1
|
Satapathy SS, Bhuyan R, Pradhan AK, Satpathy S, Panda NR, Bhuyan SK. Comparative evaluation of anticancer potential of Spilanthes paniculata leaf and flower essential oil using annexin V and orosphere formation in oral cancer cell line. Med Oncol 2024; 41:259. [PMID: 39369172 DOI: 10.1007/s12032-024-02523-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Spilanthes paniculata, a member of the Asteraceae family, is predominantly used as a traditional remedy in addressing oral ailments, particularly gum infections and sore throat. The flowers are chewed to alleviate toothache immediately. This study evaluated a comparison between the essential oils of leaf and flower derived from Spilanthes paniculata targeting the SCC9 oral cell lines using in vitro and in silico approaches. The anticancer activity was performed through an MTT assay, apoptosis assays using annexin V and orospheres formation assays. Molecular docking was performed between five selected phytocompounds against the p53 protein by using AutoDock 4.2.6 software. The results confirmed that the flower essential oil significantly reduced the cell viability in a dose-dependent manner, with an IC50 value of 113.95 µg/mL. The apoptosis assays showed that the flower essential oil was approximately 2.5 times more effective in inducing early apoptosis at 50 µg/mL compared to the essential oil of the leaf. The orosphere formation assays further confirmed the anticancer potential of the flower essential oil. Spathulenol exhibited strong hydrogen bonding with the p53 protein. The ADMET prediction tools were used to predict the in silico pharmacokinetics and drug-like properties of the phytoconstituents. The results suggested that Spathulenol and Nerolidol have high gastrointestinal absorption (GIA), with estimated solubility (ESOL) values of - 3.17 and - 3.22, respectively, falling within the optimal range. These findings suggest that the flower's essential oil efficiently prevented the proliferation of oral cancer and observed a notable degree of cell death, inducing apoptosis and suggesting its significant antiproliferative activity against cancerous cell line which could be explored further for therapeutic applications.
Collapse
Affiliation(s)
- Soumya Subhashree Satapathy
- Centre for Biotechnology, School of Pharmaceutical Science, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Ruchi Bhuyan
- Department of Medical Research, IMS and SUM Hospital, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
- Institute of Dental Science, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Arun Kumar Pradhan
- Centre for Biotechnology, School of Pharmaceutical Science, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Soumya Satpathy
- Department of Medical Research, IMS and SUM Hospital, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Nihar Ranjan Panda
- Department of Medical Research, IMS and SUM Hospital, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Sanat Kumar Bhuyan
- Institute of Dental Science, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India.
| |
Collapse
|
2
|
Ananta, Benerjee S, Tchounwou PB, Kumar S. Mechanistic update of Trisenox in blood cancer. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023; 5:100166. [PMID: 38074774 PMCID: PMC10701371 DOI: 10.1016/j.crphar.2023.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 10/28/2023] [Accepted: 11/14/2023] [Indexed: 02/12/2024] Open
Abstract
Acute promyelocytic leukemia (APL)/blood cancer is M3 type of acute myeloid leukemia (AML) formed inside bone marrow through chromosomal translocation mutation usually between chromosome 15 & 17. It accounts around 10% cases of AML worldwide. Trisenox (TX/ATO) is used in chemotherapy for treatment of all age group of APL patients with highest efficacy and survival rate for longer period. High concentration of TX inhibits growth of APL cells by diverse mechanism however, it cures only PML-RARα fusion gene/oncogene containing APL patients. TX resistant APL patients (different oncogenic make up) have been reported from worldwide. This review summarizes updated mechanism of TX action via PML nuclear bodies formation, proteasomal degradation, autophagy, p53 activation, telomerase activity, heteromerization of pRb & E2F, and regulation of signaling mechanism in APL cells. We have also provided important information of combination therapy of TX with other molecules mechanism of action in acute leukemia cells. It provides updated information of TX action for researcher which may help finding new target for further research in APL pathophysiology or new TX resistant APL patients drug designing.
Collapse
Affiliation(s)
- Ananta
- Department of Life Sciences, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - Swati Benerjee
- Department of Life Sciences, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - Paul B. Tchounwou
- RCMI Center for Urban Health Disparities Research and Innovation, Morgan State University, Baltimore, MD 21251, USA
| | - Sanjay Kumar
- Department of Life Sciences, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya, India
| |
Collapse
|
3
|
Cierpikowski P, Leszczyszyn A, Bar J. The Role of Hedgehog Signaling Pathway in Head and Neck Squamous Cell Carcinoma. Cells 2023; 12:2083. [PMID: 37626893 PMCID: PMC10453169 DOI: 10.3390/cells12162083] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth leading malignancy worldwide, with a poor prognosis and limited treatment options. Molecularly targeted therapies for HNSCC are still lacking. However, recent reports provide novel insights about many molecular alterations in HNSCC that may be useful in future therapies. Therefore, it is necessary to identify new biomarkers that may provide a better prediction of the disease and promising targets for personalized therapy. The poor response of HNSCC to therapy is attributed to a small population of tumor cells called cancer stem cells (CSCs). Growing evidence indicates that the Hedgehog (HH) signaling pathway plays a crucial role in the development and maintenance of head and neck tissues. The HH pathway is normally involved in embryogenesis, stem cell renewal, and tissue regeneration. However, abnormal activation of the HH pathway is also associated with carcinogenesis and CSC regulation. Overactivation of the HH pathway was observed in several tumors, including basal cell carcinoma, that are successfully treated with HH inhibitors. However, clinical studies about HH pathways in HNSCC are still rare. In this review, we summarize the current knowledge and recent advances regarding the HH pathway in HNSCC and discuss its possible implications for prognosis and future therapy.
