1
|
Wang B, Sun D, Li H, Chen J. A bird's eye view of the potential role of NFKBIA in pan-cancer. Heliyon 2024; 10:e31204. [PMID: 38813139 PMCID: PMC11133827 DOI: 10.1016/j.heliyon.2024.e31204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
In the 21st century, cancer remains a serious threat to people's health and has become a prominent public health problem. NFKBIA is involved in the pathological process of many diseases including cancer, but its specific role in pan-cancer has not yet been fully elucidated. This study aims to deepen the understanding of cancer pathology by analyzing the potential functions of NFKBIA in pan-cancer. We used TCGA data to analyze differences of expression of NFKBIA in pan-cancer. We explored the prognostic value, clinical relevance, immune relevance, potential biological function, and diagnosis and treatment value of NFKBIA in pan-cancer through bioinformatics analysis. This study found that in pan-cancer, NFKBIA exhibits differences in expression, which correlate with the prognosis, diagnosis, treatment value and clinical and immune parameters. We have identified that Aspirin, Astaxanthin and Bardoxolone methyl are expected to play a potential therapeutic role in pan-cancer. The results of this study will help to improve our understanding of the role and potential mechanism of NFKBIA in cancer pathology, which may provide guidance for cancer-related research and clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Difang Sun
- Department of Ophthalmology, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Haifeng Li
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jinli Chen
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Bahremand K, Aghaz F, Bahrami K. Enhancing Cisplatin Efficacy with Low Toxicity in Solid Breast Cancer Cells Using pH-Charge-Reversal Sericin-Based Nanocarriers: Development, Characterization, and In Vitro Biological Assessment. ACS OMEGA 2024; 9:14017-14032. [PMID: 38560009 PMCID: PMC10976391 DOI: 10.1021/acsomega.3c09361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/23/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Platinum-based chemotherapeutic agents are widely employed in cancer treatment because of their effectiveness in targeting DNA. However, this indiscriminate action often affects both cancerous and normal cells, leading to severe side effects and highlighting the need for innovative approaches in achieving precise drug delivery. Nanotechnology presents a promising avenue for addressing these challenges. Protein-based nanocarriers exhibit promising capabilities in the realm of cancer drug delivery with silk sericin nanoparticles standing out as a leading contender. This investigation focuses on creating a sericin-based nanocarrier (SNC) featuring surface charge reversal designed to effectively transport cisplatin (Cispt-SNC) into MCF-7 breast cancer cells. Utilizing AutoDock4.2, our molecular docking analyses identified key amino acids and revealed distinctive conformational clusters, providing insights into the drug-protein interaction landscape and highlighting the potential of sericin as a carrier for controlled drug release. The careful optimization and fabrication of sericin as the carrier material were achieved through flash nanoprecipitation, a straightforward and reproducible method that is devoid of intricate equipment. The physicochemical properties of SNCs and Cispt-SNCs, particularly concerning size, surface charge, and morphology, were evaluated using dynamic light scattering (DLS) and scanning electron microscopy (SEM). Chemical and conformational analyses of the nanocarriers were conducted using Fourier-transform infrared spectroscopy (FTIR) and circular dichroism (CD), and elemental composition analysis was performed through energy-dispersive X-ray spectroscopy (EDX). This approach aimed to achieve the smallest nanoparticle size for Cispt-SNCs (180 nm) and high drug encapsulation efficiency (84%) at an optimal sericin concentration of 0.1% (w/v), maintaining a negative net charge at a physiological pH (7.4). Cellular uptake and cytotoxicity were investigated in MCF-7 breast cancer cells. SNCs demonstrated stability and exhibited a pH-dependent drug release behavior, aligning with the mildly acidic tumor microenvironment (pH 6.0-7.0). Efficient cellular uptake of Cispt-SNC, along with DNA fragmentation and chromatin condensation, was found at pH 6, leading to cell apoptosis. These results collectively indicate the potential of SNCs for achieving controlled drug release in a tumor-specific context. Our in vitro studies reveal the cytotoxicity of both cisplatin and Cispt-SNCs on MCF-7 cells. Cisplatin significantly reduced cell viability at 10 μM concentration (IC50), and the unique combination of sericin and cisplatin showcased enhanced cell viability compared to cisplatin alone, suggesting that controlled drug release is indicated by a gradient decrease in cell viability and highlighting SNCs as promising carriers. The study underscores the promise of protein-based nanocarriers in advancing targeted drug delivery for cancer therapy.
Collapse
Affiliation(s)
- Kiana Bahremand
- Nano Drug Delivery
Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Faranak Aghaz
- Nano Drug Delivery
Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Kiumars Bahrami
- Nanoscience and Nanotechnology
Research Center (NNRC), Razi University, Kermanshah 67144-14971, Iran
| |
Collapse
|
3
|
Oza PP, Kashfi K. The Triple Crown: NO, CO, and H 2S in cancer cell biology. Pharmacol Ther 2023; 249:108502. [PMID: 37517510 PMCID: PMC10529678 DOI: 10.1016/j.pharmthera.2023.108502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) are three endogenously produced gases with important functions in the vasculature, immune defense, and inflammation. It is increasingly apparent that, far from working in isolation, these three exert many effects by modulating each other's activity. Each gas is produced by three enzymes, which have some tissue specificities and can also be non-enzymatically produced by redox reactions of various substrates. Both NO and CO share similar properties, such as activating soluble guanylate cyclase (sGC) to increase cyclic guanosine monophosphate (cGMP) levels. At the same time, H2S both inhibits phosphodiesterase 5A (PDE5A), an enzyme that metabolizes sGC and exerts redox regulation on sGC. The role of NO, CO, and H2S in the setting of cancer has been quite perplexing, as there is evidence for both tumor-promoting and pro-inflammatory effects and anti-tumor and anti-inflammatory activities. Each gasotransmitter has been found to have dual effects on different aspects of cancer biology, including cancer cell proliferation and apoptosis, invasion and metastasis, angiogenesis, and immunomodulation. These seemingly contradictory actions may relate to each gas having a dual effect dependent on its local flux. In this review, we discuss the major roles of NO, CO, and H2S in the context of cancer, with an effort to highlight the dual nature of each gas in different events occurring during cancer progression.
