1
|
Arfeen M, Srivastava A, Srivastava N, Khan RA, Almahmoud SA, Mohammed HA. Design, classification, and adverse effects of NSAIDs: A review on recent advancements. Bioorg Med Chem 2024; 112:117899. [PMID: 39217686 DOI: 10.1016/j.bmc.2024.117899] [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: 06/25/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Inflammation is a hallmark of many diseases, including cancer, neurodegenerative diseases like Alzheimer's, type II diabetes, rheumatoid arthritis, and asthma. Nonsteroidal anti-inflammatory drugs (NSAIDs) have been a cornerstone in the management of various inflammatory, pain, and fever-related conditions. As a result, NSAIDs have found their applications in new therapeutic areas. NSAIDs are known to act by inhibiting the cyclooxygenase (COX) pathway. In recent years, new strategies have been proposed to counter inflammation and develop safer COX inhibitors. This review discusses the design of new COX inhibitors, the derivatization of conventional NSAIDs, and their biological applications. The review also presents an integrated classification of NSAIDs incorporating both traditional chemical-based and function-based approaches, including a brief overview of the NSAIDs of natural origins. Additionally, the review addresses adverse effects associated with different NSAIDs, including effects associated with cardiovascular, renal, and hepatic complications emphasizing the need for the development of new and safer COX inhibitors.
Collapse
Affiliation(s)
- Minhajul Arfeen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Ashish Srivastava
- PSIT-Pranveer Singh Institute of Technology, (Pharmacy), Kanpur, UP 209305, India.
| | - Noopur Srivastava
- Six Sigma Institute of Technology and Science, Rudrapur, Uttarkhand 263153, India.
| | - Riaz A Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Suliman A Almahmoud
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia.
| |
Collapse
|
2
|
Qayyum S, Jabeen A, Ashraf S, Seraj F, Khan KM, Siddiqui RA, Ul-Haq Z. Oxadiazole Derivatives of Diclofenac as an Anti-proliferative Agent for B-cell Non-Hodgkin Lymphoma: An In vitro and In Silico Studies. Med Chem 2024; 20:443-451. [PMID: 38279758 DOI: 10.2174/0115734064290905231228110023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Non-Hodgkin lymphoma of B cell origin is the common type of lymphoma- related malignancy with poor response rate with conventional front-line therapies. AIM The aim of the present study was to investigate the potential of new anti-inflammatory oxadiazole derivatives of Diclofenac as an anti-lymphoma agent through in vitro and in silico approaches. METHODS Anti-lymphoma potential was evaluated by alamar blue technique. MTT assay employed for cytotoxicity. Gene and protein expression studies was performed by qRT-PCR and ELISA respectively. Docking studies was performed by using MOE program. RESULTS Among five diclofenac derivatives, (II) showed promising anti-lymphoma effects, where it inhibited the expression of BCL-2, p-38 MAPK and TGF-β in both follicular and Burkitt's lymphoma cells and was non-toxic against normal human fibroblast cells. The in silico studies against BCL-2 revealed that the unsubstituted Sulphur group in (II) is involved in the crucial interactions with the binding site residue. CONCLUSION The compound (II) can be a potential therapeutic candidate for B-cell non-Hodgkin lymphoma and deserves further development as a novel anti-lymphoma agent.
Collapse
Affiliation(s)
- Shaista Qayyum
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Almas Jabeen
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Sajda Ashraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Faiza Seraj
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Khalid Mohammad Khan
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Rafat Ali Siddiqui
- Food Chemistry and Nutritional Science Research Laboratory, Virginia State University, Petersburg, VA, USA
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| |
Collapse
|
3
|
Zhao L, Hsiao T, Stonesifer C, Daniels J, Garcia-Saleem TJ, Choi J, Geskin L, Rook AH, Wood GS. The Robust Tumoricidal Effects of Combined BET/HDAC Inhibition in Cutaneous T-Cell Lymphoma Can Be Reproduced by ΔNp73 Depletion. J Invest Dermatol 2022; 142:3253-3261.e4. [PMID: 35787399 PMCID: PMC9691518 DOI: 10.1016/j.jid.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/11/2022] [Accepted: 06/07/2022] [Indexed: 01/05/2023]
Abstract
Combined BET inhibitor/histone deacetylase inhibitor treatment induces marked apoptosis of cutaneous T-cell lymphoma (CTCL) with minimal normal T-cell toxicity. At 96 hours when apoptosis was extensive, a majority of CTCL lines showed ≥2-fold suppression of T-cell survival factors (e.g., AKT1, BCL2 antiapoptotic factors, BIRC5, CD40, CD70, GADD45A, PRKCA, TNFRSF1B, ΔNp73) and ≥2-fold upregulation of proapoptotic factors and tumor suppressors (e.g., ATM, BAK, BIM, multiple caspases, FHIT, HIC1, MGMT, NOD1) (P < 0.05). The largest alterations were in TP73 isoform expression, resulting in increased TAp73/ΔNp73 ratios in CTCL lines and leukemic Sézary cells. Targeted ΔNp73 inhibition by small interfering RNA knockdown resulted in robust CTCL apoptosis comparable with that induced by BET inhibitor/histone deacetylase inhibitor with minimal normal T-cell toxicity. Chromatin immunoprecipitation analysis showed that BET inhibitor/histone deacetylase inhibitor treatment reduced RNA polymerase II binding to ΔNp73, MYC, and AKT1 while increasing its binding to TAp73. CTCL skin lesions expressed both TAp73 and ΔNp73 isoforms in situ. In aggregate, these findings implicate TAp73/ΔNp73 balance as a major factor governing CTCL survival, show that the expression of p73 isoforms can be altered by molecular biological and pharmaceutical means, show that p73 isoforms are expressed across the entire CTCL clinical spectrum, and identify the p73 pathway as a potential target for therapeutics.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tony Hsiao
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Connor Stonesifer
- Department of Dermatology, Columbia University, New York, New York, USA
| | - Jay Daniels
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Ilinois, USA
| | | | - Jaehyuk Choi
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Ilinois, USA
| | - Larisa Geskin
- Department of Dermatology, Columbia University, New York, New York, USA
| | - Alain H Rook
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gary S Wood
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
4
|
Deb J, Lakshman TR, Ghosh I, Jana SS, Paine TK. Mechanistic studies of in vitro anti-proliferative and anti-inflammatory activities of the Zn(ii)-NSAID complexes of 1,10-phenanthroline-5,6-dione in MDA-MB-231 cells. Dalton Trans 2021; 49:11375-11384. [PMID: 32766641 DOI: 10.1039/d0dt01721c] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Two zinc(ii)-NSAID complexes [(phendione)ZnII(NPR)2(H2O)2] (1) and [(phendione)ZnII(MFN)2] (2) (HNPR = naproxen and HMFN = mefenamic acid) of 1,10-phenanthroline-5,6-dione (phendione) were isolated and characterized to evaluate their potential as anti-cancer agents. Each of the complexes contains two equivalents of NSAID per zinc(ii)-phendione unit. The complexes are stable in solution under cell culture conditions. Cytotoxic assay on the human breast cancer cell line (MDA-MB-231) reveals that the anti-proliferative activity of phendione is retained in both the complexes. The anti-inflammatory properties of NSAIDs are also preserved in the metal complexes as evident from the PGE2 assay. Both 1 and 2 exhibit selective COX-1 inhibition at a low concentration. Furthermore, the zinc(ii)-naproxen complex (1) disrupts the intercellular bridges displaying in vitro delay in cellular migration and down-regulation of EMT-related genes. The mechanistic studies indicate that the ternary complexes are more active compared to cisplatin and have the potential to overcome cisplatin resistance in MDA MB 231 cells. These findings demonstrate that the zinc(ii)-NSAID complexes are worthy of further in vivo studies for their promising anti-tumor potential.