Collapse
Affiliation(s)
- Piotr Cierpikowski
- Department of Maxillofacial Surgery, The Ludwik Rydygier Specialist Hospital, Osiedle Zlotej Jesieni 1, 31-826 Krakow, Poland
| | - Anna Leszczyszyn
- Dental Surgery Outpatient Clinic, 4th Military Clinical Hospital, Weigla 5, 53-114 Wroclaw, Poland;
| | - Julia Bar
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| |
Collapse
|
4
|
Nogueira RLR, de Araújo TBS, Valverde LF, Silva VAO, Cavalcante BRR, Rossi EA, Allahdadi KJ, dos Reis MG, Pereira TA, Coletta RD, Bezerra DP, de Freitas Souza BS, Dias RB, Rocha CAG. Arsenic Trioxide Triggers Apoptosis of Metastatic Oral Squamous Cells Carcinoma with Concomitant Downregulation of GLI1 in Hedgehog Signaling. Biomedicines 2022; 10:biomedicines10123293. [PMID: 36552049 PMCID: PMC9775978 DOI: 10.3390/biomedicines10123293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Given the lack of advances in Oral Squamous Cell Carcinoma (OSCC) therapy in recent years, pharmacological strategies to block OSCC-related signaling pathways have gained prominence. The present study aimed to evaluate the therapeutic potential of Arsenic Trioxide (ATO) concerning its antitumoral effects and the inhibition of the Hedgehog (HH) pathway in OSCC. Initially, ATO cytotoxicity was assessed in a panel of cell lines. Cell viability, cell cycle, death patterns, and cell morphology were analyzed, as well as the effect of ATO on the expression of HH pathway components. After the cytotoxic assay, HSC3 cells were chosen for all in vitro assays. ATO increased apoptotic cell death and nuclear fragmentation in the sub-G1 cell cycle phase and promoted changes in cell morphology. In addition, the reduced expression of GLI1 indicated that ATO inhibits HH activity. The present study provides evidence of ATO as an effective cytotoxic drug for oral cancer treatment in vitro.
Collapse
Affiliation(s)
- Raphael Luís Rocha Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Taís Bacelar Sacramento de Araújo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
| | - Ludmila Faro Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
| | - Viviane Aline Oliveira Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Bruno Raphael Ribeiro Cavalcante
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Erik Aranha Rossi
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
| | - Kyan James Allahdadi
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
| | - Mitermayer Galvão dos Reis
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Thiago Almeida Pereira
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ricardo D. Coletta
- Department of Oral Diagnosis, School of Dentistry University of Campinas, Piracicaba 13414-903, São Paulo, Brazil
- Graduate Program in Oral Biology, School of Dentistry University of Campinas, Piracicaba 13414-903, São Paulo, Brazil
| | - Daniel Pereira Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
| | - Rosane Borges Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
| | - Clarissa A. Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
- Correspondence: ; Tel.: +55-71-3176-2209
| |
Collapse
|
5
|
Kumar S, Tchounwou PB. p53 as a unique target of action of cisplatin in acute leukaemia cells. J Cell Mol Med 2022; 26:4727-4739. [PMID: 35946055 PMCID: PMC9443951 DOI: 10.1111/jcmm.17502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/18/2022] [Accepted: 06/26/2022] [Indexed: 12/03/2022] Open
Abstract
Acute promyelocytic leukaemia (APL) occurs in approximately 10% of acute myeloid leukaemia patients. Arsenic trioxide (ATO) has been for APL chemotherapy, but recently several ATO-resistant cases have been reported worldwide. Cisplatin (CDDP) enhances the toxicity of ATO in ovarian, lung cancer, chronic myelogenous leukaemia, and HL-60 cells. Hence, the goal of this study was to investigate a novel target of CDDP action in APL cells, as an alternate option for the treatment of ATO-resistant APL patients. We applied biochemical, molecular, confocal microscopy and advanced gene editing (CRISPR-Cas9) techniques to elucidate the novel target of CDDP action and its functional mechanism in APL cells. Our main findings revealed that CDDP activated p53 in APL cells through stress signals catalysed by ATM and ATR protein kinases, CHK1 and CHK2 phosphorylation at Ser 345 and Thr68 residues, and downregulation and dissociation of MDM2-DAXX-HAUSP complex. Our functional studies confirmed that CDDP-induced repression of MDM2-DAXX-HAUSP complex was significantly reversed in both nutilin-3-treated KG1a and p53-knockdown NB4 cells. Our findings also showed that CDDP stimulated an increased number of promyelocytes with dense granules, activated p53 expression, and downregulated MDM2 in liver and bone marrow of APL mice. Principal conclusion of our study highlights a novel mode of action of CDDP targeting p53 expression which may provide a basis for designing new anti-leukaemic compounds for treatment of APL patients.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD‐RCMI Center for Health Disparities ResearchJackson State UniversityJacksonMississippiUSA
- Department of life Sciences, School of Earth, Biological, and Environmental SciencesCentral University South BiharGayaIndia
| | - Paul B. Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD‐RCMI Center for Health Disparities ResearchJackson State UniversityJacksonMississippiUSA
| |
Collapse
|
6
|
Lima DG, do Amaral GCLS, Planello AC, Borgato GB, Guimarães GN, de Souza AP. Combined therapy with cisplatin and 5-AZA-2CdR modifies methylation and expression of DNA repair genes in oral squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2022; 15:131-144. [PMID: 35414841 PMCID: PMC8986466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
The methylation and expression of DNA repair system genes has been studied in several tumor types. These genes have been associated with resistance to chemotherapy treatments by epigenetic regulation. Studies have yet to show the effects of combined therapy using an epigenetic drug (5-aza-2CdR) and cisplatin (CDDP) on DNA repair genes in oral squamous cell carcinoma (OSCC). This study proposed to investigate the effects of CDDP in combination with 5-aza-2CdR on the methylation of MGMT and MLH1 genes in oral cancer cells. Oral squamous cell carcinoma cell lineages (SCC-9, SCC-15, and SCC-25) were submitted to 72 hours of treatment: 0.1 μM CDDP (or 4.44 μM SCC-9), 0.1 μM and 0.3 μM 5-aza-2CdR (or 1 μM and 3 μM SCC-9), and the drugs in combination. Cell viability was assessed by MTT, DNA methylation of MGMT and MLH1 genes by Methylation Sensitivity High-Resolution Melting (MS-HRM), and the relative expression of the genes by RT-qPCR. The results show that all treatments reduced cell viability; however, in SCC-15 and SCC-9 (IC50 value), 5-aza-2CdR promotes cell sensitization to cytotoxic effect of cisplatin. The MGMT promoter region was 100% demethylated in the SCC-15 and SCC-25 cells but partially (50%) methylated in SCC-9 before drug treatment. Treatment with IC50 CDDP value kept the methylation status and decreased MGMT expression in SCC-9; MGMT gene in SCC-15 and SCC-25 cells became downregulated after treatment with 5-aza-2CdR. MLH1 was demethylated, but the treatments with low-doses and combined drugs decreased the expression in SCC-9 and SCC-25; however high doses of 5-aza-2CdR and drug combination with IC50 value CDDP increased expression of MLH1 in SCC-9. The data presented suggest that epigenetic drugs associated with chemotherapy have clinical translational potential as a therapy strategy to avoid or reverse cancer resistance, requiring further investigation.