Collapse
Affiliation(s)
- Palak P Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York 10091, USA.
| |
Collapse
|
4
|
Aghaz F, Asadi Z, Sajadimajd S, Kashfi K, Arkan E, Rahimi Z. Codelivery of resveratrol melatonin utilizing pH responsive sericin based nanocarriers inhibits the proliferation of breast cancer cell line at the different pH. Sci Rep 2023; 13:11090. [PMID: 37422485 PMCID: PMC10329705 DOI: 10.1038/s41598-023-37668-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023] Open
Abstract
Protein-based nanocarriers have demonstrated good potential for cancer drug delivery. Silk sericin nano-particle is arguably one of the best in this field. In this study, we developed a surface charge reversal sericin-based nanocarrier to co-deliver resveratrol and melatonin (MR-SNC) to MCF-7 breast cancer cells as combination therapy. MR-SNC was fabricated with various sericin concentrations via flash-nanoprecipitation as a simple and reproducible method without complicated equipment. The nanoparticles were subsequently characterized for their size, charge, morphology and shape by dynamic light scattering (DLS) and scanning electron microscope (SEM). Nanocarriers chemical and conformational analysis were done by fourier transform infrared spectroscopy (FT-IR) and circular dichroism (CD) respectively. In vitro drug release was determined at different pH values (7.45, 6.5 and 6). The cellular uptake and cytotoxicity were studies using breast cancer MCF-7 cells. MR-SNC fabricated with the lowest sericin concentration (0.1%), showed a desirable 127 nm size, with a net negative charge at physiological pH. Sericin structure was preserved entirely in the form of nano-particles. Among the three pH values we applied, the maximum in vitro drug release was at pH 6, 6.5, and 7.4, respectively. This pH dependency showed the charge reversal property of our smart nanocarrier via changing the surface charge from negative to positive in mildly acidic pH, destructing the electrostatic interactions between sericin surface amino acids. Cell viability studies demonstrated the significant toxicity of MR-SNC in MCF-7 cells at all pH values after 48 h, suggesting a synergistic effect of combination therapy with the two antioxidants. The efficient cellular uptake of MR-SNC, DNA fragmentation and chromatin condensation was found at pH 6. Nutshell, our result indicated proficient release of the entrapped drug combination from MR-SNC in an acidic environment leading to cell apoptosis. This work introduces a smart pH-responsive nano-platform for anti-breast cancer drug delivery.
Collapse
Affiliation(s)
- Faranak Aghaz
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Asadi
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soraya Sajadimajd
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | - Elham Arkan
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Zohreh Rahimi
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
5
|
Fan B, Mohammed A, Huang Y, Luo H, Zhang H, Tao S, Xu W, Liu Q, He T, Jin H, Sun M, Sun M, Yun Z, Zhao R, Wu G, Li X. Can Aspirin Use Be Associated With the Risk or Prognosis of Bladder Cancer? A Case-Control Study and Meta-analytic Assessment. Front Oncol 2021; 11:633462. [PMID: 34350107 PMCID: PMC8327774 DOI: 10.3389/fonc.2021.633462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/30/2021] [Indexed: 12/03/2022] Open
Abstract
Aspirin, widely used to prevent cardiovascular disease, had been linked to the incidence of bladder cancer (BCa). Existing studies focusing on Chinese populations are relatively rare, especially for Northeast China. Meanwhile, relevant studies on the effects of aspirin on the occurrence or prognosis of BCa are inconsistent or even controversial. First, in the case control study, logistic regression analysis was used to investigate the association between aspirin intake and risk of BCa including 1121 patients with BCa and the 2242 controls. Subsequently, Kaplan-Meier curve and Cox regression analyses were applied to explore the association between aspirin intake and clinicopathological factors which may predict overall survival (OS) and recurrence-free survival (RFS) of BCa patients. Finally, we quantificationally combined the results with those from the published literature evaluating aspirin intake and its effects on the occurrence, outcome of surgery and prognosis of BCa by meta-analysis up to May 1, 2021.Our case-control study demonstrated that the regular use of aspirin was not associated with a reduced incidence of BCa (P=0.175). Stratified analyses of sex showed that aspirin intake did not lead to a lower risk of BCa in female patients (P=0.063). However, the male population who regularly took aspirin had a lower incidence of BCa (OR=0.748, 95% CI= 0.584-0.958, P=0.021). Subgroup analyses stratified by smoking found a significant reduction in the risk of BCa in current smokers with aspirin intake (OR=0.522, 95% CI=0.342-0.797, P=0.002). In terms of prognosis of BCa, patients with a history of aspirin intake did not had a markedly longer OS or RFS than those with no history of aspirin intake by Kaplan-Meier curves. Stratified analysis by sex showed no correlation between aspirin intake and the recurrence or survival of BCa for either male or female patients. However, in people younger than 68, aspirin intake seemed to have prolonged effects for overall survival (HR=3.876; 95% CI=1.326-11.325, P=0.019). Then, we performed a meta-analysis and the combined results from 19 articles and our study involving more than 39524 BCa cases indicated that aspirin intake was not associated with the occurrence of BCa (P=0.671). Subgroup analysis by whether regular use of aspirin, by the mean duration of use of aspirin, by sex, by smoking exposure, by research region and by study type also supported the above results. In terms of the impact of aspirin intake on the prognosis of patients with BCa, 11 articles and our study involving 8825 BCa cases were eligible. The combined results showed that patients with aspirin intake did not have significantly influence on survival, recurrence, progression and metastasis than those without aspirin intake. On the whole, both our retrospective study and literature meta-analysis suggested a lack of a strong relevant association between the use of aspirin and the incidence or prognosis of BCa. Thus, additional long-term follow-up prospective research is warranted to clarify the association of aspirin with BCa incidence and prognosis.