Collapse
Affiliation(s)
- Jolly Deb
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Triloke Ranjan Lakshman
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Ivy Ghosh
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Siddhartha Sankar Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Tapan Kanti Paine
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
5
|
Pavlidis A, Piperi C, Papadavid E. Novel therapeutic approaches for cutaneous T cell lymphomas. Expert Rev Clin Immunol 2021; 17:629-641. [PMID: 33890833 DOI: 10.1080/1744666x.2021.1919085] [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] [Indexed: 12/13/2022]
Abstract
Introduction: Cutaneous T-cell lymphoma (CTCL) is a rare non-Hodgkin's lymphoma, characterized by malignant T cells infiltrating the skin. CTCL exhibits vast heterogeneity which complicates diagnosis and therapeutic strategies. Current CTCL treatment includes skin-directed therapies (such as topical corticosteroid, topical mechlorethamine, topical bexarotene, ultraviolet phototherapy and localized radiotherapy), total skin electron beam therapy and systemic therapies. Elucidation of molecular and signaling pathways underlying CTCL pathogenesis leads to identification of innovative and personalized treatment schemes.Areas covered: The authors reviewed the molecular and immunological aspects of CTCL with special focus on Mycosis Fungoides (MF), Sézary Syndrome (SS) and associated systemic treatment. A literature search was conducted in PubMed and Web of Science for peer-reviewed articles published until November 2020. Novel treatment approaches including retinoids, targeted therapies, immune checkpoint and JAK/STAT inhibitors, histones deacetylase (HDAC) and mTOR inhibitors as well as proteasome inhibitors, are discussed as potential therapeutic tools for the treatment of CTCL.Expert opinion: Novel therapeutic agents exhibit potential beneficial effects in CTCL patients of high need for therapy such as refractory early stage cutaneous and advanced stage disease. Therapeutic schemes employing a combination of novel agents with current treatment options may prove valuable for the future management of CTCL patients.
Collapse
Affiliation(s)
- Antreas Pavlidis
- 2nd Department of Dermatology and Venereal Diseases, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Papadavid
- 2nd Department of Dermatology and Venereal Diseases, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
6
|
Sumarni U, Reidel U, Eberle J. Targeting Cutaneous T-Cell Lymphoma Cells by Ingenol Mebutate (PEP005) Correlates with PKCδ Activation, ROS Induction as Well as Downregulation of XIAP and c-FLIP. Cells 2021; 10:cells10050987. [PMID: 33922439 PMCID: PMC8146015 DOI: 10.3390/cells10050987] [Citation(s) in RCA: 3] [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] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 01/25/2023] Open
Abstract
New therapeutic strategies are needed for cutaneous T-cell lymphoma (CTCL), and the plant extract ingenol mebutate (PEP005) may be considered. PEP005 has been approved for actinic keratosis, and proapoptotic activities were described in different cancer cells. Here, we aimed to investigate its efficacy in four CTCL cell lines and its mode of action. While HuT-78 and HH responded with induced apoptosis as well as with loss of cell viability and cell proliferation, MyLa and SeAx remained resistant. Interestingly, both sensitive and resistant cells showed caspase-8 activation and enhanced levels of reactive oxygen species (ROS), while final caspase-3 activation was restricted to sensitive cells. Apoptosis induction was prevented by the caspase inhibitor QVD-Oph as well as by the antioxidant vitamin E. Caspase activation by PEP005 may be explained to some extent by the downregulation of the caspase antagonistic proteins c-FLIP and XIAP in sensitive cells, whereas both proteins were strongly expressed in resistant cells. Finally, PEP005 resulted in the activation of proapoptotic PKCδ, and the PKC inhibitor bisindolylmaleimide I reduced apoptosis, caspase-3 processing and ROS production, as well as restored cell viability. In conclusion, PKCδ appeared as a central player in apoptosis regulation in CTCL cells, also suggesting its therapeutic targeting.