Collapse
Affiliation(s)
- Dieila Giomo Lima
- Department of Bioscience, Piracicaba Dental School, University of Campinas Piracicaba, São Paulo, Brazil
| | | | - Aline Cristiane Planello
- Department of Bioscience, Piracicaba Dental School, University of Campinas Piracicaba, São Paulo, Brazil
| | - Gabriell Bonifacio Borgato
- Department of Bioscience, Piracicaba Dental School, University of Campinas Piracicaba, São Paulo, Brazil
| | - Gustavo Narvaes Guimarães
- Department of Bioscience, Piracicaba Dental School, University of Campinas Piracicaba, São Paulo, Brazil
| | - Ana Paula de Souza
- Department of Bioscience, Piracicaba Dental School, University of Campinas Piracicaba, São Paulo, Brazil
| |
Collapse
|
7
|
Cheng Y, Li S, Gao L, Zhi K, Ren W. The Molecular Basis and Therapeutic Aspects of Cisplatin Resistance in Oral Squamous Cell Carcinoma. Front Oncol 2021; 11:761379. [PMID: 34746001 PMCID: PMC8569522 DOI: 10.3389/fonc.2021.761379] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a kind of malignant tumors with low survival rate and prone to have early metastasis and recurrence. Cisplatin is an alkylating agent which induces DNA damage through the formation of cisplatin-DNA adducts, leading to cell cycle arrest and apoptosis. In the management of advanced OSCC, cisplatin-based chemotherapy or chemoradiotherapy has been considered as the first-line treatment. Unfortunately, only a portion of OSCC patients can benefit from cisplatin treatment, both inherent resistance and acquired resistance greatly limit the efficacy of cisplatin and even cause treatment failure. Herein, this review outline the underlying mechanisms of cisplatin resistance in OSCC from the aspects of DNA damage and repair, epigenetic regulation, transport processes, programmed cell death and tumor microenvironment. In addition, this review summarizes the strategies applicable to overcome cisplatin resistance, which can provide new ideas to improve the clinical therapeutic outcome of OSCC.
Collapse
Affiliation(s)
- Yali Cheng
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Shaoming Li
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Ling Gao
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
8
|
El-Daly SM, Gouhar SA, Gamal-Eldeen AM, Abdel Hamid FF, Ashour MN, Hassan NS. Synergistic Effect of α-Solanine and Cisplatin Induces Apoptosis and Enhances Cell Cycle Arrest in Human Hepatocellular Carcinoma Cells. Anticancer Agents Med Chem 2020; 19:2197-2210. [PMID: 31566136 DOI: 10.2174/1871520619666190930123520] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 01/09/2023]
Abstract
AIM The clinical application of cisplatin is limited by severe side effects associated with high applied doses. The synergistic effect of a combination treatment of a low dose of cisplatin with the natural alkaloid α-solanine on human hepatocellular carcinoma cells was evaluated. METHODS HepG2 cells were exposed to low doses of α-solanine and cisplatin, either independently or in combination. The efficiency of this treatment modality was evaluated by investigating cell growth inhibition, cell cycle arrest, and apoptosis enhancement. RESULTS α-solanine synergistically potentiated the effect of cisplatin on cell growth inhibition and significantly induced apoptosis. This synergistic effect was mediated by inducing cell cycle arrest at the G2/M phase, enhancing DNA fragmentation and increasing apoptosis through the activation of caspase 3/7 and/or elevating the expression of the death receptors DR4 and DR5. The induced apoptosis from this combination treatment was also mediated by reducing the expression of the anti-apoptotic mediators Bcl-2 and survivin, as well as by modulating the miR-21 expression. CONCLUSION Our study provides strong evidence that a combination treatment of low doses of α-solanine and cisplatin exerts a synergistic anticancer effect and provides an effective treatment strategy against hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sherien M El-Daly
- Medical Biochemistry Department, Medical Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt.,Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki 12622, Cairo, Egypt
| | - Shaimaa A Gouhar
- Medical Biochemistry Department, Medical Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Amira M Gamal-Eldeen
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki 12622, Cairo, Egypt.,Biochemistry Department, National Research Centre, Dokki, Cairo, Egypt.,Clinical Laboratory Department, College of Applied Medical Sciences, Taif University, At Taif 26521, Saudi Arabia
| | - Fatma F Abdel Hamid
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Magdi N Ashour
- Medical Biochemistry Department, Medical Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Nahla S Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Hu WC, Teo WH, Huang TF, Lee TC, Lo JF. Combinatorial Low Dose Arsenic Trioxide and Cisplatin Exacerbates Autophagy via AMPK/STAT3 Signaling on Targeting Head and Neck Cancer Initiating Cells. Front Oncol 2020; 10:463. [PMID: 32351887 PMCID: PMC7174769 DOI: 10.3389/fonc.2020.00463] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly lethal disease with high-level of epidemic both in the world and Taiwan. Previous studies support that head and neck cancer-initiating cells (HN-CICs), a subpopulation of cancer cells with enhanced stemness properties, contribute to therapy resistance and tumor recurrence. Arsenic trioxide (As2O3; ATO) has shown to be an effective anti-cancer drug targeting acute promyelocytic leukemia (APL). Combinatorial treatment with high dose of ATO and cisplatin (CDDP) exert synergistic apoptotic effects in cancer cell lines of various solid tumors, however, it may cause of significant side effect to the patients. Nevertheless, none has reported the anti-cancerous effect of ATO/CDDP targeting HN-CICs. In this study, we aim to evaluate the low dose combination of ATO with conventional chemo-drugs CDDP treatment on targeting HN-CICs. We first analyzed the inhibitory tumorigenicity of co-treatment with ATO and chemo-drugs on HN-CICs which are enriched from HNSCC cells. We observed that ATO/CDDP therapeutic regimen successfully synergized the cell death on HN-CICs with a Combination Index (CI) <1 by Chou-Talalay's analysis in vitro. Interestingly, the ATO/CDDP regimen also induced exaggerated autophagy on HN-CICs. Additionally, this drug combination strategy also empowered both preventive and therapeutic effect by in vivo xenograft assays. Finally, we provide the underlying molecular mechanisms of ATO-based therapeutic regimen on HN-CICs. Together, low dose of combinatorial ATO/CDDP regimen induced cell death as well as exacerbated autophagy via AMPK-STAT3 mediated pathway in HN-CICs.
