diagnosis is ~ 100% in first five years, cancer progression and mortality is imminent in subsequent years. The slow progression to the lethal form of breast cancer has prompted development of multiple avenues to delay the progression, metastasis and mortality using potent prevention strategies, including the use of nutraceuticals. Oleanolic acid (OA) and ursolic acid (UA) are two triterpenoids found in edible plant parts-fruits and seeds with potent cancer preventive, and selective cytotoxic activities against multiple cancers including breast cancer. We conducted cytotoxic assays of the combination of OA and UA. We found the combination has enhanced efficacy as compared to OA or UA alone. The combination of OA and UA and UA alone caused cell death by increased autophagy but not apoptosis in both MCF7 and MB231 human breast cancer cells. Further analysis revealed increased autopagosomes and autophagic flux, inhibition of either process reduced cytotoxicity, indicating cytotoxic autophagy is the primary mechanism of their action. Therefore, a combination of OA and UA with conventional therapies could enhance their therapeutic efficacy while limiting systemic toxicities of existing therapies. Citation Format: Jie Gao, Kenza Mamouni, Georgios Kallifatidis, Siva Panda, Muthusamy Thangaraju, Bal L. Lokeshwar. Breast cancer prevention by triterpenoids from allspice [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 5069.
- Published
- 2019
44. Abstract 123A: The critical role of interleukin-8 chemokine axis in the development of benign prostatic hyperplasia (BPH)
- Author
-
Diandra K. Smith, Natasha Venugopal, Martha K. Terris, and Bal L. Lokeshwar
- Subjects
Cancer Research ,Oncology - Abstract
Benign prostatic hyperplasia (BPH), a non-malignant proliferative disease of the prostate, has a global incidence of over 210 million men aged 50 years or older. BPH is essentially a non-cancerous (non-malignant tumor) with significant morbidity. The severity varies from mild urinary retention to severe lower urinary tract symptoms (LUTS) accounting for greater than 50% of hospital visits for difficulty with urination, retention, and pain, and close to 40% of the prostate surgeries. BPH strongly associates with prostate volume that begins to increase in men aged 40 or higher. We investigated the role of chemokine and chemokine receptors in three immortalized cell lines and primary cultures derived from fresh BPH tissue following transurethral resection of the prostate. We analyzed the levels of chemokines and chemokine receptors secreted in the culture medium and in the mRNA from cells by qPCR, western blotting and Cytokine arrays. The epithelial identity of primary cultures was established by analyzing cytokeratin expression. Two natural products isolated from Allspice were identified that inhibit the growth of BPH cultures. A cell line identified as the BPH progenitor, a BPH intermediary cell line and a cell line with frank BPH features secreted a large excess of chemokine, predominantly CXCL-VIII or Interleukin-8 (IL-8). Unusually high amount of IL-8 was secreted by BPH progenitor cells and intermediate cells as compared to a stable normal prostate cell line (RWPE1) derived from the peripheral zone of the prostate, which did not secrete any IL-8 or express its receptor. The investigation found that expression of CXC receptors CXCR1, CXCR2 and an IL-8 inducible CXCR (CXCR7) was absent in progenitor cells and in intermediate cells, but was highly expressed in the cell line derived from BPH tissue and more significantly, in primary cultures of BPH tissue. Further, primary cultures and progenitor cells expressed high levels of immune stimulatory factors indicating PMN and monocyte infiltration leading to potential chronic inflammation of the transitional zone of the prostate. The natural products from Allspice inhibited all members of the IL-8 axis and proliferation, suggesting their potential preventative potential for BPH. These results demonstrate how a novel chemokine axis is likely to enhance pathogenesis of BPH and suggest several avenues to control the abnormal growth of prostate in aging male. Note: This abstract was not presented at the meeting. Citation Format: Diandra K. Smith, Natasha Venugopal, Martha K. Terris, Bal L. Lokeshwar. The critical role of interleukin-8 chemokine axis in the development of benign prostatic hyperplasia (BPH) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 123A.