Collapse
MESH Headings
- Apoptosis
- CASP8 and FADD-Like Apoptosis Regulating Protein/antagonists & inhibitors
- Cell Cycle
- Cell Movement
- Cell Proliferation
- Diterpenes/pharmacology
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Lymphoma, T-Cell, Cutaneous/drug therapy
- Lymphoma, T-Cell, Cutaneous/genetics
- Lymphoma, T-Cell, Cutaneous/metabolism
- Lymphoma, T-Cell, Cutaneous/pathology
- Protein Kinase C-delta/genetics
- Protein Kinase C-delta/metabolism
- Reactive Oxygen Species/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Tumor Cells, Cultured
- X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors
Collapse
|
7
|
Rindler K, Bauer WM, Jonak C, Wielscher M, Shaw LE, Rojahn TB, Thaler FM, Porkert S, Simonitsch-Klupp I, Weninger W, Mayerhoefer ME, Farlik M, Brunner PM. Single-Cell RNA Sequencing Reveals Tissue Compartment-Specific Plasticity of Mycosis Fungoides Tumor Cells. Front Immunol 2021; 12:666935. [PMID: 33968070 PMCID: PMC8097053 DOI: 10.3389/fimmu.2021.666935] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022] Open
Abstract
Mycosis fungoides (MF) is the most common primary cutaneous T-cell lymphoma. While initially restricted to the skin, malignant cells can appear in blood, bone marrow and secondary lymphoid organs in later disease stages. However, only little is known about phenotypic and functional properties of malignant T cells in relationship to tissue environments over the course of disease progression. We thus profiled the tumor micromilieu in skin, blood and lymph node in a patient with advanced MF using single-cell RNA sequencing combined with V-D-J T-cell receptor sequencing. In skin, we identified clonally expanded T-cells with characteristic features of tissue-resident memory T-cells (TRM, CD69+CD27-NR4A1+RGS1+AHR+). In blood and lymph node, the malignant clones displayed a transcriptional program reminiscent of a more central memory-like phenotype (KLF2+TCF7+S1PR1+SELL+CCR7+), while retaining tissue-homing receptors (CLA, CCR10). The skin tumor microenvironment contained potentially tumor-permissive myeloid cells producing regulatory (IDO1) and Th2-associated mediators (CCL13, CCL17, CCL22). Given their expression of PVR, TNFRSF14 and CD80/CD86, they might be under direct control by TIGIT+CTLA4+CSF2+TNFSF14+ tumor cells. In sum, this study highlights the adaptive phenotypic and functional plasticity of MF tumor cell clones. Thus, the TRM-like phenotype enables long-term skin residence of MF cells. Their switch to a TCM-like phenotype with persistent skin homing molecule expression in the circulation might explain the multi-focal nature of MF.
Collapse
Affiliation(s)
- Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Wielscher
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Lisa E Shaw
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Thomas B Rojahn
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Felix M Thaler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Stefanie Porkert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Marius E Mayerhoefer
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Brandolini L, Antonosante A, Giorgio C, Bagnasco M, d'Angelo M, Castelli V, Benedetti E, Cimini A, Allegretti M. NSAIDs-dependent adaption of the mitochondria-proteasome system in immortalized human cardiomyocytes. Sci Rep 2020; 10:18337. [PMID: 33110169 PMCID: PMC7591859 DOI: 10.1038/s41598-020-75394-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
The progressive consumption growth of non-steroidal anti-inflammatory drugs (NSAIDs) has progressively raised the attention toward the gastrointestinal, renal, and cardiovascular toxicity. Increased risk of cardiovascular diseases was strictly associated with the usage of COX-2 selective NSAIDs. Other studies allowed to clarify that the cardiovascular risk is not limited to COX-2 selective but also extended to non-selective NSAIDs, such as Diclofenac and Ketoprofen. To date, although a less favorable cardiovascular risk profile for Diclofenac as compared to Ketoprofen is reported, the mechanisms through which NSAIDs cause adverse cardiovascular events are not entirely understood. The present study aimed to evaluate the effects of Ketoprofen in comparison with Diclofenac in immortalized human cardiomyocytes. The results obtained highlight the dose-dependent cardiotoxicity of Diclofenac compared to Ketoprofen. Despite both drugs induce the increase in ROS production, decrease of mitochondrial membrane potential, and proteasome activity modulation, only Diclofenac exposure shows a marked alteration of these intracellular parameters, leading to cell death. Noteworthy, Diclofenac decreases the proteasome 26S DC and this scenario may be dependent on the intracellular overload of oxidized proteins. The data support the hypothesis that immortalized human cardiomyocytes exposed to Ketoprofen are subjected to tolerable stress events, conversely Diclofenac exposition triggers cell death.
Collapse
Affiliation(s)
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | | | | | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy. .,Sbarro Institute for Cancer Research and Molecular Medicine and Centre for Biotechnology, Temple University, Philadelphia, USA.
| | | |
Collapse
|
9
|
Bindu S, Mazumder S, Bandyopadhyay U. Non-steroidal anti-inflammatory drugs (NSAIDs) and organ damage: A current perspective. Biochem Pharmacol 2020; 180:114147. [PMID: 32653589 PMCID: PMC7347500 DOI: 10.1016/j.bcp.2020.114147] [Citation(s) in RCA: 659] [Impact Index Per Article: 164.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Owing to the efficacy in reducing pain and inflammation, non-steroidal anti-inflammatory drugs (NSAIDs) are amongst the most popularly used medicines confirming their position in the WHO's Model List of Essential Medicines. With escalating musculoskeletal complications, as evident from 2016 Global Burden of Disease data, NSAID usage is evidently unavoidable. Apart from analgesic, anti-inflammatory and antipyretic efficacies, NSAIDs are further documented to offer protection against diverse critical disorders including cancer and heart attacks. However, data from multiple placebo-controlled trials and meta-analyses studies alarmingly signify the adverse effects of NSAIDs in gastrointestinal, cardiovascular, hepatic, renal, cerebral and pulmonary complications. Although extensive research has elucidated the mechanisms underlying the clinical hazards of NSAIDs, no review has extensively collated the outcomes on various multiorgan toxicities of these drugs together. In this regard, the present review provides a comprehensive insight of the existing knowledge and recent developments on NSAID-induced organ damage. It precisely encompasses the current understanding of structure, classification and mode of action of NSAIDs while reiterating on the emerging instances of NSAID drug repurposing along with pharmacophore modification aimed at safer usage of NSAIDs where toxic effects are tamed without compromising the clinical benefits. The review does not intend to vilify these 'wonder drugs'; rather provides a careful understanding of their side-effects which would be beneficial in evaluating the risk-benefit threshold while rationally using NSAIDs at safer dose and duration.