Collapse
Affiliation(s)
- Wei-Chun Hu
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Wan-Huai Teo
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Tung-Fu Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Te-Chang Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jeng-Fan Lo
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
10
|
Miodragović Ð, Swindell EP, Waxali ZS, Bogachkov A, O'Halloran TV. Beyond Cisplatin: Combination Therapy with Arsenic Trioxide. Inorganica Chim Acta 2019; 496:119030. [PMID: 32863421 PMCID: PMC7453736 DOI: 10.1016/j.ica.2019.119030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Platinum drugs (cisplatin, oxaliplatin, and carboplatin) and arsenic trioxide are the only commercial inorganic non-radioactive anticancer drugs approved by the US Food and Drug Administration. Numerous efforts are underway to take advantage of the synergy between the anticancer activity of cisplatin and arsenic trioxide - two drugs with strikingly different mechanisms of action. These include co-encapsulation of the two drugs in novel nanoscale delivery systems as well as the development of small molecule agents that combine the activity of these two inorganic materials. Several of these new molecular entities containing Pt-As bonds have broad anticancer activity, are robust in physiological buffer solutions, and form stable complexes with biopolymers. This review summarizes results from a number of preclinical studies involving the combination of cisplatin and As2O3, co-encapsulation and nanoformulation efforts, and the chemistry and cytotoxicity of the first member of platinum anticancer agents with an arsenous acid moiety bound to the platinum(II) center: arsenoplatins.
Collapse
Affiliation(s)
- Ðenana Miodragović
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Northeastern Illinois University, 5500 North St Louis Avenue, Chicago, Illinois 60625, United States
| | - Elden P Swindell
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Zohra Sattar Waxali
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Abraham Bogachkov
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Thomas V O'Halloran
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
11
|
Rocha FV, Farias RL, Lima MA, Batista VS, Nascimento-Júnior NM, Garrido SS, Leopoldino AM, Goto RN, Oliveira AB, Beck J, Landvogt C, Mauro AE, Netto AVG. Computational studies, design and synthesis of Pd(II)-based complexes: Allosteric inhibitors of the Human Topoisomerase-IIα. J Inorg Biochem 2019; 199:110725. [PMID: 31374424 DOI: 10.1016/j.jinorgbio.2019.110725] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/20/2019] [Accepted: 05/27/2019] [Indexed: 12/30/2022]
Abstract
Herein, a robust docking protocol was developed by using a low-cost workflow to highlight the modulation at ATPase domain from Human Topoisomerase-IIα (TOP2A) towards four novel Pd(II)-complexes bearing N,S-donor ligands. In vitro TOP2A inhibition assay confirmed the ability of them to prevent the enzyme functions into concentration ranging at 6.25-25μM. These results exhibited more effectivity than anticancer agent etoposide (35μM) and merbarone (40-50μM). The compounds were screened via Resazurin assay against MCF-7, MDA-MB-231 (Human breast), DU-145 (Human prostate), A549 (Human lung) and Cal27 (Human tongue) tumor cell lines revealing great cytotoxic effects, primarily to MCF-7 (IC50=1.81-4.46μM). As well, 1-4 exhibited their selectivity index (SI) higher than cisplatin against HEK-293 (human kidney) normal cells, at least 11.6-fold (SI1-4=1.4-5.0; SIcis=0.12). Further, Red Blood Cell hemolytic test suggested in vitro non-toxic character for compound 4, previously evaluated as the most effective TOP2A inhibitor.
Collapse
Affiliation(s)
- Fillipe V Rocha
- UFSCar - Univ Federal de São Carlos, Departamento de Química, São Carlos, Brazil.
| | - Renan L Farias
- UNESP - Univ Estadual Paulista, Instituto de Química, Departamento de Química Geral e Inorgânica, Araraquara, Brazil
| | - Mauro A Lima
- UFSCar - Univ Federal de São Carlos, Departamento de Química, São Carlos, Brazil
| | - Victor S Batista
- UNESP - Univ Estadual Paulista, Instituto de Química, Departamento de Química Orgânica, Araraquara, Brazil
| | | | - Saulo S Garrido
- UNESP - Univ Estadual Paulista, Instituto de Química, Departamento de Bioquímica e Tecnologia Química, Araraquara, Brazil
| | - Andréia M Leopoldino
- USP - Univ de São Paulo, Department of Clinical Analyses, Toxicology and Food Sciences, Ribeirão Preto, Brazil
| | - Renata N Goto
- USP - Univ de São Paulo, Department of Clinical Analyses, Toxicology and Food Sciences, Ribeirão Preto, Brazil
| | - Adriano B Oliveira
- UFS - Univ Federal de Sergipe, Departamento de Química, São Cristóvão, Brazil
| | - Johannes Beck
- Rheinische Friedrich-Wilhelms-Universität Bonn, Institut für Anorganische Chemie, Bonn, Germany
| | - Christian Landvogt
- Rheinische Friedrich-Wilhelms-Universität Bonn, Institut für Anorganische Chemie, Bonn, Germany
| | - Antônio E Mauro
- UNESP - Univ Estadual Paulista, Instituto de Química, Departamento de Química Geral e Inorgânica, Araraquara, Brazil
| | - Adelino V G Netto
- UNESP - Univ Estadual Paulista, Instituto de Química, Departamento de Química Geral e Inorgânica, Araraquara, Brazil
| |
Collapse
|
12
|
Kumar S, Farah IO, Tchounwou PB. Trisenox induces cytotoxicity through phosphorylation of mitogen-activated protein kinase molecules in acute leukemia cells. J Biochem Mol Toxicol 2018; 32:e22207. [PMID: 30091188 PMCID: PMC6192836 DOI: 10.1002/jbt.22207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/23/2018] [Accepted: 07/06/2018] [Indexed: 01/03/2023]
Abstract
Trisenox (TX) has been used successfully for the treatment of acute promyelocytic leukemia (APL) patients. TX-induced cytotoxicity in APL cells remains poorly understood. In this study, we investigated the molecular mechanism of TX cytotoxicity using APL cell lines. We assessed TX toxicity by quantitatively measuring lactate dehydrogenase levels. Inhibition of cell cycle progression was assessed by confocal microscopy of Ki-67 expression. Apoptosis was evaluated by Western blot analysis of apoptotic proteins expression, immunocytochemistry, and confocal imaging of annexin V and propidium iodide. Mitogen-activated protein kinase (MAPK) signaling cascade was analyzed by Western blot analysis and inhibitor-based experiments with APL cells. We found that TX-induced cytotoxicity inhibited APL cell cycle progression. TX also induced significant (P < 0.05) changes in the expression levels of apoptotic molecules and activated the phosphorylation of MAPK signaling pathways in APL cells. Understanding the mechanism of TX cytotoxicity would be helpful in the design of new APL drugs.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, College of Science, Engineering and Technology
