1
|
Abstract
INTRODUCTION High-risk HPV infections are related to several epithelial cancers. Despite the availability of prophylactic vaccines, HPV infections are still responsible for about 5% of all human malignancies worldwide. While therapeutic vaccines are ongoing clinical trials, genotoxic agents and surgical interventions represent current clinical treatments, with no specific anti-HPV drugs yet available in the clinics. AREAS COVERED We offer a comprehensive report of small molecules in preclinical studies proposed as potential anticancer agents against HPV-driven tumors. Given the importance of HPV oncoproteins for cancer maintenance, particularly E6 and E7, we present a classification of both non-targeted and targeted agents, with a further subdivision of the latter into two categories according to their either direct or indirect activity against viral protein functions. EXPERT OPINION Prophylactic vaccines can prevent the insurgence of HPV-related cancers, but have no effect against pre-existing infections. Moreover, their high cost, genotype-restricted effect and the growing worldwide distrust for vaccines make the availability of a specific drug an unmet medical need. Different viral early proteins emerge as ideal candidates for drug development. We highlight the most promising strategies and address future challenges in this field to herald the prospect of a specific therapeutic regimen against HPV-related cancers.
Collapse
Affiliation(s)
- Lorenzo Messa
- Department of Molecular Medicine, University of Padua, Padua, 35121, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, 35121, Italy.,Clinical Microbiology and Virology Unit, Padua University Hospital, Padua, Italy
| |
Collapse
|
2
|
Programmed cell death, redox imbalance, and cancer therapeutics. Apoptosis 2021; 26:385-414. [PMID: 34236569 DOI: 10.1007/s10495-021-01682-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/06/2023]
Abstract
Cancer cells are disordered by nature and thus featured by higher internal redox level than healthy cells. Redox imbalance could trigger programmed cell death if exceeded a certain threshold, rendering therapeutic strategies relying on redox control a possible cancer management solution. Yet, various programmed cell death events have been consecutively discovered, complicating our understandings on their associations with redox imbalance and clinical implications especially therapeutic design. Thus, it is imperative to understand differences and similarities among programmed cell death events regarding their associations with redox imbalance for improved control over these events in malignant cells as well as appropriate design on therapeutic approaches relying on redox control. This review addresses these issues and concludes by bringing affront cold atmospheric plasma as an emerging redox controller with translational potential in clinics.
Collapse
|
3
|
MartĂnez-NoĂ«l G, Vieira VC, Szajner P, Lilienthal EM, Kramer RE, Boyland KA, Smith JA, Howley PM. Live cell, image-based high-throughput screen to quantitate p53 stabilization and viability in human papillomavirus positive cancer cells. Virology 2021; 560:96-109. [PMID: 34051479 DOI: 10.1016/j.virol.2021.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/16/2021] [Accepted: 05/16/2021] [Indexed: 11/16/2022]
Abstract
Approximately 5% of cancers are caused by high-risk human papillomaviruses. Although very effective preventive vaccines will reduce this cancer burden significantly over the next several decades, they have no therapeutic effect for those already infected and remaining at risk for malignant progression of hrHPV lesions. HPV-associated cancers are dependent upon the expression of the viral E6 and E7 oncogenes. The oncogenic function of hrHPV E6 relies partially on its ability to induce p53 degradation. Since p53 is generally wildtype in hrHPV-associated cancers, p53 stabilization arrests proliferation, induces apoptosis and/or results in senescence. Here we describe a live cell, image-based high-throughput screen to identify compounds that stabilize p53 and/or affect viability in HPV-positive cancer HeLa cells. We validate the robustness and potential of this screening assay by assessing the activities of approximately 6,500 known bioactive compounds, illustrating its capability to function as a platform to identify novel therapeutics for hrHPV.