Collapse
Affiliation(s)
- Samik Bindu
- Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal 736101 India
| | - Somnath Mazumder
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Uday Bandyopadhyay
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, West Bengal, India; Division of Molecular Medicine, Bose Institute, P-1/12, CIT Rd, Scheme VIIM, Kankurgachi, Kolkata, West Bengal 700054 India.
| |
Collapse
|
10
|
Soltan MY, Sumarni U, Assaf C, Langer P, Reidel U, Eberle J. Key Role of Reactive Oxygen Species (ROS) in Indirubin Derivative-Induced Cell Death in Cutaneous T-Cell Lymphoma Cells. Int J Mol Sci 2019; 20:ijms20051158. [PMID: 30866411 PMCID: PMC6429192 DOI: 10.3390/ijms20051158] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 01/05/2023] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) may develop a highly malignant phenotype in its late phase, and patients may profit from innovative therapies. The plant extract indirubin and its chemical derivatives represent new and promising antitumor strategies. This first report on the effects of an indirubin derivative in CTCL cells shows a strong decrease of cell proliferation and cell viability as well as an induction of apoptosis, suggesting indirubin derivatives for therapy of CTCL. As concerning the mode of activity, the indirubin derivative DKP-071 activated the extrinsic apoptosis cascade via caspase-8 and caspase-3 through downregulation of the caspase antagonistic proteins c-FLIP and XIAP. Importantly, a strong increase of reactive oxygen species (ROS) was observed as an immediate early effect in response to DKP-071 treatment. The use of antioxidative pre-treatment proved the decisive role of ROS, which turned out upstream of all other proapoptotic effects monitored. Thus, reactive oxygen species appear as a highly active proapoptotic pathway in CTCL, which may be promising for therapeutic intervention. This pathway can be efficiently activated by an indirubin derivative.
Collapse
Affiliation(s)
- Marwa Y Soltan
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
- Department of Dermatology and Venereology, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt.
| | - Uly Sumarni
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Chalid Assaf
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
- Clinic for Dermatology and Venereology, Helios Klinikum Krefeld, Lutherplatz 40, 47805 Krefeld, Germany.
| | - Peter Langer
- Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 3a, 18059 Rostock, Germany.
- Leibniz Institute of Catalysis at the University of Rostock e.V., Albert-Einstein-Str. 29a, 18059 Rostock, Germany.
| | - Ulrich Reidel
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jürgen Eberle
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
11
|
Banti CN, Hatzidimitriou AG, Kourkoumelis N, Hadjikakou SK. Diclofenac conjugates with biocides through silver(I) ions (CoMeD's); Development of a reliable model for the prediction of anti-proliferation of NSAID's-silver formulations. J Inorg Biochem 2019; 194:7-18. [PMID: 30798079 DOI: 10.1016/j.jinorgbio.2019.01.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 01/13/2023]
Abstract
The conjugation of diclofenac (DICLH), a Non-Steroidal Anti-inflammatory Drug (NSAID), with biocides such as dimethyl sulfoxide (DMSO) and triphenylphosphine (TPP), through silver(I) ions, results into the chemical [Agn(DICL)n(L)m]k (L = DMSO and n = 2, m = 2, k = infinite (1); L = TPP and n = 1, m = 2, k = 1 (2)). The compounds were characterized by m.p., FT-IR, UV-vis and 1H NMR spectroscopic techniques. The crystal and molecular structures of 1-2 were determined by X-ray crystallography. The in vitro cytotoxic activity of 1-2 against the human breast adenocarcinoma cancer cells MCF-7 (hormone dependent) and MDA-MB-231 (hormone independent) reveals that the 1 inhibits the MCF-7 rather than the MDA-MB-231 cells, suggesting hormone mimetic behaviour. Compound 2 inhibits both cancerous cell lines, stronger than cisplatin. Both compounds inhibit MCF-7 cells migration. Compounds 1-2, exhibit, lower toxicity against fetal lung fibroblast (MRC-5) cells than cisplatin. Their genotoxicity was evaluated on MRC-5 cells. The molecular mechanism of 1-2 against MCF-7 cells was clarified by (i) their cell cycle arrest study (ii) their mitochondrial membrane permeability (iii) their binding affinity towards Calf Thymus (CT)-DNA and (iv) their inhibitory activity against the enzyme lipoxygenase (LOX). Regression analysis of the data obtained for [Ag(NSAID)(Ar3P)m] (NSAID = p‑hydroxy‑benzoic acid (p-HO-BZAH), salicylic acid (SALH2), aspirin (ASPH), naproxen (NAPRH), nimesulide (NIMH); L = TPP, Tri(p‑tolyl)phosphine (TPTP), Tri(o‑tolyl)phosphine (TOTP), Tri(m‑tolyl)phosphine (TMTP); m = 2 or 3) and [Ag(DICL)2(DMSO)2]k (k = infinite) was performed. Considering the biological results (IC50) as dependent variable a theoretical equation is obtained for these compounds. The calculated IC50 values are compared satisfactorily with the corresponding experimental inhibitory activity of the complexes.
Collapse
Affiliation(s)
- Christina N Banti
- Inorganic Chemistry Laboratory, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece.
| | | | | | - Sotiris K Hadjikakou
- Inorganic Chemistry Laboratory, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
12
|
Lakshman TR, Deb J, Ghosh I, Sarkar S, Paine TK. Combining anti-inflammatory and anti-proliferative activities in ternary metal-NSAID complexes of a polypyridylamine ligand. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2018.11.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
13
|
Menacher G, Steinritz D, Schmidt A, Popp T, Worek F, Gudermann T, Thiermann H, Balszuweit F. Effects of anti-inflammatory compounds on sulfur mustard injured cells: Recommendations and caveats suggested by in vitro cell culture models. Toxicol Lett 2018; 293:91-97. [DOI: 10.1016/j.toxlet.2017.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/07/2017] [Accepted: 09/11/2017] [Indexed: 11/27/2022]
|
14
|
Metformin and epothilone A treatment up regulate pro-apoptotic PARP-1, Casp-3 and H2AX genes and decrease of AKT kinase level to control cell death of human hepatocellular carcinoma and ovary adenocarcinoma cells. Toxicol In Vitro 2017; 47:48-62. [PMID: 29117515 DOI: 10.1016/j.tiv.2017.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/20/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023]
Abstract
High mortality rates in ovarian and liver cancer are largely a result of resistance to currently used chemotherapy. Here, we investigated genotoxic and pro-oxidant effects of metformin (MET) and epothilone A (A) in combination with respect to apoptosis in HepG2 and SKOV-3 cancer cells. Reactive oxygen species (ROS) was studied using 2',7'-dichlorodihydrofluoresein diacetate, and samples were analyzed for the presence and absence of the N-acetylcysteine (NAC). Expression of genes involved in programmed cell death, oxidative and alkylating DNA damage was measured. Probes were analyzed in the presence of Akt or nuclear factor-κB inhibitor. Compared to either drug alone, combination of epothilone A and metformin was more potent; decreased Akt level; and elevated percentage of apoptotic cells, induced cell cycle arrest at G1 phase and elevated the sub-G1 cell population by increasing the mRNA level of caspase-3, poly (ADP-ribose) polymerase-1 and H2AX. The anticancer effect of the drug combination was partially reversed by NAC supplementation, suggesting that ROS generation is required to induce apoptosis. The present study demonstrates that novel combination such as epothilone A and MET show promise in expanding ovarian and liver cancer therapy.