Collapse
Affiliation(s)
- Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Alradhi Mohammed
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuanbin Huang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hong Luo
- Clinical Medicine, Dalian Medical University, Dalian, China
| | - Hongxian Zhang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shenghua Tao
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Weijiao Xu
- Clinical Medicine, Dalian Medical University, Dalian, China
| | - Qian Liu
- Medical Imaging, Dalian Medical University, Dalian, China
| | - Tao He
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huidan Jin
- Department of Anaesthesiology, Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Mengfan Sun
- Department of Pharmacy, Zhongshan College of Dalian Medical University, Dalian, China
| | - Man Sun
- Clinical Medicine, Dalian Medical University, Dalian, China
| | - Zhifei Yun
- Clinical Medicine, Dalian Medical University, Dalian, China
| | - Rui Zhao
- Department of Pharmacy, Zhongshan College of Dalian Medical University, Dalian, China
| | - Guoyu Wu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiancheng Li
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Wnt/β-Catenin Signaling Pathway as Chemotherapeutic Target in Breast Cancer: An Update on Pros and Cons. Clin Breast Cancer 2020; 20:361-370. [DOI: 10.1016/j.clbc.2020.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
|
7
|
Zhou J, Di M, Han H. Upregulation of SHIP2 participates in the development of breast cancer via promoting Wnt/β-catenin signaling. Onco Targets Ther 2019; 12:7067-7077. [PMID: 31564892 PMCID: PMC6722435 DOI: 10.2147/ott.s223422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Src homology 2-containing inositol 5-phosphatase 2 (SHIP2) gene is associated with arthrosclerosis, gastric cancer and diabetes. In this study, we revealed that overexpression of SHIP2 is closely implicated with the development of breast cancer (BC). Methods The BC tissue and adjacent cancerous tissue were obtained from BC patients who had underwent mastectomy. BC cells with either overexpression or knockdown of SHIP2 were analyzed to determine cell proliferation, migration, invasion and apoptosis using the CCK-8 assay, colony formation assay, scratch assay, transwell assay and flow cytometry, respectively. A rat BC xenograft model was constructed to explore the role of SHIP2 on tumor growth in vivo. Results The expression levels of SHIP2 in BC tissues and cells were significantly higher than those in adjacent tissues and normal breast cells, respectively. Silencing SHIP2 suppressed BC cells proliferation and promoted apoptosis. Overexpression of SHIP2 enhanced the cells migration/invasion in BC. Moreover, SHIP2 participated in the Wnt/β-catenin pathway by regulating GSK-3β and its downstream genes. β-Catenin activator LiCl could significantly eliminate the effect of si-SHIP2 on BC cells. Moreover, overexpression of SHIP2 increased tumor volume and weight in rat model, and Wnt/β-catenin pathway inhibitor ICG001 reversed the promoting effect of SHIP2 on tumorigenesis. Conclusion Upregulation of SHIP2 could increase the migration, invasion, proliferation, and decrease apoptosis in BC cells. Moreover, SHIP2 participated in the progression of BC via activating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Oncology, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, People's Republic of China
| | - Manman Di
- Department of Oncology, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, People's Republic of China
| | - Hui Han
- Department of Oncology, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, People's Republic of China
| |
Collapse
|
8
|
Guenzle J, Garrelfs NWC, Goeldner JM, Weyerbrock A. Cyclooxygenase (COX) Inhibition by Acetyl Salicylic Acid (ASA) Enhances Antitumor Effects of Nitric Oxide in Glioblastoma In Vitro. Mol Neurobiol 2019; 56:6046-6055. [DOI: 10.1007/s12035-019-1513-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/24/2019] [Indexed: 02/06/2023]
|
9
|
Ayyadevara S, Balasubramaniam M, Kakraba S, Alla R, Mehta JL, Shmookler Reis RJ. Aspirin-Mediated Acetylation Protects Against Multiple Neurodegenerative Pathologies by Impeding Protein Aggregation. Antioxid Redox Signal 2017; 27:1383-1396. [PMID: 28537433 PMCID: PMC5661865 DOI: 10.1089/ars.2016.6978] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS Many progressive neurological disorders, including Alzheimer's disease (AD), Huntington's disease, and Parkinson's disease (PD), are characterized by accumulation of insoluble protein aggregates. In prospective trials, the cyclooxygenase inhibitor aspirin (acetylsalicylic acid) reduced the risk of AD and PD, as well as cardiovascular events and many late-onset cancers. Considering the role played by protein hyperphosphorylation in aggregation and neurodegenerative diseases, and aspirin's known ability to donate acetyl groups, we asked whether aspirin might reduce both phosphorylation and aggregation by acetylating protein targets. RESULTS Aspirin was substantially more effective than salicylate in reducing or delaying aggregation in human neuroblastoma cells grown in vitro, and in Caenorhabditis elegans models of human neurodegenerative diseases in vivo. Aspirin acetylates many proteins, while reducing phosphorylation, suggesting that acetylation may oppose phosphorylation. Surprisingly, acetylated proteins were largely excluded from compact aggregates. Molecular-dynamic simulations indicate that acetylation of amyloid peptide energetically disfavors its association into dimers and octamers, and oligomers that do form are less compact and stable than those comprising unacetylated peptides. INNOVATION Hyperphosphorylation predisposes certain proteins to aggregate (e.g., tau, α-synuclein, and transactive response DNA-binding protein 43 [TDP-43]), and it is a critical pathogenic marker in both cardiovascular and neurodegenerative diseases. We present novel evidence that acetylated proteins are underrepresented in protein aggregates, and that aggregation varies inversely with acetylation propensity after diverse genetic and pharmacologic interventions. CONCLUSIONS These results are consistent with the hypothesis that aspirin inhibits protein aggregation and the ensuing toxicity of aggregates through its acetyl-donating activity. This mechanism may contribute to the neuro-protective, cardio-protective, and life-prolonging effects of aspirin. Antioxid. Redox Signal. 27, 1383-1396.