| | - Ibrahim O. Farah
- Department of Biology, Jackson State University, 1400 J.R Lynch Street, Box18750, Jackson, Mississippi, MS39217, USA
| | - Paul B. Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, College of Science, Engineering and Technology
| |
Collapse
|
13
|
Oral Administration of Clinical Stage Drug Candidate SENS-401 Effectively Reduces Cisplatin-induced Hearing Loss in Rats. Otol Neurotol 2018; 38:1355-1361. [PMID: 28796092 DOI: 10.1097/mao.0000000000001546] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
HYPOTHESIS SENS-401, an oral clinical-stage drug, may reduce cisplatin-induced hearing loss and cochlear damage in an in vivo model. BACKGROUND Cisplatin is commonly associated with hearing loss, causing significant learning and behavioral difficulties in the pediatric cancer population, and for which there are currently no clinical solutions. SENS-401 has previously been shown to improve acoustic trauma-induced hearing loss in vivo. METHODS The effect of SENS-401 (R-azasetron besylate) on cisplatin IC50 values was evaluated in a panel of cisplatin-sensitive cell lines (NIH:OVCAR-3, SK-N-AS, NCI-H460, FaDu). Auditory brainstem response and distortion product otoacoustic emission tests were performed in a rat model of cisplatin-induced hearing-loss (8 mg/kg, day 1) at baseline, and after 14 days of SENS-401 (6.6, 13.2, 26.4 mg/kg/d). Cochlear outer hair cells were counted after immunolabeling for myosin-VIIa. RESULTS Cisplatin cytotoxicity was not impacted by the addition of SENS-401 (up to 10 μM) in any of the cell types evaluated. In vivo, all SENS-401 doses significantly improved auditory brainstem response threshold shift (up to 30 dB) and distortion product otoacoustic emission amplitude loss (up to 19 dB) over placebo. Body weight and survival were not significantly different between rats receiving placebo and those receiving 26.4 mg/kg SENS-401. Significantly more surviving outer hair cells were present after SENS-401 treatment compared with placebo (p < 0.001), with up to 11-fold more in the basal turn of the cochlea. CONCLUSION In vivo and in vitro data support the otoprotective potential and tolerability of SENS-401 without impacting chemotherapeutic potential. Oral SENS-401 is a promising candidate for treating cisplatin-induced ototoxicity.
Collapse
|
14
|
Upreti M, Jyoti A, Johnson SE, Swindell EP, Napier D, Sethi P, Chan R, Feddock JM, Weiss HL, O'Halloran TV, Evers BM. Radiation-enhanced therapeutic targeting of galectin-1 enriched malignant stroma in triple negative breast cancer. Oncotarget 2018; 7:41559-41574. [PMID: 27223428 PMCID: PMC5173078 DOI: 10.18632/oncotarget.9490] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/04/2016] [Indexed: 12/14/2022] Open
Abstract
Currently there are no FDA approved targeted therapies for Triple Negative Breast Cancer (TNBC). Ongoing clinical trials for TNBC have focused primarily on targeting the epithelial cancer cells. However, targeted delivery of cytotoxic payloads to the non-transformed tumor associated-endothelium can prove to be an alternate approach that is currently unexplored. The present study is supported by recent findings on elevated expression of stromal galectin-1 in clinical samples of TNBC and our ongoing findings on stromal targeting of radiation induced galectin-1 by the anginex-conjugated arsenic-cisplatin loaded liposomes using a novel murine tumor model. We demonstrate inhibition of tumor growth and metastasis in response to the multimodal nanotherapeutic strategy using a TNBC model with orthotopic tumors originating from 3D tumor tissue analogs (TTA) comprised of tumor cells, endothelial cells and fibroblasts. The ‘rigorous’ combined treatment regimen of radiation and targeted liposomes is also shown to be well tolerated. More importantly, the results presented provide a means to exploit clinically relevant radiation dose for concurrent receptor mediated enhanced delivery of chemotherapy while limiting overall toxicity. The proposed study is significant as it falls in line with developing combinatorial therapeutic approaches for stroma-directed tumor targeting using tumor models that have an appropriate representation of the TNBC microenvironment.
Collapse
Affiliation(s)
- Meenakshi Upreti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA.,Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Amar Jyoti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA.,Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Sara E Johnson
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Elden P Swindell
- Department of Chemistry, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Dana Napier
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Pallavi Sethi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA.,Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Ryan Chan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Jonathan M Feddock
- Department of Radiation Medicine, University of Kentucky Chandler Hospital, Lexington, KY, USA
| | - Heidi L Weiss
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Thomas V O'Halloran
- Department of Chemistry, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - B Mark Evers
- Department of Pathology, University of Kentucky, Lexington, KY, USA.,Department of Surgery, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
15
|
Yan D, Cai X, Feng Y. miR-183 Modulates Cell Apoptosis and Proliferation in Tongue Squamous Cell Carcinoma SCC25 Cell Line. Oncol Res 2016; 24:399-404. [PMID: 28281960 PMCID: PMC7838737 DOI: 10.3727/096504016x14685034103239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
This study was designed to investigate the role of miR-183 in modulating cell growth and apoptosis of tongue squamous cell carcinoma SCC25 cell line. Human squamous epithelial cell and squamous cell carcinoma cell line SCC25 was used, and miR-183 was inhibited. Cell growth, colony formation, and apoptotic rate, as well as the expression of caspase 3 and BCL-xL, were detected. Results showed that miR-183 was significantly overexpressed in the SCC25 cell line when compared with normal control. The miR-183 inhibitor reduced cell growth and colony formation, while the apoptosis percentage was significantly increased. The expression of activated caspase 3 and BCL-xL was obviously up- and downregulated in siRNA-transfected cells, respectively. In conclusion, miR-183 contributed to cell growth and proliferation, and suppressed cell apoptosis in SCC25 cells. Therefore, miR-183 might serve as a therapeutic target in tongue squamous cell carcinoma (TSCC).