- Published
- 2019
45. Chemokines and Chemokine Receptors as Promoters of Prostate Cancer Growth and Progression
- Author
-
Nicole Salazar, Miguel Castellan, Bal L. Lokeshwar, and Samir S. Shirodkar
- Subjects
Male ,CCR1 ,Chemokine ,biology ,Cell Survival ,Chemokine receptor CCR5 ,Prostatic Neoplasms ,Article ,Gene Expression Regulation, Neoplastic ,Chemokine receptor ,Cell Transformation, Neoplastic ,Growth factor receptor ,Disease Progression ,Genetics ,biology.protein ,Cancer research ,Humans ,Receptors, Chemokine ,CCR10 ,CXC chemokine receptors ,Chemokines ,Neoplasm Metastasis ,Molecular Biology ,Signal Transduction ,CCL21 - Abstract
Prostate cancer (CaP) is estimated to be first in incidence among cancers, with more than 240,000 new cases in 2012 in the United States. Chemokines and their receptors provide survival, proliferation, and invasion characteristics to CaP cells in both primary sites of cancer and metastatic locations. The emerging data demonstrate that many chemokines and their receptors are involved in the multistep process of CaP, leading to metastasis, and, further, that these factors act cooperatively to enhance other mechanisms of tumor cell survival, growth, and metastasis. Changes of chemokine receptor cohorts may be necessary to activate tumor-promoting signals. Chemokine receptors can activate downstream effectors, such as mitogen-activated protein kinases, by complex mechanisms of ligand-dependent activation of cryptic growth factors; guanosine triphosphate–binding, protein-coupled activation of survival kinases; or transactivation of other receptors such as ErbB family members. We describe vanguard research in which more than the classic view of chemokine receptor biology was clarified. Control of chemokines and inhibition of their receptor activation may add critical tools to reduce tumor growth, especially in chemo-hormonal refractory CaP that is both currently incurable and the most aggressive form of the disease, accounting for most of the more than 28,000 annual deaths.
- Published
- 2013
46. Achyranthes aspera (Apamarg) leaf extract inhibits human pancreatic tumor growth in athymic mice by apoptosis
- Author
-
Norman H. Altman, Pochi R. Subbarayan, Mansoor M. Ahmed, Isildinha M. Reis, Sakhi Philip, Bach Ardalan, Malancha Sarkar, Bal L. Lokeshwar, Shamaladevi Nagaraja Rao, and Pradeep Kumar
- Subjects
Pathology ,medicine.medical_specialty ,Gene Expression ,Mice, Nude ,Apoptosis ,Pharmacology ,Mice ,In vivo ,Pancreatic tumor ,Pancreatic cancer ,Drug Discovery ,medicine ,Animals ,Humans ,RNA, Messenger ,Phosphorylation ,Achyranthes ,biology ,Caspase 3 ,Plant Extracts ,Achyranthes aspera ,business.industry ,Cancer ,medicine.disease ,biology.organism_classification ,Antineoplastic Agents, Phytogenic ,Xenograft Model Antitumor Assays ,Medicine, Ayurvedic ,Pancreatic Neoplasms ,Plant Leaves ,Toxicity ,business ,Proto-Oncogene Proteins c-akt ,Injections, Intraperitoneal ,Phytotherapy - Abstract
Achyranthes aspera (Family Amaranthacea) is used for cancer therapy by ayurvedic medical practitioners in India. However, due to the non formal nature of its use, there are no systematic studies validating its medicinal properties. Thus, it's utility as an anti cancer agent remains anecdotal. Earlier, we demonstrated A. aspera to exhibit time and dose-dependent preferential cytotoxicity to cultured human pancreatic cancer cells. In this report we validate in vivo anti tumor properties of A. aspera.The in vivo anti tumor activity of leaf extract (LE) was tested by intraperitoneal (IP) injections into athymic mice harboring human pancreatic tumor subcutaneous xenograft. Toxicity was monitored by recording changes in behavioral, histological, hematological and body weight parameters.Dosing LE to athymic mice by I.P. injection for 32 days showed no adverse reactions in treated mice. Compared to the control set, IP administration of LE to tumor bearing mice significantly reduced both tumor weight and volume. Gene expression analysis using Real time PCR methods revealed that LE significantly induced caspase-3 mRNA (p0.001) and suppressed expression of the pro survival kinase Akt-1 (p0.05). TUNEL assay and immunohistochemistry confirmed apoptosis induction by activation of caspase-3 and inhibiting Akt phosphorylation in treated sets. These results are in agreement with RT PCR data.Taken together, these data suggest A. aspera to have potent anti cancer property.