Collapse
Affiliation(s)
- Srinivas Ayyadevara
- 1 Central Arkansas Veterans Healthcare System, Research & Development Service , Little Rock, Arkansas.,2 Department of Geriatrics, Reynolds Institute on Aging, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Meenakshisundaram Balasubramaniam
- 1 Central Arkansas Veterans Healthcare System, Research & Development Service , Little Rock, Arkansas.,2 Department of Geriatrics, Reynolds Institute on Aging, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Samuel Kakraba
- 3 Bioinformatics Program, University of Arkansas for Medical Sciences, and University of Arkansas at Little Rock , Little Rock, Arkansas
| | - Ramani Alla
- 1 Central Arkansas Veterans Healthcare System, Research & Development Service , Little Rock, Arkansas.,2 Department of Geriatrics, Reynolds Institute on Aging, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Jawahar L Mehta
- 1 Central Arkansas Veterans Healthcare System, Research & Development Service , Little Rock, Arkansas.,4 Divison of Cardiology, Department of Medicine, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Robert J Shmookler Reis
- 1 Central Arkansas Veterans Healthcare System, Research & Development Service , Little Rock, Arkansas.,2 Department of Geriatrics, Reynolds Institute on Aging, University of Arkansas for Medical Sciences , Little Rock, Arkansas.,3 Bioinformatics Program, University of Arkansas for Medical Sciences, and University of Arkansas at Little Rock , Little Rock, Arkansas
| |
Collapse
|
10
|
Plenchette S, Romagny S, Laurens V, Bettaieb A. [NO and cancer: itinerary of a double agent]. Med Sci (Paris) 2016; 32:625-33. [PMID: 27406774 DOI: 10.1051/medsci/20163206027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Protein S-nitrosylation is now recognized as a ubiquitous regulatory mechanism. Like any post-translational modifications, S-nitrosylation is critical for the control of numerous cellular processes. It is now clear that S-nitrosylation is playing a double game, enhancing or inhibiting the tumor growth or the induction of cell death. Thanks to research aimed at demonstrating NO cytotoxic effects, new therapeutic strategies based on NO donor drugs have emerged. Although therapeutic NO donors can target a large number of proteins, the cellular mechanism is still not fully understood. This review reflects the current state of knowledge on S-nitrosylated proteins that take part of the oncogenic and apoptotic signaling, putting forward proteins with potential interest in cancer therapy.
Collapse
Affiliation(s)
- Stéphanie Plenchette
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Sabrina Romagny
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Véronique Laurens
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Ali Bettaieb
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| |
Collapse
|
11
|
Nitric Oxide-Releasing Aspirin Suppresses NF-κB Signaling in Estrogen Receptor Negative Breast Cancer Cells in Vitro and in Vivo. Molecules 2015; 20:12481-99. [PMID: 26184135 PMCID: PMC6331965 DOI: 10.3390/molecules200712481] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/11/2015] [Accepted: 07/06/2015] [Indexed: 01/29/2023] Open
Abstract
Estrogen receptor negative (ER(-)) breast cancer is aggressive, responds poorly to current treatments and has a poor prognosis. The NF-κB signaling pathway is implicated in ER(-) tumorigenesis. Aspirin (ASA) is chemopreventive against ER(+) but not for ER(-) breast cancers. Nitric oxide-releasing aspirin (NO-ASA) is a safer ASA where ASA is linked to an NO-releasing moiety through a spacer. In vitro, we investigated anti-proliferation effects of NO-ASA (para- and meta-isomers) against ER(-) breast cancer cells MDA-MB-231 and SK-BR-23, effects on NF-κB signaling, and reactive oxygen species by standard techniques. In vivo, effects of NO-ASA were evaluated in a mouse xenograft model using MDA-MB-231 cells. p-NO-ASA inhibited the growth of MDA-MB-231 and SK-BR-3 cells at 24 h, the respective IC50s were 13 ± 2 and 17 ± 2 μM; ASA had an IC50 of >3000 μM in both cell lines. The IC50s for m-NO-ASA in MDA-MB-231 and SK-BR-3 were 173 ± 15 and 185 ± 12 μM, respectively, therefore, implying p-NO-ASA as a stronger inhibitor of growth p-NO-ASA reduced cell growth by inhibiting proliferation, inducing apoptosis and causing G0/G1 cell cycle block. Activation of NF-κB was inhibited by both isomers as demonstrated by decreases in NF-κB-DNA binding and luciferase activity at 24 h, However, m-NO-ASA produced transient effects at 3 h such as increased NF-κB-DNA-binding, increased levels of nuclear p50, even though both isomers inhibited IκB degradation. Increase in nuclear p50 by m-NO-ASA was associated with translocation of p50 in to the nucleus as observed by immunoflouresence at 3 h. NO-ASA induced reactive oxygen species (ROS) as evidenced by overall increases in both H2DCFDA (2',7'-dichlorodihydrofluorescein) and DHE (dihydroethidium)-derived fluorescence. Inhibition of ROS by N-acetyl-cysteine reversed the m-NO-ASA-mediated translocation of p50 in to the nucleus. In xenografts, p-NO-ASA inhibited tumor growth by inhibiting proliferation (PCNA and tumor volume), inducing apoptosis (TUNEL positive cells) and reducing NF-κB expression. Both isomers inhibit cancer cells, inhibit NF-κB pathway and induce ROS, and have potential as anticancer compounds.
Collapse
|
12
|
Basudhar D, Cheng RC, Bharadwaj G, Ridnour LA, Wink DA, Miranda KM. Chemotherapeutic potential of diazeniumdiolate-based aspirin prodrugs in breast cancer. Free Radic Biol Med 2015; 83:101-14. [PMID: 25659932 PMCID: PMC4441830 DOI: 10.1016/j.freeradbiomed.2015.01.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/05/2015] [Accepted: 01/13/2015] [Indexed: 12/12/2022]
Abstract
Diazeniumdiolate-based aspirin prodrugs have previously been shown to retain the anti-inflammatory properties of aspirin while protecting against the common side effect of stomach ulceration. Initial analysis of two new prodrugs of aspirin that also release either nitroxyl (HNO) or nitric oxide (NO) demonstrated increased cytotoxicity toward human lung carcinoma cells compared to either aspirin or the parent nitrogen oxide donor. In addition, cytotoxicity was significantly lower in endothelial cells, suggesting cancer-specific sensitivity. To assess the chemotherapeutic potential of these new prodrugs in treatment of breast cancer, we studied their effect both in cultured cells and in a nude mouse model. Both prodrugs reduced growth of breast adenocarcinoma cells more effectively than the parent compounds while not being appreciably cytotoxic in a related nontumorigenic cell line (MCF-10A). The HNO donor also was more cytotoxic than the related NO donor. The basis for the observed specificity was investigated in terms of impact on metabolism, DNA damage and repair, apoptosis, angiogenesis and metastasis. The results suggest a significant pharmacological potential for treatment of breast cancer.