Collapse
Affiliation(s)
- Dayong Yan
- *Oral and Maxillofacial Surgery, The Central Hospital of Zhengzhou, University of Zhengzhou, Zhengzhou, Henan, P.R. China
| | - Xiaoqing Cai
- *Oral and Maxillofacial Surgery, The Central Hospital of Zhengzhou, University of Zhengzhou, Zhengzhou, Henan, P.R. China
| | - Yu Feng
- †Anesthesia Medicine, The Central Hospital of Zhengzhou, University of Zhengzhou, Zhengzhou, Henan, P.R. China
| |
Collapse
|
16
|
Kumar S, Tchounwou PB. Molecular mechanisms of cisplatin cytotoxicity in acute promyelocytic leukemia cells. Oncotarget 2016; 6:40734-46. [PMID: 26486083 PMCID: PMC4747365 DOI: 10.18632/oncotarget.5754] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/19/2015] [Indexed: 11/25/2022] Open
Abstract
Cis-diamminedichloroplatinum (II) (cisplatin) is a widely used anti-tumor drug for the treatment of a broad range of human malignancies with successful therapeutic outcomes for head and neck, ovarian, and testicular cancers. It has been found to inhibit cell cycle progression and to induce oxidative stress and apoptosis in acute promyelocytic leukemia (APL) cells. However, its molecular mechanisms of cytotoxic action are poorly understood. We hypothesized that cisplatin induces cytotoxicity through DNA adduct formation, oxidative stress, transcriptional factors (p53 and AP-1), cell cycle regulation, stress signaling and apoptosis in APL cells. We used the APL cell line as a model, and applied a variety of molecular tools to elucidate the cytotoxic mode of action of cisplatin. We found that cisplatin inhibited cell proliferation by a cytotoxicity, characterized by DNA damage and modulation of oxidative stress. Cisplatin also activated p53 and phosphorylated activator protein (AP-1) component, c-Jun at serine (63, 73) residue simultaneously leading to cell cycle arrest through stimulation of p21 and down regulation of cyclins and cyclin dependent kinases in APL cell lines. It strongly activated the intrinsic pathway of apoptosis through alteration of the mitochondrial membrane potential, release of cytochrome C, and up-regulation of caspase 3 activity. It also down regulated the p38MAPK pathway. Overall, this study highlights the molecular mechanisms that underline cisplatin toxicity to APL cells, and provides insights into selection of novel targets and/or design of therapeutic agents to treat APL.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, Mississippi 39217, USA
| | - Paul B Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, Mississippi 39217, USA
| |
Collapse
|
17
|
Mitochondrial fission determines cisplatin sensitivity in tongue squamous cell carcinoma through the BRCA1-miR-593-5p-MFF axis. Oncotarget 2016; 6:14885-904. [PMID: 25912308 PMCID: PMC4558123 DOI: 10.18632/oncotarget.3659] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/28/2015] [Indexed: 02/05/2023] Open
Abstract
Cisplatin has been widely employed as a cornerstone chemotherapy treatment for a wide spectrum of solid neoplasms; increasing tumor responsiveness to cisplatin has been a topic of interest for the past 30 years. Strong evidence has indicated that mitochondrial fission participates in the regulation of apoptosis in many diseases; however, whether mitochondrial fission regulates cisplatin sensitivity remains poorly understood. Here, we show that MFF mediated mitochondrial fission and apoptosis in tongue squamous cell carcinoma (TSCC) cells after cisplatin treatment and that miR-593-5p was downregulated in this process. miR-593-5p attenuated mitochondrial fission and cisplatin sensitivity by targeting the 3' untranslated region sequence of MFF and inhibiting its translation. In exploring the underlying mechanism of miR-593-5p downregulation, we observed that BRCA1 transactivated miR-593-5p expression and attenuated cisplatin sensitivity in vitro. The BRCA1-miR-593-5p-MFF axis also affected cisplatin sensitivity in vivo. Importantly, in a retrospective analysis of multiple centers, we further found that the BRCA1-miR-593-5p-MFF axis was significantly associated with cisplatin sensitivity and the survival of patients with TSCC. Together, our data reveal a model for mitochondrial fission regulation at the transcriptional and post-transcriptional levels; we also reveal a new pathway for BRCA1 in determining cisplatin sensitivity through the mitochondrial fission program.
Collapse
|
18
|
Rivera C. Essentials of oral cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:11884-11894. [PMID: 26617944 PMCID: PMC4637760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/20/2015] [Indexed: 06/05/2023]
Abstract
Oral cancer is one of the 10 most common cancers in the world, with a delayed clinical detection, poor prognosis, without specific biomarkers for the disease and expensive therapeutic alternatives. This review aims to present the fundamental aspects of this cancer, focused on squamous cell carcinoma of the oral cavity (OSCC), moving from its definition and epidemiological aspects, addressing the oral carcinogenesis, oral potentially malignant disorders, epithelial precursor lesions and experimental methods for its study, therapies and future challenges. Oral cancer is a preventable disease, risk factors and natural history is already being known, where biomedical sciences and dentistry in particular are likely to improve their poor clinical indicators.
Collapse
Affiliation(s)
- César Rivera
- Department of Basic Biomedical Sciences, University of Talca (UTALCA) TalcaMaule Region, Chile
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas (UNICAMP) PiracicabaSão Paulo, Brazil
- Mass Spectrometry Laboratory, Brazilian Biosciences National Laboratory (LNBio), National Center for Research in Energy and Materials (CNPEM) CampinasSão Paulo, Brazil
| |
Collapse
|
19
|
Manna A, De Sarkar S, De S, Bauri AK, Chattopadhyay S, Chatterjee M. The variable chemotherapeutic response of Malabaricone-A in leukemic and solid tumor cell lines depends on the degree of redox imbalance. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:713-723. [PMID: 26141757 DOI: 10.1016/j.phymed.2015.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 05/12/2015] [Accepted: 05/20/2015] [Indexed: 06/04/2023]
Abstract