Collapse
Affiliation(s)
- Gustavo MartĂnez-NoĂ«l
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Valdimara CorrĂŞa Vieira
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Patricia Szajner
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Erin M Lilienthal
- ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Rebecca E Kramer
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Kathleen A Boyland
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Jennifer A Smith
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA; ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Peter M Howley
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Lourenço de Freitas N, Deberaldini MG, Gomes D, Pavan AR, Sousa Â, Dos Santos JL, Soares CP. Histone Deacetylase Inhibitors as Therapeutic Interventions on Cervical Cancer Induced by Human Papillomavirus. Front Cell Dev Biol 2021; 8:592868. [PMID: 33634093 PMCID: PMC7901962 DOI: 10.3389/fcell.2020.592868] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
The role of epigenetic modifications on the carcinogenesis process has received a lot of attention in the last years. Among those, histone acetylation is a process regulated by histone deacetylases (HDAC) and histone acetyltransferases (HAT), and it plays an important role in epigenetic regulation, allowing the control of the gene expression. HDAC inhibitors (HDACi) induce cancer cell cycle arrest, differentiation, and cell death and reduce angiogenesis and other cellular events. Human papillomaviruses (HPVs) are small, non-enveloped double-stranded DNA viruses. They are major human carcinogens, being intricately linked to the development of cancer in 4.5% of the patients diagnosed with cancer worldwide. Long-term infection of high-risk (HR) HPV types, mainly HPV16 and HPV18, is one of the major risk factors responsible for promoting cervical cancer development. In vitro and in vivo assays have demonstrated that HDACi could be a promising therapy to HPV-related cervical cancer. Regardless of some controversial studies, the therapy with HDACi could target several cellular targets which HR-HPV oncoproteins could be able to deregulate. This review article describes the role of HDACi as a possible intervention in cervical cancer treatment induced by HPV, highlighting the main advances reached in the last years and providing insights for further investigations regarding those agents against cervical cancer.
Collapse
Affiliation(s)
- Natália Lourenço de Freitas
- Department of Clinical Analysis, School of Pharmaceutical Sciences, SĂŁo Paulo State University (UNESP), Araraquara, Brazil
| | - Maria Gabriela Deberaldini
- Drugs and Medicines Department, School of Pharmaceutical Science, SĂŁo Paulo State University (UNESP), Araraquara, Brazil
- Institute of Chemistry, SĂŁo Paulo State University (UNESP), Araraquara, Brazil
| | - Diana Gomes
- CICS-UBI – Health Science Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Aline Renata Pavan
- Drugs and Medicines Department, School of Pharmaceutical Science, SĂŁo Paulo State University (UNESP), Araraquara, Brazil
- Institute of Chemistry, SĂŁo Paulo State University (UNESP), Araraquara, Brazil
| | - Ă‚ngela Sousa
- CICS-UBI – Health Science Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Jean Leandro Dos Santos
- Drugs and Medicines Department, School of Pharmaceutical Science, SĂŁo Paulo State University (UNESP), Araraquara, Brazil
| | - Christiane P. Soares
- Department of Clinical Analysis, School of Pharmaceutical Sciences, SĂŁo Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
5
|
Zhao X, Sun W, Ren Y, Lu Z. Therapeutic potential of p53 reactivation in cervical cancer. Crit Rev Oncol Hematol 2020; 157:103182. [PMID: 33276182 DOI: 10.1016/j.critrevonc.2020.103182] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 10/23/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
Cervical cancer (CC) is one of most common malignancies affecting women worldwide. To date, surgical resection is the only effective radical remedy for CC at its early stages, while the prognosis of metastatic or recurrent CC is very poor. Dysfunction of the tumor suppressor p53 due to aberrant expression, post-translational modification, mutations, SNPs, and LOH as well as sequestration by viral antigens and MDM2/HDM2-mediated degradation is closely associated with the therapeutic insensitivity and relapse of many malignancies, including CC. Accumulating studies have demonstrated that restoration of p53 activity can induce cell cycle arrest and apoptosis, eliminate radio- and chemotherapy resistance, and inhibit tumor growth in CC cells. Therefore, activation of wild-type p53 as well as restoration of p53 function seems appealing as a therapeutic strategy. In this review, we focus on the potential roles of p53 reactivation in CC treatment and their underlying molecular mechanisms towards the development of novel therapies.