Collapse
Affiliation(s)
- Debashree Basudhar
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Robert C Cheng
- Radiation Biology Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gaurav Bharadwaj
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Lisa A Ridnour
- Radiation Biology Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A Wink
- Radiation Biology Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katrina M Miranda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
13
|
Basudhar D, Bharadwaj G, Cheng RY, Jain S, Shi S, Heinecke JL, Holland RJ, Ridnour LA, Caceres VM, Spadari-Bratfisch RC, Paolocci N, Velázquez-Martínez CA, Wink DA, Miranda KM. Synthesis and chemical and biological comparison of nitroxyl- and nitric oxide-releasing diazeniumdiolate-based aspirin derivatives. J Med Chem 2013; 56:7804-20. [PMID: 24102516 DOI: 10.1021/jm400196q] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Structural modifications of nonsteroidal anti-inflammatory drugs (NSAIDs) have successfully reduced the side effect of gastrointestinal ulceration without affecting anti-inflammatory activity, but they may increase the risk of myocardial infarction with chronic use. The fact that nitroxyl (HNO) reduces platelet aggregation, preconditions against myocardial infarction, and enhances contractility led us to synthesize a diazeniumdiolate-based HNO-releasing aspirin and to compare it to an NO-releasing analogue. Here, the decomposition mechanisms are described for these compounds. In addition to protection against stomach ulceration, these prodrugs exhibited significantly enhanced cytotoxcity compared to either aspirin or the parent diazeniumdiolate toward nonsmall cell lung carcinoma cells (A549), but they were not appreciably toxic toward endothelial cells (HUVECs). The HNO-NSAID prodrug inhibited cylcooxgenase-2 and glyceraldehyde 3-phosphate dehydrogenase activity and triggered significant sarcomere shortening on murine ventricular myocytes compared to control. Together, these anti-inflammatory, antineoplasic, and contractile properties suggest the potential of HNO-NSAIDs in the treatment of inflammation, cancer, or heart failure.
Collapse
Affiliation(s)
- Debashree Basudhar
- Department of Chemistry and Biochemistry, University of Arizona , Tucson, Arizona 85721, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
The proapoptotic effect of traditional and novel nonsteroidal anti-inflammatory drugs in mammalian and yeast cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:504230. [PMID: 23983899 PMCID: PMC3747411 DOI: 10.1155/2013/504230] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/08/2013] [Indexed: 12/16/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have long been used to treat pain, fever, and inflammation. However, mounting evidence shows that NSAIDs, such as aspirin, have very promising antineoplastic properties. The chemopreventive, antiproliferative behaviour of NSAIDs has been associated with both their inactivation of cyclooxygenases (COX) and their ability to induce apoptosis via pathways that are largely COX-independent. In this review, the various proapoptotic pathways induced by traditional and novel NSAIDs such as phospho-NSAIDs, hydrogen sulfide-releasing NSAIDs and nitric oxide-releasing NSAIDs in mammalian cell lines are discussed, as well as the proapoptotic effects of NSAIDs on budding yeast which retains the hallmarks of mammalian apoptosis. The significance of these mechanisms in terms of the role of NSAIDs in effective cancer prevention is considered.
Collapse
|
15
|
Laschak M, Spindler KD, Schrader AJ, Hessenauer A, Streicher W, Schrader M, Cronauer MV. JS-K, a glutathione/glutathione S-transferase-activated nitric oxide releasing prodrug inhibits androgen receptor and WNT-signaling in prostate cancer cells. BMC Cancer 2012; 12:130. [PMID: 22462810 PMCID: PMC3376035 DOI: 10.1186/1471-2407-12-130] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 03/30/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Nitric oxide (NO) and its oxidative reaction products have been repeatedly shown to block steroid receptor function via nitrosation of zinc finger structures in the DNA-binding domain (DBD). In consequence NO-donors could be of special interest for the treatment of deregulated androgen receptor(AR)-signaling in castration resistant prostate cancer (CRPC). METHODS Prostate cancer (PCa) cells were treated with JS-K, a diazeniumdiolate derivate capable of generating large amounts of intracellular NO following activation by glutathione S-transferase. Generation of NO was determined indirectly by the detection of nitrate in tissue culture medium or by immunodetection of nitrotyrosine in the cytoplasm. Effects of JS-K on intracellular AR-levels were determined by western blotting. AR-dimerization was analyzed by mammalian two hybrid assay, nuclear translocation of the AR was visualized in PCa cells transfected with a green fluorescent AR-Eos fusion protein using fluorescence microscopy. Modulation of AR- and WNT-signalling by JS-K was investigated using reporter gene assays. Tumor cell proliferation following JS-K treatment was measured by MTT-Assay. RESULTS The NO-releasing compound JS-K was shown to inhibit AR-mediated reporter gene activity in 22Rv1 CRPC cells. Inhibition of AR signaling was neither due to an inhibition of nuclear import nor to a reduction in AR-dimerization. In contrast to previously tested NO-donors, JS-K was able to reduce the intracellular concentration of functional AR. This could be attributed to the generation of extremely high intracellular levels of the free radical NO as demonstrated indirectly by high levels of nitrotyrosine in JS-K treated cells. Moreover, JS-K diminished WNT-signaling in AR-positive 22Rv1 cells. In line with these observations, castration resistant 22Rv1 cells were found to be more susceptible to the growth inhibitory effects of JS-K than the androgen dependent LNCaP which do not exhibit an active WNT-signaling pathway. CONCLUSIONS Our results suggest that small molecules able to inhibit WNT- and AR-signaling via NO-release represent a promising platform for the development of new compounds for the treatment of CRPC.