PURPOSE The 'two-faced' character of reactive oxygen species (ROS) plays an important role in cancer biology by acting as secondary messengers in intracellular signaling cascades, enhancing cell proliferation and survival, thereby sustaining the oncogenic phenotype. Conversely, enhanced generation of ROS can trigger an oxidative assault leading to a redox imbalance translating into an apoptotic cell death. Intrinsically, cancer cells have higher basal levels of ROS which if supplemented by additional oxidative insult by pro-oxidants can be cytotoxic, an example being Malabaricone-A (MAL-A). MAL-A is a plant derived diarylnonanoid, purified from fruit rind of the plant Myristica malabarica whose anti-cancer activity has been demonstrated in leukemic cell lines, the modality of cell death being apoptosis. This study aimed to compare the degree of effectiveness of MAL-A in leukemic vs. solid tumor cell lines. METHODS The cytotoxicity of MAL-A was evaluated by the MTS-PMS cell viability assay in leukemic cell lines (MOLT3, K562 and HL-60) and compared with solid tumor cell lines (MCF7, A549 and HepG2); further studies then proceeded with MOLT3 vs. MCF7 and A549. The contribution of redox imbalance in MAL-A induced cytotoxicity was confirmed by pre-incubating cells with an antioxidant, N-acetyl-L-cysteine (NAC) or a thiol depletor, buthionine sulfoximine (BSO). MAL-A induced redox imbalance was quantitated by flow cytometry, by measuring the generation of ROS and levels of non protein thiols using dichlorofluorescein diacetate (CM-H2DCFDA) and 5-chloromethylfluorescein diacetate (CMFDA) respectively. The activities of glutathione peroxidase (GPx), superoxide dismutase, catalase (CAT), NAD(P)H dehydrogenase (quinone 1) NQO1 and glutathione-S-transferase GST were measured spectrophotometrically. The mitochondrial involvement of MAL-A induced cell death was measured by evaluation of cardiolipin peroxidation using 10-N-nonyl acridine orange (NAO), transition pore activity with calcein-AM, while the mitochondrial transmembrane electrochemical gradient (∆ψ(m)) was measured by JC-1, fluorescence being acquired in a flow cytometer. The apoptotic mode of cell death was evaluated by double staining with annexin V-FITC and propidium iodide (PI), cell cycle analysis by flow cytometry and caspase-3 activity spectrophotometrically. The expression of Nrf2 and HO-1 was examined by western blotting. RESULTS MAL-A demonstrated a higher degree of cytotoxicity in three leukemic cell lines whose IC50 ranged from 12.70 ± 0.10 to 18.10 ± 0.95 µg/ml, whereas in three solid tumor cell lines, the IC50 ranged from 28.10 ± 0.58 to 55.26 ± 5.90 µg/ml. This higher degree of cytotoxicity in MOLT3, a leukemic cell line was due to a higher induction of redox imbalance, evident by both an increased generation of ROS and concomitant depletion of thiols. This was confirmed by pre-incubation with NAC and BSO, wherein NAC decreased MAL-A induced cytotoxicity by 2.04 fold while BSO enhanced MAL-A cytotoxicity and decreased the IC50 by 5.60 fold. However, in solid tumor cell lines (MCF7 and A549), NAC minimally decreased MAL-A induced cytotoxicity, and BSO increased the IC50 by 1.96 and 2.39 fold respectively. Furthermore, the generation of ROS by MAL-A increased maximally in MOLT3 as the fluorescence increased from 44.28 ± 7.85 to 273.99 ± 32.78, and to a lesser degree in solid tumor cell lines, MCF7 (44.28 ± 14.89 to 207.97 ± 70.64) and A549 (37.87 ± 3.24 to 147.12 ± 38.53). In all three cell lines there was a concomitant depletion of thiols as in MOLT3, the GMFC decreased from 340.65 ± 60.39 to 62.67 ± 11.32, in MCF7 (277.82 ± 50.32 to 100.39 ± 31.93) and in A549 (274.05 ± 59.13 to 83.15 ± 21.43). In MOLT3 as compared to MCF7 and A549, decrease in the activities of GPx, CAT, NQO1 and GST was substantially greater. In all cell lines, the MAL-A induced redox imbalance translated into triggering of initial mitochondrial apoptotic events. Here again, MAL-A induced a higher degree of cardiolipin peroxidation in MOLT3 (67.01%) than MCF7 and A549 (29.15% and 44.30%), as also down regulated the mitochondrial transition pore activity from baseline to a higher extent, GMFC being 48.05 ± 2.37 to 10.70 ± 3.97 (MOLT3), 43.55 ± 3.36 to 15.36 ± 0.60 (MCF7) and 39.58 ± 0.4 to 12.65 ± 1.56 (A549). Perturbation of mitochondrial membrane potential evident by a decrease in the ratio of red/green (J-aggregates/monomers) was 134 fold (14.73/0.11) in MOLT3, 45 fold in MCF7 (20.72/0.46) and 34 fold in A549 (22.01/0.64). The extent of apoptosis using a similar concentration of MAL-A was maximal in MOLT3, wherein a 105 fold increase in annexin V binding was evident (0.83 ± 0.51 to 87.08 ± 9.85%) whereas it increased by 43.11 fold in MCF7 (0.69 ± 0.30 to 29.75 ± 11.79%) and 47.52 fold in A549 (0.61 ± 0.31 to 28.99 ± 17.21%). MAL-A induced apoptosis was also associated with a higher degree of caspase-3 activity in MOLT3 vs. MCF7 or A549 which translated into halting of cell cycle progression, evident by an increment in the sub-G0/G1 population [19.26 fold in MOLT3 (0.95 ± 0.45 vs. 18.30 ± 1.90%), 11.01 fold in MCF7 (0.97 ± 0.37 vs. 10.68 ± 0.69%) and 8.58 fold in A549 (1.06 ± 0.45 vs. 9.10 ± 1.05%)]. MAL-A effectively inhibited Nrf2 and HO-1, more prominently in MOLT3. Furthermore, the decreased expression of Nrf2 in MOLT3 correlated with the decreased activities of NQO1 and GST, suggesting that targeting of the Nrf2 anti-oxidant pathway could be considered. CONCLUSION Taken together, MAL-A a pro-oxidant compound is likely to be more effective in leukemias, meriting further pharmacological consideration.
Collapse
Affiliation(s)
- Alak Manna
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244B, Acharya JC Bose Road, Kolkata 700 020, India
| | - Sritama De Sarkar
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244B, Acharya JC Bose Road, Kolkata 700 020, India
| | - Soumita De
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244B, Acharya JC Bose Road, Kolkata 700 020, India
| | - Ajay K Bauri
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | | | - Mitali Chatterjee
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244B, Acharya JC Bose Road, Kolkata 700 020, India .
| |
Collapse
|
20
|
Fan S, Chen WX, Lv XB, Tang QL, Sun LJ, Liu BD, Zhong JL, Lin ZY, Wang YY, Li QX, Yu X, Zhang HQ, Li YL, Wen B, Zhang Z, Chen WL, Li JS. miR-483-5p determines mitochondrial fission and cisplatin sensitivity in tongue squamous cell carcinoma by targeting FIS1. Cancer Lett 2015; 362:183-91. [DOI: 10.1016/j.canlet.2015.03.045] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 03/26/2015] [Accepted: 03/28/2015] [Indexed: 12/21/2022]
|
21
|
Abstract
Arsenic is an enigmatic xenobiotic that causes a multitude of chronic diseases including cancer and also is a therapeutic with promise in cancer treatment. Arsenic causes mitotic delay and induces aneuploidy in diploid human cells. In contrast, arsenic causes mitotic arrest followed by an apoptotic death in a multitude of virally transformed cells and cancer cells. We have explored the hypothesis that these differential effects of arsenic exposure are related by arsenic disruption of mitosis and are differentiated by the target cell's ability to regulate or modify cell cycle checkpoints. Functional p53/CDKN1A axis has been shown to mitigate the mitotic block and to be essential to induction of aneuploidy. More recent preliminary data suggest that microRNA modulation of chromatid cohesion also may play a role in escape from mitotic block and in generation of chromosomal instability. Other recent studies suggest that arsenic may be useful in treatment of solid tumors when used in combination with other cytotoxic agents such as cisplatin.