Collapse
Affiliation(s)
- Xiangxuan Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China.
| | - Wei Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China
| | - Ying Ren
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China
| | - Zaiming Lu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China
| |
Collapse
|
6
|
Kumar A, Rathi E, Hariharapura RC, Kini SG. Is viral E6 oncoprotein a viable target? A critical analysis in the context of cervical cancer. Med Res Rev 2020; 40:2019-2048. [PMID: 32483862 DOI: 10.1002/med.21697] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/15/2022]
Abstract
An understanding of the pathology of cervical cancer (CC) mediated by E6/E7 oncoproteins of high-risk human papillomavirus (HPV) was developed by late 80's. But if we look at the present scenario, not a single drug could be developed to inhibit these oncoproteins and in turn, be used specifically for the treatment of CC. The readers are advised not to presume the "viability of E6 protein" as mentioned in the title relates to just druggability of E6. The viability aspect will cover almost everything a researcher should know to develop E6 inhibitors until the preclinical stage. Herein, we have analysed the achievements and shortcomings of the scientific community in the last four decades in targeting HPV E6 against CC. Role of all HPV proteins has been briefly described for better perspective with a little detailed discussion of the role of E6. We have reviewed the articles from 1985 onward, reporting in vitro inhibition of E6. Recently, many computational studies have reported potent E6 inhibitors and these have also been reviewed. Subsequently, a critical analysis has been reported to cover the in vitro assay protocols and in vivo models to develop E6 inhibitors. A paragraph has been devoted to the role of public policy to fight CC employing vaccines and whether the vaccine against HPV has quenched the zeal to develop drugs against it. The review concludes with the challenges and the way forward.
Collapse
Affiliation(s)
- Avinash Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ekta Rathi
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raghu Chandrashekar Hariharapura
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Suvarna G Kini
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
7
|
Souza FDO, Sorbo JM, Regasini LO, Bolzani VDS, Rosa JC, Czernys ÉDS, Valente V, Moreira TF, Navegante G, Fernandes BC, Soares CP. Nitensidine B affects proteins of the glycolytic pathway and induces apoptosis in cervical carcinoma cells immortalized by HPV16. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 48:179-186. [PMID: 30195876 DOI: 10.1016/j.phymed.2018.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 05/21/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Cervical cancer, the fourth most common type of cancer among women worldwide, accounts for approximately 12% of all types of malignancies that affect women. Natural products have contributed significantly to the development of modern therapies; approximately 70% of the drugs available for chemotherapy are naturally based products. PURPOSE The purpose of this study was to examine the biological activities of nitensidine B (NTB), a guanidinic alkaloid isolated from the leaves of Pterogyne nitens Tul. (Fabaceae) in a cervical cancer cell line. METHODS In vitro experiments were performed using cervical carcinoma cells immortalized by human papillomavirus type 16 (HPV16, SiHa cells), since epidemiological and molecular studies have demonstrated robust associations between the etiologies of cervical cancer and HPV infection. Cytotoxicity as well as the effect of NTB treatment on intracellular signals of apoptosis, fragmentation of internucleosomal DNA via terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and levels of apoptosis effectors (Caspase 3/7) were evaluated. In addition, differential proteomic analysis (iTRAQ) and protein validation using western blot were performed. RESULTS The cytotoxicity of NTB treatment in the SiHa cell line was concentration-dependent, with the minimum inhibitory concentration of 50% of the cells of 40.98 µM. In the TUNEL assay, SiHa cell apoptosis with 3/7 caspase activation was reported at 12 h following treatment. Differential proteomic analysis by iTRAQ demonstrated that proteins of the glycolytic pathway, aldolase A, alpha-enolase, pyruvate kinase, and glyceraldehyde 3-phosphate dehydrogenase were underexpressed. CONCLUSION These results indicated that NTB could play a role in decreasing glycolysis . Since tumor cells prefer the glycolytic pathway to generate energy, these findings suggest that NTB may be a reliable model for the study of human cervical cancer cell lines immortalized by HPV16, however more experiments can be performed.