- Published
- 2012
47. Designing a broad-spectrum integrative approach for cancer prevention and treatment
- Author
-
Block, Keith I, Gyllenhaal, Charlotte, Lowe, Leroy, Amedei, Amedeo, Amin, ARM Ruhul, Amin, Amr, Aquilano, Katia, Arbiser, Jack, Arreola, Alexandra, Arzumanyan, Alla, Ashraf, S Salman, Azmi, Asfar S, Benencia, Fabian, Bhakta, Dipita, Bilsland, Alan, Bishayee, Anupam, Blain, Stacy W, Block, Penny B, Boosani, Chandra S, Carey, Thomas E, Carnero, Amancio, Carotenuto, Marianeve, Casey, Stephanie C, Chakrabarti, Mrinmay, Chaturvedi, Rupesh, Chen, Georgia Zhuo, Chen, Helen, Chen, Sophie, Chen, Yi Charlie, Choi, Beom K, Ciriolo, Maria Rosa, Coley, Helen M, Collins, Andrew R, Connell, Marisa, Crawford, Sarah, Curran, Colleen S, Dabrosin, Charlotta, Damia, Giovanna, Dasgupta, Santanu, DeBerardinis, Ralph J, Decker, William K, Dhawan, Punita, Diehl, Anna Mae E, Dong, Jin-Tang, Dou, Q Ping, Drew, Janice E, Elkord, Eyad, El-Rayes, Bassel, Feitelson, Mark A, Felsher, Dean W, Ferguson, Lynnette R, Fimognari, Carmela, Firestone, Gary L, Frezza, Christian, Fujii, Hiromasa, Fuster, Mark M, Generali, Daniele, Georgakilas, Alexandros G, Gieseler, Frank, Gilbertson, Michael, Green, Michelle F, Grue, Brendan, Guha, Gunjan, Halicka, Dorota, Helferich, William G, Heneberg, Petr, Hentosh, Patricia, Hirschey, Matthew D, Hofseth, Lorne J, Holcombe, Randall F, Honoki, Kanya, Hsu, Hsue-Yin, Huang, Gloria S, Jensen, Lasse D, Jiang, Wen G, Jones, Lee W, Karpowicz, Phillip A, Keith, W Nicol, Kerkar, Sid P, Khan, Gazala N, Khatami, Mahin, Ko, Young H, Kucuk, Omer, Kulathinal, Rob J, Kumar, Nagi B, Kwon, Byoung S, Le, Anne, Lea, Michael A, Lee, Ho-Young, Lichtor, Terry, Lin, Liang-Tzung, Locasale, Jason W, Lokeshwar, Bal L, Longo, Valter D, Lyssiotis, Costas A, MacKenzie, Karen L, Malhotra, Meenakshi, Marino, Maria, Martinez-Chantar, Maria L, and Matheu, Ander
- Subjects
Cancer hallmarks ,Phytochemicals ,Oncology and Carcinogenesis ,Drug Resistance ,Antineoplastic Agents ,Targeted therapy ,Genetic Heterogeneity ,Rare Diseases ,Neoplasms ,Phytogenic ,Tumor Microenvironment ,2.1 Biological and endogenous factors ,Humans ,Molecular Targeted Therapy ,Oncology & Carcinogenesis ,Aetiology ,Precision Medicine ,Integrative medicine ,Cancer ,Prevention ,Multi-targeted ,Orphan Drug ,Good Health and Well Being ,5.1 Pharmaceuticals ,Neoplasm ,Development of treatments and therapeutic interventions ,Signal Transduction - Abstract
Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.