Collapse
|
15
|
Fu W, Yi S, Qiu L, Sun J, Tu P, Wang Y. BCL11B-Mediated Epigenetic Repression Is a Crucial Target for Histone Deacetylase Inhibitors in Cutaneous T-Cell Lymphoma. J Invest Dermatol 2017; 137:1523-1532. [PMID: 28288848 DOI: 10.1016/j.jid.2017.02.980] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/13/2017] [Accepted: 02/24/2017] [Indexed: 10/20/2022]
Abstract
The treatment options for advanced cutaneous T-cell lymphoma (CTCL) are limited because of its unclear pathogenesis. Histone deacetylase (HDAC) inhibitors (HDACis) are recently developed therapeutics approved for refractory CTCL. However, the response rate is relatively low and unpredictable. Previously, we discovered that BCL11B, a key T-cell development regulator, was aberrantly overexpressed in mycosis fungoides, the most common CTCL, as compared with benign inflammatory skin. In this study, we identified a positive correlation between BCL11B expression and sensitivity to HDACi in CTCL lines. BCL11B suppression in BCL11B-high cells induced cell apoptosis by de-repressing apoptotic pathways and showed synergistic effects with suberoylanilide hydroxamic acid (SAHA), a pan-HDACi. Next, we identified the physical interaction and shared downstream genes between BCL11B and HDAC1/2 in CTCL lines. This interaction was essential in the anti-apoptosis effect of BCL11B, and the synergism between BCL11B suppression and HDACi treatment. Further, in clinical samples from 46 mycosis fungoides patients, BCL11B showed increased but varied expression in advanced tumor stage. Analysis of four patients receiving SAHA treatment suggested a positive correlation between BCL11B expression and favorable response to SAHA treatment. In conclusion, BCL11B may serve as a therapeutic target and a useful marker for improving HDACi efficacy in advanced CTCL.
Collapse
Affiliation(s)
- Wenjing Fu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Department of Dermatology and Venerology, Binzhou Medical University Hospital, Binzhou, China
| | - Shengguo Yi
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Lei Qiu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Jingru Sun
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Ping Tu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Yang Wang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
16
|
Targeting Thioredoxin-1 by dimethyl fumarate induces ripoptosome-mediated cell death. Sci Rep 2017; 7:43168. [PMID: 28233787 PMCID: PMC5324128 DOI: 10.1038/srep43168] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/20/2017] [Indexed: 01/11/2023] Open
Abstract
Constitutively active NFκB promotes survival of many cancers, especially T-cell lymphomas and leukemias by upregulating antiapoptotic proteins such as inhibitors of apoptosis (IAPs) and FLICE-like inhibitory proteins (cFLIPs). IAPs and cFLIPs negatively regulate the ripoptosome, which mediates cell death in an apoptotic or necroptotic manner. Here, we demonstrate for the first time, that DMF antagonizes NFκB by suppressing Thioredoxin-1 (Trx1), a major regulator of NFκB transcriptional activity. DMF-mediated inhibition of NFκB causes ripoptosome formation via downregulation of IAPs and cFLIPs. In addition, DMF promotes mitochondrial Smac release and subsequent degradation of IAPs, further enhancing cell death in tumor cells displaying constitutive NFκB activity. Significantly, CTCL patients treated with DMF display substantial ripoptosome formation and caspase-3 cleavage in T-cells. DMF induces cell death predominantly in malignant or activated T-cells. Further, we show that malignant T-cells can die by both apoptosis and necroptosis, in contrast to resting T-cells, which are restricted to apoptosis upon DMF administration. In summary, our data provide new mechanistic insight in the regulation of cell death by targeting NFκB via Trx1 in cancer. Thus, interference with Trx1 activity is a novel approach for treatment of NFκB-dependent tumors.
Collapse
|
17
|
Hassan HM, Varney ML, Chaturvedi NK, Joshi SS, Weisenburger DD, Singh RK, Dave BJ. Modulation of p73 isoforms expression induces anti-proliferative and pro-apoptotic activity in mantle cell lymphoma independent of p53 status. Leuk Lymphoma 2016; 57:2874-2889. [PMID: 27074052 PMCID: PMC5967247 DOI: 10.3109/10428194.2016.1165814] [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] [Indexed: 12/13/2022]
Abstract
Mantle cell lymphoma (MCL) is characterized by a clinically aggressive course with frequent relapse and poor survival. The p53 pathway is frequently dysregulated and p53 status predicts clinical outcome. In this report, we investigated whether modulation of p73 isoforms by diclofenac inhibits cell growth, induces apoptosis and/or cell cycle arrest in MCL relative to p53 status. Wild-type p53 [Granta-519 and JVM-2], mutant p53 [Jeko-1 and Mino-1] expressing cells, therapy resistant cell lines, and primary human cells isolated from MCL patients were used. Overexpression of pro-apoptotic TAp73 enhanced MCL cell apoptosis. Diclofenac induced a concentration- and duration-dependent increase in TAp73, cell cycle arrest, cell death, and inhibited MCL cell growth independent of p53 status. Diclofenac treatment was associated with increased activity of caspases 3, 7, and 8 and induction of p53 transcriptional target genes. These studies demonstrate the potential for diclofenac as novel therapeutic agent in MCL independent of p53 status.