Collapse
Affiliation(s)
- Martin Laschak
- Department of Urology, Ulm University, Prittwitzstrasse 43, 89075, Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
16
|
Chattopadhyay M, Kodela R, Nath N, Dastagirzada YM, Velázquez-Martínez CA, Boring D, Kashfi K. Hydrogen sulfide-releasing NSAIDs inhibit the growth of human cancer cells: a general property and evidence of a tissue type-independent effect. Biochem Pharmacol 2011; 83:715-22. [PMID: 22222427 DOI: 10.1016/j.bcp.2011.12.018] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 12/09/2011] [Accepted: 12/15/2011] [Indexed: 12/21/2022]
Abstract
Hydrogen sulfide-releasing non-steroidal anti-inflammatory drugs (HS-NSAIDs) are an emerging novel class of compounds with significant anti-inflammatory properties. They consist of a traditional NSAID to which an H(2)S-releasing moiety is covalently attached. We examined the effects of four different HS-NSAIDs on the growth properties of eleven different human cancer cell lines of six different tissue origins. Human colon, breast, pancreatic, prostate, lung, and leukemia cancer cell lines were treated with HS-aspirin, -sulindac, -iburofen, -naproxen, and their traditional counterparts. HS-NSAIDs inhibited the growth of all cancer cell lines studied, with potencies of 28- to >3000-fold greater than that of their traditional counterparts. HS-aspirin (HS-ASA) was consistently the most potent. HS-NSAIDs inhibited cell proliferation, induced apoptosis, and caused G(0)/G(1) cell cycle block. Metabolism of HS-ASA by colon cells showed that the acetyl group of ASA was hydrolyzed rapidly, followed by hydrolysis of the ester bond linking the salicylate anion to the H(2)S releasing moiety, producing salicylic acid and ADT-OH from which H(2)S is released. In reconstitution studies, ASA and ADT-OH were individually less active than the intact HS-ASA towards cell growth inhibition. Additionally, the combination of these two components representing a fairly close approximation to the intact HS-ASA, was 95-fold less active than the intact HS-ASA for growth inhibition. Taken together, these results demonstrate that HS-NSAIDs have potential anti-growth activity against a wide variety of human cancer cells.
Collapse
Affiliation(s)
- Mitali Chattopadhyay
- Department of Physiology and Pharmacology, Sophie Davis School of Biomedical Education, City University of New York Medical School, New York, NY 10031, United States
| | | | | | | | | | | | | |
Collapse
|
17
|
Chattopadhyay M, Kodela R, Nath N, Barsegian A, Boring D, Kashfi K. Hydrogen sulfide-releasing aspirin suppresses NF-κB signaling in estrogen receptor negative breast cancer cells in vitro and in vivo. Biochem Pharmacol 2011; 83:723-32. [PMID: 22209867 DOI: 10.1016/j.bcp.2011.12.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 12/09/2011] [Accepted: 12/15/2011] [Indexed: 01/04/2023]
Abstract
Hormone-dependent estrogen receptor positive (ER+) breast cancers generally respond well to anti-estrogen therapy. Unfortunately, hormone-independent estrogen receptor negative (ER-) breast cancers are aggressive, respond poorly to current treatments and have a poor prognosis. New approaches and targets are needed for the prevention and treatment of ER- breast cancer. The NF-κB signaling pathway is strongly implicated in ER- tumor genesis, constituting a possible target for treatment. Hydrogen sulfide-releasing aspirin (HS-ASA), a novel and safer derivative of aspirin, has shown promise as an anti-cancer agent. We examined the growth inhibitory effect of HS-ASA via alterations in cell proliferation, cell cycle phase transitions, and apoptosis, using MDA-MB-231 cells as a model of triple negative breast cancer. Tumor xenografts in mice, representing human ER- breast cancer, were evaluated for reduction in tumor size, followed by immunohistochemical analysis for proliferation, apoptosis and expression of NF-κB. HS-ASA suppressed the growth of MDA-MB-231 cells by induction of G(0)/G(1) arrest and apoptosis, down-regulation of NF-κB, reduction of thioredoxin reductase activity, and increased levels reactive oxygen species. Tumor xenografts in mice, were significantly reduced in volume and mass by HS-ASA treatment. The decrease in tumor mass was associated with inhibition of cell proliferation, induction of apoptosis and decrease in NF-κB levels in vivo. HS-ASA has anti-cancer potential against ER- breast cancer and merits further study.
Collapse
Affiliation(s)
- Mitali Chattopadhyay
- Department of Physiology and Pharmacology, Sophie Davis School of Biomedical Education, City University of New York Medical School, New York, NY 10031, United States
| | | | | | | | | | | |
Collapse
|
18
|
Gehrke I, Razavi R, Poll-Wolbeck SJ, Berkessel A, Hallek M, Kreuzer KA. The Antineoplastic Effect of Nitric Oxide-Donating Acetylsalicylic Acid (NO-ASA) in Chronic Lymphocytic Leukemia (CLL) Cells is Highly Dependent on its Positional Isomerism. Ther Adv Hematol 2011; 2:279-89. [PMID: 23556096 DOI: 10.1177/2040620711416272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Chronic Lymphocytic Leukemia (CLL) is not curable in patients that are not eligible for allogeneic stem cell transplantation. Therefore, new treatment options are highly desirable. Chemically modified nonsteroidal anti-inflammatory drugs (NSAIDs), such as nitric-oxide-donating acetylsalicylic acid (NO-ASA), have been described to possess antineoplastic capacity. Recently, we could demonstrate a potent apoptosis induction in primary CLL cells in vitro and tumor growth inhibition by para-NO-ASA in a xenograft mouse model. However, little is known about the impact of positional isomerism of NO-ASA on its antineoplastic capacity in CLL. METHODS Primary CLL cells were treated with the meta-or para-isomer of NO-ASA at varying concentrations and durations. Viability was assessed flow cytometrically by annexin V-FITC/PI staining and by CellTiter-Glo luminescence cell viability assay. Caspase and PARP cleavage as well as involvement of β-catenin/Lef-1 signaling was determined by immunoblotting. For caspase inhibition, BD™ ApoBlock was used. Nude mice were xenografted with JVM3 cells and treated with meta-NO-ASA, para-NO-ASA or vehicle control. RESULTS The meta-isomer was entirely ineffective in inducing CLL cell apoptosis in concentrations up to 100 μM, while para-NO-ASA acted in the low micromolar range. meta-NO-ASA, in contrast to para-NO-ASA, did not alter caspase activity. While para-NO-ASA action involved inhibition of β-catenin/Lef-1 signaling, meta-NO-ASA did not show any impact on this signaling pathway. Further, meta-NO-ASA did not significantly reduce tumor growth in a CLL xenograft mouse model, while para-NO-ASA was highly potent. CONCLUSION We conclude that positional isomerism is crucial for the antineoplastic effect of NO-ASA in CLL. It can be suggested that the para-isomer, but not the meta-isomer, generates a chemical structure which is essential for the neoplastic effect of NO-ASA.