Collapse
Affiliation(s)
- Felipe de Oliveira Souza
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Highway Araraquara JaĂş, Km 01, Campos Ville, Araraquara, Sao Paulo, Brazil
| | - Juliana Maria Sorbo
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Highway Araraquara JaĂş, Km 01, Campos Ville, Araraquara, Sao Paulo, Brazil
| | - LuĂs Octávio Regasini
- Department of Chemistry and Environmental Sciences of the Institute of Biosciences, Letters and Exact Sciences of the Sao Paulo State University, Cristovao Colombo street, 2265, Sao Jose do Rio Preto, Sao Paulo, Brazil
| | - Vanderlan da Silva Bolzani
- Department of Organic Chemistry, Institute of Chemistry, SĂŁo Paulo State University, 355, 14800-900 Araraquara, Brazil
| | - José César Rosa
- Center of Protein Chemistry of Department of Cellular Molecular Biology and Pathogen Bioagents of the Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Avenue Bandeirantes, 3900, Ribeirao Preto, Sao Paulo, Brazil
| | - Érica da Silva Czernys
- Center of Protein Chemistry of Department of Cellular Molecular Biology and Pathogen Bioagents of the Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Avenue Bandeirantes, 3900, Ribeirao Preto, Sao Paulo, Brazil
| | - Valéria Valente
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Highway Araraquara JaĂş, Km 01, Campos Ville, Araraquara, Sao Paulo, Brazil
| | - ThaĂs Fernanda Moreira
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Highway Araraquara JaĂş, Km 01, Campos Ville, Araraquara, Sao Paulo, Brazil
| | - Geovana Navegante
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Highway Araraquara JaĂş, Km 01, Campos Ville, Araraquara, Sao Paulo, Brazil
| | - Barbara Colatto Fernandes
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Highway Araraquara JaĂş, Km 01, Campos Ville, Araraquara, Sao Paulo, Brazil
| | - Christiane Pienna Soares
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Highway Araraquara JaĂş, Km 01, Campos Ville, Araraquara, Sao Paulo, Brazil.
| |
Collapse
|
8
|
A quantitative LumiFluo assay to test inhibitory compounds blocking p53 degradation induced by human papillomavirus oncoprotein E6 in living cells. Sci Rep 2018; 8:6020. [PMID: 29662081 PMCID: PMC5902497 DOI: 10.1038/s41598-018-24470-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/29/2018] [Indexed: 12/25/2022] Open
Abstract
High-risk human papillomaviruses (HR-HPVs) are the causative agents for the onset of several epithelial cancers in humans. The deregulated expression of the viral oncoproteins E6 and E7 is the driving force sustaining the progression of malignant transformation in pre-neoplastic lesions. Targeting the viral E6 oncoprotein through inhibitory compounds can counteract the survival of cancer cells due to the reactivation of p53-mediated pathways and represents an intriguing strategy to treat HPV-associated neoplasias. Here, we describe the development of a quantitative and easy-to-perform assay to monitor the E6-mediated degradation of p53 in living cells to be used for small-molecule testing. This assay allows to unbiasedly determine whether a compound can protect p53 from the E6-mediated degradation in cells, through a simple 3-step protocol. We validated the assay by testing two small molecules, SAHA and RITA, reported to impair the E6-mediated p53 degradation. Interestingly, we observed that only SAHA efficiently rescued p53, while RITA could not provide the same degree of protection. The possibility to specifically and quantitatively monitor the ability of a selected compound to rescue p53 in a cellular context through our LumiFluo assay could represent an important step towards the successful development of anti-HPV drugs.
Collapse
|
9
|
DasGupta T, Nweze EI, Yue H, Wang L, Jin J, Ghosh SK, Kawsar HI, Zender C, Androphy EJ, Weinberg A, McCormick TS, Jin G. Human papillomavirus oncogenic E6 protein regulates human β-defensin 3 (hBD3) expression via the tumor suppressor protein p53. Oncotarget 2017; 7:27430-44. [PMID: 27034006 PMCID: PMC5053661 DOI: 10.18632/oncotarget.8443] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/17/2016] [Indexed: 01/24/2023] Open
Abstract
Human β-defensin-3 (hBD3) is an epithelial cell-derived innate immune regulatory molecule overexpressed in oral dysplastic lesions and fosters a tumor-promoting microenvironment. Expression of hBD3 is induced by the epidermal growth factor receptor signaling pathway. Here we describe a novel pathway through which the high-risk human papillomavirus type-16 (HPV-16) oncoprotein E6 induces hBD3 expression in mucosal keratinocytes. Ablation of E6 by siRNA induces the tumor suppressor p53 and diminishes hBD3 in HPV-16 positive CaSki cervical cancer cells and UM-SCC-104 head and neck cancer cells. Malignant cells in HPV-16-associated oropharyngeal cancer overexpress hBD3. HPV-16 E6 induces hBD3 mRNA expression, peptide production and gene promoter activity in mucosal keratinocytes. Reduction of cellular levels of p53 stimulates hBD3 expression, while activation of p53 by doxorubicin inhibits its expression in primary oral keratinocytes and CaSki cells, suggesting that p53 represses hBD3 expression. A p53 binding site in the hBD3 gene promoter has been identified by using electrophoretic mobility shift assays and chromatin immunoprecipitation (ChIP). In addition, the p63 protein isoform ΔNp63α, but not TAp63, stimulated transactivation of the hBD3 gene and was co-expressed with hBD3 in head and neck cancer specimens. Therefore, high-risk HPV E6 oncoproteins may stimulate hBD3 expression in tumor cells to facilitate tumorigenesis of HPV-associated head and neck cancer.