Collapse
Affiliation(s)
- Hesham M. Hassan
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michelle L. Varney
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagendra K. Chaturvedi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shantaram S. Joshi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Rakesh K. Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhavana J. Dave
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Human Genetics Laboratories, Munroe Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
18
|
Dimethyl fumarate restores apoptosis sensitivity and inhibits tumor growth and metastasis in CTCL by targeting NF-κB. Blood 2016; 128:805-15. [PMID: 27268084 DOI: 10.1182/blood-2016-01-694117] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/26/2016] [Indexed: 12/19/2022] Open
Abstract
Despite intensive efforts in recent years, a curative therapy for cutaneous T-cell lymphoma (CTCL) has not yet been developed. Therefore, the establishment of new therapeutic approaches with higher efficacy rates and milder side effects is strongly desired. A characteristic feature of the malignant T-cell population in CTCL is resistance toward cell death resulting from constitutive NF-κB activation. Therefore, NF-κB-dependent cell death resistance represents an interesting therapeutic target in CTCL because an NF-κB-directed therapy would leave bystander T cells widely unaffected. We investigated the effects of dimethyl fumarate (DMF) on CTCL cells in vitro and in vivo. DMF induced cell death in primary patient-derived CD4(+) cells and CTCL cell lines, but hardly in T cells from healthy donors. DMF-induced cell death was linked specifically to NF-κB inhibition. To study the impact of DMF in vivo, we developed 2 CTCL xenograft mouse models with different cutaneous localizations of the T-cell infiltrate. DMF treatment delayed the growth of CTCL tumors and prevented formation of distant metastases. In addition, DMF induced increased cell death in primary CTCL tumors and in liver metastases. In summary, DMF treatment represents a remarkable therapeutic option in CTCL because it restores CTCL apoptosis in vitro and in preclinical models in vivo and prevents spreading of the disease to distant sites. DMF treatment is of particular promise in CTCL because DMF is already in successful clinical use in the treatment of psoriasis and multiple sclerosis allowing fast translation into clinical studies in CTCL.
Collapse
|
19
|
Bi XW, Wang L, Zhang WW, Yan SM, Sun P, Xia Y, Li ZM, Jiang WQ. The pretreatment albumin to globulin ratio predicts survival in patients with natural killer/T-cell lymphoma. PeerJ 2016; 4:e1742. [PMID: 26966671 PMCID: PMC4782740 DOI: 10.7717/peerj.1742] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/07/2016] [Indexed: 12/19/2022] Open
Abstract
Background. The pretreatment albumin to globulin ratio (AGR) has been reported to be a predictor of survival in several types of cancer. The aim of this study was to evaluate the prognostic impact of AGR in patients with natural killer/T-cell lymphoma (NKTCL). Methods. We retrospectively reviewed the available serum biochemistry results for 331 NKTCL patients before treatment. AGR was calculated as albumin/(total protein-albumin), and a cut-off value of 1.3 was used to define AGR as low or high. Survival analysis was used to assess the prognostic value of AGR. Results. A low AGR (<1.3) was associated with significantly more adverse clinical features, including old age, poor performance status, advanced stage, elevated lactate dehydrogenase, B symptoms, and high International Prognostic Index (IPI) and natural killer/T-cell lymphoma prognostic index (NKPI) scores. Patients with a low AGR had a significantly lower 5-year overall survival (44.5 vs. 65.2%, P < 0.001) and progression-free survival (33.1 vs. 57.4%, P < 0.001). In the multivariate analysis, a low AGR remained an independent predictor of poorer survival. Additionally, AGR distinguished patients with different outcomes in the IPI low-risk group and in the NKPI high-risk group. Discussion. Pretreatment AGR may serve as a simple and effective predictor of prognosis in patients with NKTCL.
Collapse
Affiliation(s)
- Xi-wen Bi
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Liang Wang
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Wen-wen Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Shu-mei Yan
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Peng Sun
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yi Xia
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Zhi-ming Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Wen-qi Jiang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, PR China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| |
Collapse
|
20
|
Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-diclofenac as an anti-cancer agent. Ecancermedicalscience 2016; 10:610. [PMID: 26823679 PMCID: PMC4720497 DOI: 10.3332/ecancer.2016.610] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Diclofenac (DCF) is a well-known and widely used non-steroidal anti-inflammatory drug (NSAID), with a range of actions which are of interest in an oncological context. While there has long been an interest in the use of NSAIDs in chemoprevention, there is now emerging evidence that such drugs may have activity in a treatment setting. DCF, which is a potent inhibitor of COX-2 and prostaglandin E2 synthesis, displays a range of effects on the immune system, the angiogenic cascade, chemo- and radio-sensitivity and tumour metabolism. Both pre-clinical and clinical evidence of these effects, in multiple cancer types, is assessed and summarised and relevant mechanisms of action outlined. Based on this evidence the case is made for further clinical investigation of the anticancer effects of DCF, particularly in combination with other agents - with a range of possible multi-drug and multi-modality combinations outlined in the supplementary materials accompanying the main paper.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | | | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc; Newton MA 02459, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
21
|
Wu J, Salva KA, Wood GS. c-CBL E3 ubiquitin ligase is overexpressed in cutaneous T-cell lymphoma: its inhibition promotes activation-induced cell death. J Invest Dermatol 2014; 135:861-868. [PMID: 25140833 PMCID: PMC4324119 DOI: 10.1038/jid.2014.364] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/14/2014] [Accepted: 07/28/2014] [Indexed: 11/17/2022]
Abstract
Mycosis fungoides (MF) and Sezary syndrome (SS) are two major forms of cutaneous T-cell lymphoma (CTCL) characterized by resistance to apoptosis. A central pathway for T-cell apoptosis is activation-induced cell death (AICD) which is triggered through the T-cell receptor (TCR). This results in upregulation of FAS-ligand (FASL) and subsequent apoptosis through the FAS death receptor pathway. It has been known for more than a decade that TCR signaling is defective in CTCL; however, the underlying mechanism has not been apparent. In this report, we show that the E3 ubiquitin ligase, c-CBL, is over-expressed in CTCL and that its knockdown overcomes defective TCR signaling resulting in phosphorylation of PLCg1, calcium influx, ROS generation, up-regulation of FASL and extrinsic pathway apoptosis in CTCL cells expressing adequate FAS. In CTCL cells with suboptimal FAS expression, FAS can be upregulated epigenetically by derepression of the FAS promoter using methotrexate (MTX) which we showed previously has activity as a DNA methylation inhibitor. Using these combined strategies, FAS-low as well as FAS-high CTCL cells can be killed effectively.