Collapse
Affiliation(s)
- J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, 505 S. Hancock St, Louisville, KY, 40202, USA,
| |
Collapse
|
22
|
Autophagy interplays with apoptosis and cell cycle regulation in the growth inhibiting effect of Trisenox in HEP-2, a laryngeal squamous cancer. Pathol Oncol Res 2014; 21:103-11. [PMID: 24838151 DOI: 10.1007/s12253-014-9794-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 05/06/2014] [Indexed: 02/02/2023]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is the most common among several types of head and neck cancers. Current treatments have a poor effect on early and advanced cases, and further investigations for novel agents against LSCCs are desirable. In this study, we elucidate the cytotoxic enhancing effect of arsenic trioxide (As2O3) combined with L-buthionine sulfoximine (BSO) in LSCC. The effect of BSO with As2O3 or Cisplatin (CDDP) on cell viability was examined using 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). The reactive oxygen species (ROS) levels, cell cycle, and apoptosis were measured by flow cytometry using 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA), propidium iodide (PI) and annexin V/PI. The acidic vacuolar organelles were visualized by fluorescence microscope and quantified using flow cytometry. Neither CDDP nor As2O3 when used alone reduced the cell viability. BSO was found to enhance only As2O3 sensitivity, leading to G2/M arrest and autophagy with no correlation of ROS induction. This result suggests that modulation of glutathione enhances autophagy, which interplays with apoptosis. In this study, we obtained initial preclinical evidence for the potential efficacy of these drugs in a combined therapy protocol.
Collapse
|
23
|
Nakaoka T, Ota A, Ono T, Karnan S, Konishi H, Furuhashi A, Ohmura Y, Yamada Y, Hosokawa Y, Kazaoka Y. Combined arsenic trioxide-cisplatin treatment enhances apoptosis in oral squamous cell carcinoma cells. Cell Oncol (Dordr) 2014; 37:119-29. [PMID: 24599717 DOI: 10.1007/s13402-014-0167-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) accounts for the majority of oral cancers. Despite recent advances in OSCC diagnostics and therapeutics, the overall survival rate still remains low. Here, we assessed the efficacy of a combinatorial arsenic trioxide (ATO) and cisplatin (CDDP) treatment in human OSCC cells. METHODS The combinatorial effect of ATO/CDDP on the growth and apoptosis of OSCC cell lines HSC-2, HSC-3, and HSC-4 was evaluated using MTT and annexin V assays, respectively. Chou-Talalay analyses were preformed to evaluate the combinatorial effects of ATO/CDDP on the dose-reduction index (DRI). To clarify the mechanism underlying the ATO/CDDP anticancer effect, we also examined the involvement of reactive oxygen species (ROS) in ATO/CDDP-induced apoptosis. RESULTS Combination index (CI) analyses revealed that a synergistic interaction of ATO and CDDP elicits a wide range of effects in HSC-2 cells, with CI values ranging from 0.78 to 0.90, where CI < 1 defines synergism. The CI values in HSC-3 and HSC-4 cells ranged from 0.34 to 0.45 and from 0.60 to 0.92, respectively. In addition, ATO/CDDP yielded favorable DRI values ranging from 1.6-fold to 7.71-fold dose reduction. Compared to mono-therapy, ATO/CDDP combinatorial therapy significantly augmented the loss of mitochondrial potential, caspase-3/7 activity and subsequent apoptosis. These changes were all abrogated by the antioxidant N-acetylcysteine. CONCLUSIONS This study provides the first evidence for a synergistic ATO/CDDP anticancer (apoptotic) activity in OSCC cells with a favorable DRI, thereby highlighting its potential as a combinational therapeutic regime in OSCC.
Collapse
Affiliation(s)
- Toshiki Nakaoka
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zheng CY, Lam SK, Li YY, Fong BMW, Mak JCW, Ho JCM. Combination of arsenic trioxide and chemotherapy in small cell lung cancer. Lung Cancer 2013; 82:222-30. [PMID: 24041618 DOI: 10.1016/j.lungcan.2013.08.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 07/22/2013] [Accepted: 08/25/2013] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Small cell lung cancer (SCLC) carries high mortality despite standard chemotherapy. Arsenic trioxide (ATO) has demonstrated clinical efficacy in leukemia and in vitro activity in various solid tumors. This study was conducted to determine the in vitro and in vivo combination effects of ATO and chemotherapy in SCLC. MATERIALS AND METHODS The in vitro model consisted of 5 SCLC cell lines (H187, H526, H69, H841 and DMS79) and the anti-proliferative effects of ATO, cisplatin, etoposide or combinations thereof were measured. Synergism was determined by calculation of the combination index (CI) according to Chou and Talalay. Assays for apoptosis, intracellular glutathione (GSH) content, and mitochondrial membrane depolarization (MMD) were performed. Arsenic content was measured by inductively coupled plasma-mass spectrometry. Expression level of MRP1, MRP2 and pH2AX was detected by Western blot while cellular pH2AX level was monitored by immunofluorescent staining. An in vivo xenograft model in nude mice was established with a H841 cell line to test the effects of drug combinations. RESULTS All 5 SCLC cell lines were sensitive to ATO, with IC(50) values (48 h) 1.6-8 μM. Synergistic or additive effects were obtained by combining cisplatin with ATO in all 5 cell lines. Combination of etoposide with ATO resulted in antagonistic or barely additive effects. Apoptotic assays and pH2AX immunofluorescent staining corroborated the synergistic combination of ATO and cisplatin. In addition, the ATO/cisplatin combination enhanced MMD, depleted GSH, downregulated MRP2 and elevated intracellular ATO content compared with either ATO or cisplatin alone. In vivo combination of ATO and cisplatin also demonstrated synergism in the H841 xenograft model. CONCLUSIONS There was clinically relevant in vitro activity of ATO in a panel of 5 SCLC cell lines. Significant synergism was demonstrated with the ATO/cisplatin combination, while antagonism was noted with the ATO/etoposide combination in both in vitro and in vivo models.
Collapse
Affiliation(s)
- Chun-yan Zheng
- Division of Respiratory Medicine, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region
| | | | | | | | | | | |
Collapse
|