Collapse
Affiliation(s)
- Twishasri DasGupta
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Emeka I Nweze
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA.,Present Address: University of Nigeria, Nsukka, Nigera
| | - Hong Yue
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Liming Wang
- Center for Molecular Cancer Diagnosis Inc., Twinsburg, OH, USA
| | - Jessica Jin
- Human Developmental and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Santosh K Ghosh
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Hameem I Kawsar
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA.,Present Address: St. Luke's Hospital, Chesterfield, MO, USA
| | - Chad Zender
- Department of Otolaryngology-Head & Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Elliot J Androphy
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Aaron Weinberg
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Thomas S McCormick
- Department of Dermatology, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ge Jin
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| |
Collapse
|
10
|
Krushkal J, Zhao Y, Hose C, Monks A, Doroshow JH, Simon R. Concerted changes in transcriptional regulation of genes involved in DNA methylation, demethylation, and folate-mediated one-carbon metabolism pathways in the NCI-60 cancer cell line panel in response to cancer drug treatment. Clin Epigenetics 2016; 8:73. [PMID: 27347216 PMCID: PMC4919895 DOI: 10.1186/s13148-016-0240-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Aberrant patterns of DNA methylation are abundant in cancer, and epigenetic pathways are increasingly being targeted in cancer drug treatment. Genetic components of the folate-mediated one-carbon metabolism pathway can affect DNA methylation and other vital cell functions, including DNA synthesis, amino acid biosynthesis, and cell growth. RESULTS We used a bioinformatics tool, the Transcriptional Pharmacology Workbench, to analyze temporal changes in gene expression among epigenetic regulators of DNA methylation and demethylation, and one-carbon metabolism genes in response to cancer drug treatment. We analyzed gene expression information from the NCI-60 cancer cell line panel after treatment with five antitumor agents, 5-azacytidine, doxorubicin, vorinostat, paclitaxel, and cisplatin. Each antitumor agent elicited concerted changes in gene expression of multiple pathway components across the cell lines. Expression changes of FOLR2, SMUG1, GART, GADD45A, MBD1, MTR, MTHFD1, and CTH were significantly correlated with chemosensitivity to some of the agents. Among many genes with concerted expression response to individual antitumor agents were genes encoding DNA methyltransferases DNMT1, DNMT3A, and DNMT3B, epigenetic and DNA repair factors MGMT, GADD45A, and MBD1, and one-carbon metabolism pathway members MTHFD1, TYMS, DHFR, MTR, MAT2A, SLC19A1, ATIC, and GART. CONCLUSIONS These transcriptional changes are likely to influence vital cellular functions of DNA methylation and demethylation, cellular growth, DNA biosynthesis, and DNA repair, and some of them may contribute to cytotoxic and apoptotic action of the drugs. This concerted molecular response was observed in a time-dependent manner, which may provide future guidelines for temporal selection of genetic drug targets for combination drug therapy treatment regimens.
Collapse
Affiliation(s)
- Julia Krushkal
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| | - Yingdong Zhao
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| | - Curtis Hose
- />Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Anne Monks
- />Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - James H. Doroshow
- />Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892 USA
| | - Richard Simon
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| |
Collapse
|
11
|
Choi YH. Induction of apoptosis by an ethanol extract of Poria cocos Wolf. in human leukemia U937Â cells. Oncol Rep 2015; 34:2533-40. [PMID: 26353048 DOI: 10.3892/or.2015.4256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/31/2015] [Indexed: 11/06/2022] Open
Abstract
Poria cocos Wolf., which belongs to the Polyporaceae family, has been widely used as an Oriental traditional herbal medicine for centuries. Its sclerotium has been reported to possess a wide spectrum of pharmacological activities, including free-radical scavenging, anti-viral, anti-microbial, anti-inflammatory and anticancer activities. However, the cellular and molecular mechanisms of apoptosis induction by P. cocos in human cancer cells are poorly understood. In the present study, we investigated the pro-apoptotic potential of an ethanol extract of P. cocos sclerotium (EEPC) in human leukemia U937 cells in vitro. We found that EEPC induced anti-proliferative effects in U937 cells in a concentration- and time-dependent manner, which was due to apoptotic induction, as evident from morphological changes and flow cytometric assays. EEPC-induced apoptosis of U937 cells was associated with an increase in the Bax:Bcl-2 ratio, the release of cytochrome c to the cytosol, and a decrease in the expression of an inhibitor of the apoptosis family of proteins. The events were accompanied by activation of caspase-8, -9 and -3, and cleaved poly(ADP-ribose) polymerase, suggesting the involvement of both the intrinsic and extrinsic apoptotic cascades. In addition, the overexpression of Bcl-2 caused a significant attenuation of EEPC-induced caspase activation, degradation of PARP, and the collapse of mitochondrial membrane potential, and thereby reversed EEPC-induced cell apoptosis and growth inhibition. Collectively, these data provide insights into the molecular mechanisms underlying EEPC-induced apoptosis in U937 cells, suggesting that EEPC may be a new therapeutic option for the treatment of leukemia.