Collapse
Affiliation(s)
- Iris Gehrke
- Department I of Internal Medicine, University at Cologne, Cologne, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Kodela R, Chattopadhyay M, Nath N, Cieciura LZ, Pospishill L, Boring D, Crowell JA, Kashfi K. Synthesis and biological activity of acetyl-protected hydroxybenzyl diethyl phosphates (EHBP) as potential chemotherapeutic agents. Bioorg Med Chem Lett 2011; 21:7146-50. [PMID: 22001089 DOI: 10.1016/j.bmcl.2011.09.075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/15/2011] [Accepted: 09/19/2011] [Indexed: 12/22/2022]
Abstract
Several acetyl-protected hydroxybenzyl diethyl phosphates (EHBPs) that are capable of forming quinone methide intermediates were synthesized and their cell growth inhibitory properties were evaluated in four different human cancer cell lines. Compounds 1, 1a, and 1b, corresponding to (4-acetyloxybenzyl diethylphosphate), (3-methyl-4-acetyloxybenzyl diethylphosphate), and (3-chloro-4-acetyloxybenzyl diethylphosphate), were significantly more potent than compounds 2 and 3, (2-acetyloxybenzyl diethylphosphate) and (3-acetyloxybenzyl diethylphosphate), respectively. Using HT-29 human colon cancer cells, compounds 1 and 3 increased apoptosis, inhibited proliferation, and caused a G(2)/M block in the cell cycle. Our data suggest that these compounds merit further investigation as potential anti-cancer agents.
Collapse
Affiliation(s)
- Ravinder Kodela
- Department of Physiology and Pharmacology, City University of New York Medical School, New York, NY 10031, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Johansson K, Ito M, Schophuizen CMS, Mathew Thengumtharayil S, Heuser VD, Zhang J, Shimoji M, Vahter M, Ang WH, Dyson PJ, Shibata A, Shuto S, Ito Y, Abe H, Morgenstern R. Characterization of new potential anticancer drugs designed to overcome glutathione transferase mediated resistance. Mol Pharm 2011; 8:1698-708. [PMID: 21851097 DOI: 10.1021/mp2000692] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Resistance against anticancer drugs remains a serious obstacle in cancer treatment. Here we used novel strategies to target microsomal glutathione transferase 1 (MGST1) and glutathione transferase pi (GSTP) that are often overexpressed in tumors and confer resistance against a number of cytostatic drugs, including cisplatin and doxorubicin (DOX). By synthetically combining cisplatin with a GST inhibitor, ethacrynic acid, to form ethacraplatin, it was previously shown that cytosolic GST inhibition was improved and that cells became more sensitive to cisplatin. Here we show that ethacraplatin is easily taken up by the cells and can reverse cisplatin resistance in MGST1 overexpressing MCF7 cells. A second and novel strategy to overcome GST mediated resistance involves using GST releasable cytostatic drugs. Here we synthesized two derivatives of DOX, 2,4-dinitrobenzenesulfonyl doxorubicin (DNS-DOX) and 4-mononitrobenzenesulfonyl doxorubicin (MNS-DOX) and showed that they are substrates for MGST1 and GSTP (releasing DOX). MGST1 overexpressing cells are resistant to DOX. The resistance is partially reversed by DNS-DOX. Interestingly, the less reactive MNS-DOX was more cytotoxic to cells overexpressing MGST1 than control cells. It would appear that, by controlling the reactivity of the prodrug, and thereby the DOX release rate, selective toxicity to MGST1 overexpressing cells can be achieved. In the case of V79 cells, DOX resistance proportional to GSTP expression levels was noted. In this case, not only was drug resistance eliminated by DNS-DOX but a striking GSTP-dependent increase in toxicity was observed in the clonogenic assay. In summary, MGST1 and GSTP resistance to cytostatic drugs can be overcome and cytotoxicity can be enhanced in GST overexpressing cells.
Collapse
Affiliation(s)
- Katarina Johansson
- Institute of Environmental Medicine, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Nath N, Chattopadhyay M, Pospishil L, Cieciura LZ, Goswami S, Kodela R, Saavedra JE, Keefer LK, Kashfi K. JS-K, a nitric oxide-releasing prodrug, modulates ß-catenin/TCF signaling in leukemic Jurkat cells: evidence of an S-nitrosylated mechanism. Biochem Pharmacol 2010; 80:1641-9. [PMID: 20797387 PMCID: PMC6959133 DOI: 10.1016/j.bcp.2010.08.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 08/13/2010] [Accepted: 08/16/2010] [Indexed: 12/17/2022]
Abstract
β-Catenin is a central player of the Wnt signaling pathway that regulates cell-cell adhesion and may promote leukemia cell proliferation. We examined whether JS-K, an NO-donating prodrug, modulates the Wnt/β-catenin/TCF-4 signaling pathway in Jurkat T-Acute Lymphoblastic Leukemia cells. JS-K inhibited Jurkat T cell growth in a concentration and time-dependent manner. The IC(50)s for cell growth inhibition were 14±0.7 and 9±1.2μM at 24 and 48h, respectively. Treatment of the cells with JS-K for 24h, caused a dose-dependent increase in apoptosis from 16±3.3% at 10μM to 74.8±2% at 100μM and a decrease in proliferation. This growth inhibition was also due, in part, to alterations in the different phases of the cell cycle. JS-K exhibited a dose-dependent cytotoxicity as measured by LDH release at 24h. However, between 2 and 8h, LDH release was less than 20% for any indicated JS-K concentration. The β-catenin/TCF-4 transcriptional inhibitory activity was reduced by 32±8, 63±5, and 93±2% at 2, 10, and 25μM JS-K, respectively, based on luciferase reporter assays. JS-K reduced nuclear β-catenin and cyclin D1 protein levels, but cytosolic β-catenin expression did not change. Based on a time-course assay of S-nitrosylation of proteins by a biotin switch assay, S-nitrsolyation of nuclear β-catenin was determined to precede its degradation. A comparison of the S-nitrosylated nuclear β-catenin to the total nuclear β-catenin showed that β-catenin protein levels were degraded at 24h, while S-nitrosylation of β-catenin occurred earlier at 0-6h. The NO scavenger PTIO abrogated the JS-K mediated degradation of β-catenin demonstrating the need for NO.