Collapse
Affiliation(s)
- Jianqiang Wu
- Department of Dermatology, University of Wisconsin and the Middleton VA Medical Center, Madison, Wisconsin, USA
| | - Katrin A Salva
- Department of Dermatology, University of Wisconsin and the Middleton VA Medical Center, Madison, Wisconsin, USA
| | - Gary S Wood
- Department of Dermatology, University of Wisconsin and the Middleton VA Medical Center, Madison, Wisconsin, USA.
| |
Collapse
|
22
|
Indomethacin Sensitizes TRAIL-Resistant Melanoma Cells to TRAIL-Induced Apoptosis through ROS-Mediated Upregulation of Death Receptor 5 and Downregulation of Survivin. J Invest Dermatol 2014; 134:1397-1407. [DOI: 10.1038/jid.2013.471] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/24/2013] [Accepted: 10/13/2013] [Indexed: 02/07/2023]
|
23
|
Willerslev-Olsen A, Litvinov IV, Fredholm SM, Petersen DL, Sibbesen NA, Gniadecki R, Zhang Q, Bonefeld CM, Wasik MA, Geisler C, Zhou Y, Woetmann A, Sasseville D, Krejsgaard T, Odum N. IL-15 and IL-17F are differentially regulated and expressed in mycosis fungoides (MF). Cell Cycle 2014; 13:1306-12. [PMID: 24621498 DOI: 10.4161/cc.28256] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Skin lesions from mycosis fungoides (MF) patients display an increased expression of interleukin-15 (IL-15), IL-17F, and other cytokines implicated in inflammation and malignant cell proliferation in cutaneous T-cell lymphoma (CTCL). In the leukemic variant of CTCL, Sézary syndrome (SS), IL-2 and IL-15 trigger activation of the Jak-3/STAT3 pathway and transcription of IL17A gene, whereas it is unknown what causes IL-15 expression, Jak3/STAT3 activation, and production of IL-17F in MF. Here, we studied the expression and regulation of IL-15 and its relation to IL-17F in MF cell lines and skin lesions from 60 MF patients. We show that: (1) the spontaneous IL-15 mRNA expression is resistant to Jak3 and STAT3 inhibitors at concentrations that profoundly inhibit STAT3 activation and IL-17F mRNA expression; (2) anti-IL-15 antibody blocks STAT3 activation induced by exogenous IL-15 in non-malignant MF T cells, whereas the spontaneous STAT3 activation and IL-17F expression in malignant T cells is not inhibited; (3) patients display heterogeneous IL-15/IL-17F mRNA expression patterns in skin lesions; and (4) IL-15 expression (in contrast to IL-17F) is not associated with progressive disease. Taken together, these findings indicate that IL-15 and IL-17F are differentially regulated and expressed in MF. We propose that IL-15 and IL-17F are markers for different inflammatory environments and play distinct roles in the development and progression of MF.
Collapse
Affiliation(s)
- Andreas Willerslev-Olsen
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Ivan V Litvinov
- Division of Dermatology; McGill University Health Centre; Montréal, Quebec, Canada
| | - Simon M Fredholm
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - David L Petersen
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Nina A Sibbesen
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Robert Gniadecki
- Departmen of Dermatology; Copenhagen University Hospital; Bispebjerg, Copenhagen, Denmark
| | - Qian Zhang
- Department of Pathology and Laboratory Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Charlotte M Bonefeld
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Mariusz A Wasik
- Department of Pathology and Laboratory Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Carsten Geisler
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Youwen Zhou
- Department of Dermatology and Skin Science; University of British Columbia; Vancouver, British Columbia, Canada
| | - Anders Woetmann
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Denis Sasseville
- Division of Dermatology; McGill University Health Centre; Montréal, Quebec, Canada
| | - Thorbjørn Krejsgaard
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Niels Odum
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| |
Collapse
|
24
|
Gupta V, Liu S, Ando H, Ishii R, Tateno S, Kaneko Y, Yugami M, Sakamoto S, Yamaguchi Y, Nureki O, Handa H. Salicylic Acid Induces Mitochondrial Injury by Inhibiting Ferrochelatase Heme Biosynthesis Activity. Mol Pharmacol 2013; 84:824-33. [DOI: 10.1124/mol.113.087940] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
25
|
TODO MOMOKO, HORINAKA MANO, TOMOSUGI MITSUHIRO, TANAKA RYOICHI, IKAWA HARUNA, SOWA YOSHIHIRO, ISHIKAWA HIDEKI, FUJIWARA HITOSHI, OTSUJI EIGO, SAKAI TOSHIYUKI. Ibuprofen enhances TRAIL-induced apoptosis through DR5 upregulation. Oncol Rep 2013; 30:2379-84. [DOI: 10.3892/or.2013.2713] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 07/29/2013] [Indexed: 11/05/2022] Open
|
26
|
Gottfried E, Lang SA, Renner K, Bosserhoff A, Gronwald W, Rehli M, Einhell S, Gedig I, Singer K, Seilbeck A, Mackensen A, Grauer O, Hau P, Dettmer K, Andreesen R, Oefner PJ, Kreutz M. New aspects of an old drug--diclofenac targets MYC and glucose metabolism in tumor cells. PLoS One 2013; 8:e66987. [PMID: 23874405 PMCID: PMC3706586 DOI: 10.1371/journal.pone.0066987] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 05/10/2013] [Indexed: 12/21/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs such as diclofenac exhibit potent anticancer effects. Up to now these effects were mainly attributed to its classical role as COX-inhibitor. Here we show novel COX-independent effects of diclofenac. Diclofenac significantly diminished MYC expression and modulated glucose metabolism resulting in impaired melanoma, leukemia, and carcinoma cell line proliferation in vitro and reduced melanoma growth in vivo. In contrast, the non-selective COX inhibitor aspirin and the COX-2 specific inhibitor NS-398 had no effect on MYC expression and glucose metabolism. Diclofenac significantly decreased glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA), and monocarboxylate transporter 1 (MCT1) gene expression in line with a decrease in glucose uptake and lactate secretion. A significant intracellular accumulation of lactate by diclofenac preceded the observed effect on gene expression, suggesting a direct inhibitory effect of diclofenac on lactate efflux. While intracellular lactate accumulation impairs cellular proliferation and gene expression, it does not inhibit MYC expression as evidenced by the lack of MYC regulation by the MCT inhibitor α-cyano-4-hydroxycinnamic acid. Finally, in a cell line with a tetracycline-regulated c-MYC gene, diclofenac decreased proliferation both in the presence and absence of c-MYC. Thus, diclofenac targets tumor cell proliferation via two mechanisms, that is inhibition of MYC and lactate transport. Based on these results, diclofenac holds potential as a clinically applicable MYC and glycolysis inhibitor supporting established tumor therapies.