Collapse
Affiliation(s)
- Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| |
Collapse
|
12
|
Park HY, Choi IW, Kim GY, Kim BW, Kim WJ, Choi YH. Fucoidan induces G1 arrest of the cell cycle in EJ human bladder cancer cells through down-regulation of pRB phosphorylation. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2015. [DOI: 10.1016/j.bjp.2015.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
13
|
Lin K, Zhang Q, Liu Z, Yang S, Lin Y, Wen C, Zheng Y. Effects of suberoylanilide hydroxamic acid on rat cytochrome P450 enzyme activities. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5584-5590. [PMID: 26191268 PMCID: PMC4503139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 04/21/2015] [Indexed: 06/04/2023]
Abstract
Vorinostat (suberoylanilide hydroxamic acid, SAHA) is the first approved histone deacetylase (HDAC) inhibitor for the treatment of cutaneous T-cell lymphoma after progressive disease following two systemic therapies. The rats were randomly divided into SAHA groups (low, medium and high dosage) and control group. The SAHA group rats were given 12.3, 24.5, and 49 mg/kg SAHA, respectively, by continuous intragastric administration for 7 days. The influence of SAHA on the activities of CYP450 isoforms CYP2B6, CYP1A2, CYP2C19, CYP2D6 and CYP2C9 were evaluated by cocktail method, they were responsed by the changes of pharmacokinetic parameters of bupropion, phenacetin, tolbutamide, metroprolol and omeprazole. The five probe drugs were given to rats through intragastric administration, and the plasma concentration were determined by UPLC-MS/MS. The result of SAHA group compared to control group, there were statistical pharmacokinetics difference for bupropion, phenacetin, tolbutamide and metroprolol. Continuous intragastric administration for 7 days may induce the activities of CYP2C19 of rats, inhibit CYP1A2 and slightly inhibit CYP2B6 and CYP2D6 of rats. This may give advising for reasonable drug use after co-used with SAHA. The results indicated that drug co-administrated with SAHA may need dose adjustment. Furthermore, continuous intragastric administration of SAHA for 7 days, liver cell damaged, causing liver cell edema, in liver metabolism process.
Collapse
Affiliation(s)
- Kezhi Lin
- Medical Experimental Teaching Center, Wenzhou Medical UniversityWenzhou 325035, China
| | - Qingwei Zhang
- Shanghai Institute of Pharmaceutical IndustryShanghai 200437, China
| | - Zezheng Liu
- Shanghai Institute of Pharmaceutical IndustryShanghai 200437, China
| | - Suping Yang
- Shanghai Institute of Pharmaceutical IndustryShanghai 200437, China
| | - Yingying Lin
- Shanghai Institute of Pharmaceutical IndustryShanghai 200437, China
| | - Congcong Wen
- Laboratory Animal Centre, Wenzhou Medical UniversityWenzhou 325035, China
| | - Yuancai Zheng
- The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325035, China
| |
Collapse
|
14
|
Orzechowska EJ, Girstun A, Staron K, Trzcinska-Danielewicz J. Synergy of BID with doxorubicin in the killing of cancer cells. Oncol Rep 2015; 33:2143-50. [PMID: 25760094 PMCID: PMC4391587 DOI: 10.3892/or.2015.3841] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/10/2015] [Indexed: 12/19/2022] Open
Abstract
Overexpression of the BH3-interacting domain death agonist (BID) protein sensitizes certain cancer cell lines to apoptosis induced by anticancer agents, particularly by those acting through death receptors (e.g. TRAIL). Previously, we showed that recombinant BID fused with TAT cell penetrating peptide (TAT-BID) allowed for controlled delivery of BID to different cancer cell lines and moderately sensitized some of them to TRAIL or slightly to camptothecin. In the present study, we showed that TAT-BID delivered to HeLa cells strongly sensitized them to doxorubicin, as identified by cell viability and apoptosis assays. Another cell line sensitized to doxorubicin was PC3, whereas A549 and LNCaP cells were sensitized moderately or not at all, respectively. Sensitization was more pronounced at 1 ÎĽM doxorubicin administered for 48 h than for lower doses and shorter treatments. TAT-BID and doxorubicin may thus be considered as a potential therapeutic combination for cervical carcinoma and advanced prostate cancer treatment.