Collapse
Affiliation(s)
- Niharika Nath
- Department of Physiology and Pharmacology, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031, United States
- Department of Life Sciences, New York Institute of Technology, New York, NY, United States
| | - Mitali Chattopadhyay
- Department of Physiology and Pharmacology, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031, United States
| | - Liliya Pospishil
- Department of Physiology and Pharmacology, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031, United States
| | - Lucyna Z. Cieciura
- Department of Physiology and Pharmacology, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031, United States
| | - Satindra Goswami
- Department of Physiology and Pharmacology, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031, United States
| | - Ravinder Kodela
- Department of Physiology and Pharmacology, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031, United States
| | - Joseph E. Saavedra
- Basic Research Program, SAIC-Frederick Inc., National Cancer Institute at Frederick, Frederick, MD, United States
| | - Larry K. Keefer
- Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Khosrow Kashfi
- Department of Physiology and Pharmacology, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031, United States
| |
Collapse
|
22
|
Razavi R, Gehrke I, Gandhirajan RK, Poll-Wolbeck SJ, Hallek M, Kreuzer KA. Nitric Oxide–Donating Acetylsalicylic Acid Induces Apoptosis in Chronic Lymphocytic Leukemia Cells and Shows Strong Antitumor Efficacy In vivo. Clin Cancer Res 2010; 17:286-93. [DOI: 10.1158/1078-0432.ccr-10-1030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
23
|
Current World Literature. Curr Opin Support Palliat Care 2010; 4:111-20. [DOI: 10.1097/spc.0b013e32833a1dfc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Nath N, Chattopadhyay M, Kodela R, Tian S, Vlismas P, Boring D, Crowell JA, Kashfi K. Modulation of stress genes expression profile by nitric oxide-releasing aspirin in Jurkat T leukemia cells. Biochem Pharmacol 2010; 79:1759-71. [PMID: 20188076 DOI: 10.1016/j.bcp.2010.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2010] [Accepted: 02/16/2010] [Indexed: 12/31/2022]
Abstract
NO-donating aspirin (NO-ASA, para isomer) has been reported to exhibit strong growth inhibitory effect in Jurkat T-acute lymphoblastic leukemia (T-ALL) cells mediated in part by beta-catenin degradation and caspase activation, but the mechanism(s) still remains unclear. In this study, DNA oligoarrays with 263 genes were used to examine the gene expression profiles relating to stress and drug metabolism, and characterize the stress responses at IC(50) and subIC(50) concentrations of p-NO-ASA (20 and 10microM, respectively) in Jurkat T cells. A total of 22 genes related to heat shock response, apoptosis signaling, detoxifiers and Phase II enzymes, and regulators of cell growth were altered in expression by array analysis based on the expression fold change criteria of > or =1.5-fold or < or =0.65-fold. Real time quantitative RT-PCR confirmed that 20microM p-NO-ASA strongly upregulated the mRNA levels of two heat shock genes HSPA1A (41.5+/-7.01-fold) and HSPA6 (100.4+/-8.11-fold), and FOS (16.2+/-3.2-fold), moderately upregulated HSPH1 (1.71+/-0.43-fold), FMO4 (4.5+/-1.67-fold), CASP9 (1.77+/-0.03-fold), DDIT3 (5.6+/-0.51-fold), and downregulated NF-kappaB1 (0.54+/-0.01-fold) and CCND1 (0.69+/-0.06-fold). Protein levels of Hsp70, the product of HSPA1A, and fos were increased in p-NO-ASA-treated Jurkat T and HT-29 colon cancer cells in a dose-dependent manner. Silencing of Hsp70 enhanced the growth inhibitory effect of p-NO-ASA at low concentrations. The altered gene expression patterns by NO-ASA in Jurkat T cells suggest mechanisms for carcinogen metabolism, anti-proliferative activity and possible chemoprotective activity in T-ALL.
Collapse
Affiliation(s)
- Niharika Nath
- Department of Life Sciences, New York Institute of Technology, New York, NY 10023, USA.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kashfi K. Anti-inflammatory agents as cancer therapeutics. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2009; 57:31-89. [PMID: 20230759 DOI: 10.1016/s1054-3589(08)57002-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer prevention sometimes referred to as tertiary prevention or chemoprevention makes use of specific xenobiotics or drugs to prevent, delay, or retard the development of cancer. Over the last two decades or so cancer prevention has made significant strides. For example, prevention of lung cancer through smoking cessation; cervical cancer prevention through regular Pap smear tests; colon cancer prevention through screening colonoscopy; and prostate cancer reductions by prostate-specific antigen measurements in conjunction with regular prostate examinations. The seminal epidemiological observation that nonsteroidal anti-inflammatory drugs (NSAIDs) prevent colon and other cancers has provided the impetus to develop novel chemoprevention approaches against cancer. To that end, a number of "designer drugs" have been synthesized that are in different stages of development, evaluation, and deployment. Some include the cyclooxygenase-2-specific inhibitors (coxibs), nitric oxide-releasing NSAIDs (NO-NSAIDs and NONO-NSAIDs), hydrogen sulfide-releasing NSAIDs, modulators of the lipoxygenase pathway, prostanoid receptor blockers, and chemokine receptor antagonists. In addition to these novel agents, there are also a host of naturally occurring compounds/micronutrients that have chemopreventive properties. This chapter reviews these classes of compounds, their utility and mechanism(s) of action against the background of mediators that link inflammation and cancer.
Collapse
Affiliation(s)
- Khosrow Kashfi
- Department of Physiology and Pharmacology, Sophie Davis School of Biomedical Education, The City College of The City University of New York, New York 10031, USA
| |
Collapse
|