- Published
- 2015
48. Bioactive natural products for chemoprevention and treatment of castration-resistant prostate cancer
- Author
-
James J. Hoy, Georgios Kallifatidis, and Bal L. Lokeshwar
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Palliative care ,medicine.medical_treatment ,Silibinin ,Pharmacology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,medicine ,Animals ,Anticarcinogenic Agents ,Humans ,Cancer prevention ,business.industry ,Plant Extracts ,Cancer ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Diet ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Drug Screening Assays, Antitumor ,business ,Adjuvant - Abstract
Prostate cancer (PCa), a hormonally-driven cancer, ranks first in incidence and second in cancer related mortality in men in most Western industrialized countries. Androgen and androgen receptor (AR) are the dominant modulators of PCa growth. Over the last two decades multiple advancements in screening, treatment, surveillance and palliative care of PCa have significantly increased quality of life and survival following diagnosis. However, over 20% of patients initially diagnosed with PCa still develop an aggressive and treatment-refractory disease. Prevention or treatment for hormone-refractory PCa using bioactive compounds from marine sponges, mushrooms, and edible plants either as single agents or as adjuvants to existing therapy, has not been clinically successful. Major advancements have been made in the identification, testing and modification of the existing molecular structures of natural products. Additionally, conjugation of these compounds to novel matrices has enhanced their bio-availability; a big step towards bringing natural products to clinical trials. Natural products derived from edible plants (nutraceuticals), and common folk-medicines might offer advantages over synthetic compounds due to their broader range of targets, as compared to mostly single target synthetic anticancer compounds; e.g. kinase inhibitors. The use of synthetic inhibitors or antibodies that target a single aberrant molecule in cancer cells might be in part responsible for emergence of treatment refractory cancers. Nutraceuticals that target AR signaling (epigallocatechin gallate [EGCG], curcumin, and 5α-reductase inhibitors), AR synthesis (ericifolin, capsaicin and others) or AR degradation (betulinic acid, di-indolyl diamine, sulphoraphane, silibinin and others) are prime candidates for use as adjuvant or mono-therapies. Nutraceuticals target multiple pathophysiological mechanisms involved during cancer development and progression and thus have potential to simultaneously inhibit both prostate cancer growth and metastatic progression (e.g., inhibition of angiogenesis, epithelial-mesenchymal transition (EMT) and proliferation). Given their multi-targeting properties along with relatively lower systemic toxicity, these compounds offer significant therapeutic advantages for prevention and treatment of PCa. This review emphasizes the potential application of some of the well-researched natural compounds that target AR for prevention and therapy of PCa.
- Published
- 2015
49. CXC Receptor-1 Silencing Inhibits Androgen-Independent Prostate Cancer
- Author
-
Diogo O. Escudero, Nagarajarao Shamaladevi, Dominic A. Lyn, and Bal L. Lokeshwar
- Subjects
Male ,Cancer Research ,Neoplasms, Hormone-Dependent ,Blotting, Western ,Immunoblotting ,Cell ,Apoptosis ,Cell Cycle Proteins ,Biology ,Article ,Receptors, Interleukin-8A ,Immunoenzyme Techniques ,Small hairpin RNA ,Mice ,Cyclin D1 ,RNA interference ,LNCaP ,Tumor Cells, Cultured ,medicine ,Animals ,Immunoprecipitation ,Gene Silencing ,RNA, Messenger ,Phosphorylation ,RNA, Small Interfering ,Cell Proliferation ,Membrane Potential, Mitochondrial ,Mitogen-Activated Protein Kinase 3 ,Reverse Transcriptase Polymerase Chain Reaction ,Cell growth ,Cell Cycle ,Interleukin-8 ,Prostatic Neoplasms ,Transfection ,Cell cycle ,Molecular biology ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,Mutagenesis, Site-Directed ,Cancer research - Abstract