Collapse
Affiliation(s)
- Emilia Joanna Orzechowska
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Agnieszka Girstun
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Krzysztof Staron
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Joanna Trzcinska-Danielewicz
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| |
Collapse
|
15
|
Kang JS, Han MH, Kim GY, Kim CM, Chung HY, Hwang HJ, Kim BW, Choi YH. Schisandrae semen essential oil attenuates oxidative stress-induced cell damage in C2C12 murine skeletal muscle cells through Nrf2‑mediated upregulation of HO‑1. Int J Mol Med 2014; 35:453-9. [PMID: 25482391 DOI: 10.3892/ijmm.2014.2028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 11/26/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to examine the cytoprotective effects of Schisandrae semen essential oil (SSeo), purified from Schisandrae fructus, against oxidative stress-induced cell damage in C2C12 myoblasts. SSeo attenuated hydrogen peroxide (H2O2)-induced growth inhibition and exhibited scavenging activity against the intracellular reactive oxygen species (ROS) that were induced by H2O2. SSeo also inhibited comet tail formation, chromatin condensation and phosphor-histone γH2A.X expression, suggesting that it prevents H2O2-induced cellular DNA damage and apoptotic cell death. Furthermore, SSeo significantly enhanced the expression of heme oxygenase-1 (HO‑1) associated with the induction of nuclear factor erythroid-2-related factor 2 (Nrf2) in a time- and concentration‑dependent manner. In addition, the protective effect of SSeo on H2O2‑induced C2C12 cell damage was significantly inhibited by zinc protoporphyrin IX, an HO‑1 competitive inhibitor, in C2C12 cells. These findings suggest that SSeo augments the cellular antioxidant defense capacity through intrinsic free radical scavenging activity and activation of the Nrf2/HO‑1 pathway, thereby protecting the C2C12 cells from H2O2‑induced oxidative cytotoxicity. As a result, SSeo may have therapeutic potential in the development of functional foods and as the raw material for medicines to protect against oxidative stress.
Collapse
Affiliation(s)
- Ji Sook Kang
- Blue‑Bio Industry RIC and Anti‑Aging Research Center, Dongeui University, Busan 614-714, Republic of Korea
| | - Min Ho Han
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
| | - Cheol Min Kim
- Department of Biochemistry, Busan National University College of Medicine, Yangsan 626-870, Republic of Korea
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Hye Jin Hwang
- Blue‑Bio Industry RIC and Anti‑Aging Research Center, Dongeui University, Busan 614-714, Republic of Korea
| | - Byung Woo Kim
- Blue‑Bio Industry RIC and Anti‑Aging Research Center, Dongeui University, Busan 614-714, Republic of Korea
| | - Yung Hyun Choi
- Blue‑Bio Industry RIC and Anti‑Aging Research Center, Dongeui University, Busan 614-714, Republic of Korea
| |
Collapse
|
16
|
Park SE, Shin WT, Park C, Hong SH, Kim GY, Kim SO, Ryu CH, Hong SH, Choi YH. Induction of apoptosis in MDA-MB-231 human breast carcinoma cells with an ethanol extract of Cyperus rotundus L. by activating caspases. Oncol Rep 2014; 32:2461-70. [PMID: 25241797 DOI: 10.3892/or.2014.3507] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 08/29/2014] [Indexed: 11/05/2022] Open
Abstract
Cyperus rotundus L. belongs to the Cyperaceae family and is a well documented traditional medicinal herb. Its rhizome has been reported to possess wide spectrum pharmacological activities including anti-inflammatory and antioxidant activity. However, the cellular and molecular mechanisms of the anticancer activity have not been elucidated. In the present study, we investigated the pro-apoptotic effects of C. rotundu rhizomes in a human breast carcinoma MDA-MB-231 cell model. Treatment of MDA-MB-231 cells with an ethanol extract of C. rotundu rhizomes (EECR) and a methanol extract of C. rotundu rhizomes (MECR), but not a water extract of C. rotundu rhizomes, resulted in potent antiproliferative activity. The activity of the EECR was higher than that of the MECR and was associated with the induction of apoptosis. The induction of apoptosis by the EECR was associated with upregulation of death receptor 4 (DR4), DR5 and pro-apoptotic Bax, as well as downregulation of anti-apoptotic survivin and Bcl-2. EECR treatment also downregulated Bid expression and activated caspase-8 and -9, the respective initiator caspases of the extrinsic and intrinsic apoptotic pathways. The increase in mitochondrial membrane depolarization was correlated with activation of effector caspase-3 and cleavage of poly(ADP-ribose) polymerase, a vital substrate of activated caspase-3. Blockage of caspase activation by pretreatment with a pan-caspase inhibitor consistently inhibited apoptosis and abrogated growth inhibition in EECR-treated MDA-MB-231 cells. Although reactive oxygen species (ROS) increased following treatment with the EECR, inhibiting ROS with a ROS scavenger did not attenuate EECR-induced apoptosis. Furthermore, inhibitors of phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK) signaling pathways failed to reverse EECR-induced apoptosis and growth inhibition. These results suggest that the pro-apoptotic activity of the EECR may be regulated by a caspase-dependent cascade through activation of both intrinsic and extrinsic signaling pathways that is not associated with ROS generation or the PI3K/Akt and MAPK pathways.
Collapse
Affiliation(s)
- Sang Eun Park
- Department of Internal Medicine, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Won Tak Shin
- Department of Internal Medicine, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dongeui University, Busan 614-714, Republic of Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
| | - Sung Ok Kim
- Team for Scientification of Korean Medical Intervention (BK21 Plus) and Department of Herbal Pharmacology, Daegu Haany University College of Korean Medicine, Daegu 706-828, Republic of Korea
| | - Chung Ho Ryu
- Division of Applied Life Science (BK 21 Program), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 660‑701, Republic of Korea
| | - Sang Hoon Hong
- Department of Internal Medicine, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| |
Collapse
|
17
|
Fucoidan inhibits the proliferation of human urinary bladder cancer T24 cells by blocking cell cycle progression and inducing apoptosis. Molecules 2014; 19:5981-98. [PMID: 24818577 PMCID: PMC6271230 DOI: 10.3390/molecules19055981] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 11/16/2022] Open
Abstract
Although fucoidan has been shown to exert anticancer activity against several types of cancer cell lines, no reports have explored fucoidan-affected cell growth in human urinary bladder cancer cells. In this study, we investigated the anti-proliferative effects of fucoidan in human bladder cancer T24 cells. Our results indicated that fucoidan decreased the viability of T24 cells through the induction of G1 arrest and apoptosis. Fucoidan-induced G1 arrest is associated with the enhanced expression of the Cdk inhibitor p21WAF1/CIP1 and dephosphorylation of the pRB along with enhanced binding of p21 to Cdk4/6 as well as pRB to the transcription factor E2Fs. Further investigations showed the loss of mitochondrial membrane potential and the release of cytochrome c from mitochondria to cytosol, proving mitochondrial dysfunction upon fucoidan treatment with a corresponding increase in the Bax/Bcl-2 expression ratio. Fucoidan-triggered apoptosis was also accompanied by the up-regulation of Fas and truncated Bid as well as the sequential activation of caspase-8. Furthermore, a significant increased activation of caspase-9/-3 was detected in response to fucoidan treatment with the decreased expression of IAPs and degradation of PARP, whereas a pan-caspase inhibitor significantly suppressed apoptosis and rescued the cell viability reduction. In conclusion, these observations suggest that fucoidan attenuates G1-S phase cell cycle progression and serves as an important mediator of crosstalk between caspase-dependent intrinsic and extrinsic apoptotic pathways in T24 cells.
Collapse
|