The CXC receptor-1 (CXCR1) is a coreceptor for interleukin-8 (IL-8) and is expressed on both normal and tumor cells. The function of CXCR1 in prostate cancer was investigated by silencing its expression, using RNA interference. We established stable cell colonies of PC-3 cells, depleted of CXCR1, using lentiviral plasmids (pLK0.1puro) generating small hairpin RNA (shRNA) against CXCR1 mRNA. Stable shRNA transfectants (PLK1–PLK5) that express significantly reduced CXCR1 mRNA (≥90% down) and protein (≥43% down) or vector-only transfectants (PC-3V) were characterized. PLK cells showed reduced cell proliferation (down, ≥66%), due to cell cycle arrest at G1-S phase, decreases in Cyclin D1, CDK4, phosphorylated Rb, and extracellular signal-regulated kinase 1/2 levels compared with those in PC-3V cells. CXCR1 depletion lead to increases in spontaneous apoptosis by mitochondria-mediated intrinsic mechanism and increases in proapoptotic proteins (BAD, 40%; BAX, 12%), but decreases in antiapoptotic proteins (BCL2, down 38%; BCLxL, 20%). PLK2 cells grew as slow-growing tumors (decrease of 54%), compared with that of PC3V tumors in athymic mice. Ex vivo analyses of PLK2 tumor tissues showed reduced expression of Cyclin D1 and vascular endothelial growth factor, and increased apoptosis activity. Other IL-8–expressing prostate cancer cell lines also exhibited similar phenotypes when CXCR1 was depleted by CXCR1 shRNA transfection. In contrast to these cells, CXCR1 depletion had little effect on IL-8 ligand–deficient LNCaP cells. RNA interference rescue using mutated CXCR1 plasmids reversed the silencing effect of PLK2, thus demonstrating the specificity of phenotypic alteration by CXCR1 shRNA. These studies establish that CXCR1 promotes IL-8–mediated tumor growth. [Cancer Res 2009;69(21):8265–74]
- Published
- 2009
50. Epigenetic Regulation of HYAL-1 Hyaluronidase Expression
- Author
-
Bal L. Lokeshwar, Murugesan Manoharan, Vinata B. Lokeshwar, Luis E. Lopez, Steve Scherer, Pablo Gomez, Nevis Fregien, David J. Klumpp, Mark S. Soloway, Neetika Dhir, Judith Knapp, Mario W. Kramer, and Melissa A. McCornack
- Subjects
Messenger RNA ,Epigenetics of physical exercise ,RNA ,Cell Biology ,Epigenetics ,Methylation ,Biology ,Binding site ,Molecular Biology ,Biochemistry ,Chromatin immunoprecipitation ,Molecular biology ,Transcription factor - Abstract
HYAL-1 (hyaluronoglucosaminidase-1) belongs to the hyaluronidase family of enzymes that degrade hyaluronic acid. HYAL-1 is a marker for cancer diagnosis and a molecular determinant of tumor growth, invasion, and angiogenesis. The regulation of HYAL-1 expression is unknown. Real time reverse transcription-PCR using 11 bladder and prostate cancer cells and 69 bladder tissues showed that HYAL-1 mRNA levels are elevated 10-30-fold in cells/tissues that express high hyaluronidase activity. Although multiple transcription start sites (TSS) for HYAL-1 mRNA were detected in various tissues, the major TSS in many tissues, including bladder and prostate, was at nucleotide 27274 in the cosmid clone LUCA13 (AC002455). By analyzing the 1532 base sequence 5' to this TSS, using cloning and luciferase reporter assays, we identified a TACAAA sequence at position -31 and the minimal promoter region between nucleotides -93 and -38. Mutational analysis identified that nucleotides -73 to -50 (which include overlapping binding consensus sites for SP1, Egr-1, and AP-2), bases C(-71) and C(-59), and an NFkappaB-binding site (at position -15) are necessary for promoter activity. The chromatin immunoprecipitation assay identified that Egr-1, AP-2, and NFkappaB bind to the promoter in HYAL-1-expressing cells, whereas SP1 binds to the promoter in non-HYAL-1-expressing cells. 5-Aza-2'-deoxycytidine treatment, bisulfite DNA sequencing, and methylation-specific PCR revealed that HYAL-1 expression is regulated by methylation at C(-71) and C(-59); both Cs are part of the SP1/Egr-1-binding sites. Thus, HYAL-1 expression is epigenetically regulated by the binding of different transcription factors to the methylated and unmethylated HYAL-1 promoter.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.