Collapse
Affiliation(s)
- Eva Gottfried
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
- Regensburg Centre for Interventional Immunology (RCI), University of Regensburg, Regensburg, Germany
| | - Sven A. Lang
- Department of Surgery, University of Regensburg, Regensburg, Germany
| | - Kathrin Renner
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
- Regensburg Centre for Interventional Immunology (RCI), University of Regensburg, Regensburg, Germany
| | - Anja Bosserhoff
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Wolfram Gronwald
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Michael Rehli
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
- Regensburg Centre for Interventional Immunology (RCI), University of Regensburg, Regensburg, Germany
| | - Sabine Einhell
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| | - Isabel Gedig
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| | - Katrin Singer
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| | - Anton Seilbeck
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Oliver Grauer
- Department of Neurology, University of Muenster, Muenster, Germany
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Peter Hau
- Department of Neurology, University of Regensburg, Regensburg, Germany
- Wilhelm Sander NeuroOncology Unit, University of Regensburg, Regensburg, Germany
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Reinhard Andreesen
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
- Regensburg Centre for Interventional Immunology (RCI), University of Regensburg, Regensburg, Germany
| | - Peter J. Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Marina Kreutz
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
- Regensburg Centre for Interventional Immunology (RCI), University of Regensburg, Regensburg, Germany
| |
Collapse
|
27
|
Wise JF, Berkova Z, Mathur R, Zhu H, Braun FK, Tao RH, Sabichi AL, Ao X, Maeng H, Samaniego F. Nucleolin inhibits Fas ligand binding and suppresses Fas-mediated apoptosis in vivo via a surface nucleolin-Fas complex. Blood 2013; 121:4729-39. [PMID: 23599269 PMCID: PMC3674671 DOI: 10.1182/blood-2012-12-471094] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 04/10/2013] [Indexed: 12/16/2022] Open
Abstract
Resistance to Fas-mediated apoptosis is associated with poor cancer outcomes and chemoresistance. To elucidate potential mechanisms of defective Fas signaling, we screened primary lymphoma cell extracts for Fas-associated proteins that would have the potential to regulate Fas signaling. An activation-resistant Fas complex selectively included nucleolin. We confirmed the presence of nucleolin-Fas complexes in B-cell lymphoma cells and primary tissues, and the absence of such complexes in B-lymphocytes from healthy donors. RNA-binding domain 4 and the glycine/arginine-rich domain of nucleolin were essential for its association with Fas. Nucleolin colocalized with Fas on the surface of B-cell lymphoma cells. Nucleolin knockdown sensitized BJAB cells to Fas ligand (FasL)-induced and Fas agonistic antibody-induced apoptosis through enhanced binding, suggesting that nucleolin blocks the FasL-Fas interaction. Mice transfected with nucleolin were protected from the lethal effects of agonistic anti-mouse Fas antibody (Jo2) and had lower rates of hepatocyte apoptosis, compared with vector and a non-Fas-binding mutant of nucleolin. Our results show that cell surface nucleolin binds Fas, inhibits ligand binding, and thus prevents induction of Fas-mediated apoptosis in B-cell lymphomas and may serve as a new therapeutic target.
Collapse
Affiliation(s)
- Jillian F Wise
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gong J, Kumar SA, Graham G, Kumar AP. FLIP: molecular switch between apoptosis and necroptosis. Mol Carcinog 2013; 53:675-85. [PMID: 23625539 DOI: 10.1002/mc.22027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 02/13/2013] [Accepted: 02/25/2013] [Indexed: 12/13/2022]
Abstract
Cancerous growth is one of the most difficult diseases to target as there is no one clear cause, and targeting only one pathway does not generally produce quantifiable improvement. For a truly effective cancer therapy, multiple pathways must be targeted at the same time. One way to do this is to find a gene that is associated with several pathways; this approach expands the possibilities for disease targeting and enables multiple points of attack rather than one fixed point, which does not allow treatment to evolve over time as cancer does. Inducing programmed cell death (PCD) is a promising method to prevent or inhibit the progression of tumor cells. Intricate cross talk among various programmed cell death pathways including cell death by apoptosis, necroptosis or autophagy plays a critical role in the regulation of PCD. In addition, the complex and overlapping patterns of signaling and lack of understanding of such networks between these pathways generate hurdles for developing effective therapeutic approaches. This review article focuses on targeting FLIP (Fas-associated death domain-like interleukin-1-converting enzyme-like inhibitory protein) signaling as a bridge between various PCD processes as an effective approach for cancer management.
Collapse
Affiliation(s)
- JingJing Gong
- Department of Urology, School of Medicine, South Texas Veterans Health Affairs System, The University of Texas Health Science Center, San Antonio, Texas
| | | | | | | |
Collapse
|
29
|
Salicylate Selectively Kills Cochlear Spiral Ganglion Neurons by Paradoxically Up-regulating Superoxide. Neurotox Res 2013; 24:307-19. [DOI: 10.1007/s12640-013-9384-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 12/12/2022]
|
30
|
Berger A, Quast SA, Plötz M, Kammermeier A, Eberle J. Sensitization of melanoma cells for TRAIL-induced apoptosis by BMS-345541 correlates with altered phosphorylation and activation of Bax. Cell Death Dis 2013; 4:e477. [PMID: 23348591 PMCID: PMC3563986 DOI: 10.1038/cddis.2012.198] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 11/27/2012] [Accepted: 11/28/2012] [Indexed: 12/12/2022]
Abstract
Resistance to TRAIL (TNF-related apoptosis-inducing ligand)- induced apoptosis limits its therapeutic use. Different strategies of TRAIL sensitization and a dependency on Bax have been reported, but common principles of TRAIL resistance and the way of Bax activation remained poorly understood. Applying a melanoma model of TRAIL-sensitive and -resistant cell lines, efficient sensitization for TRAIL-induced apoptosis is demonstrated by the kinase inhibitor BMS-345541 (N-(1,8-dimethylimidazo(1,2-a)quinoxalin-4-yl)-1,2-ethanediamine hydrochloride), which targets IκB (inhibitor of κB proteins) kinase β (IKKβ). This effect was completely abrogated by Bax knockout as well as by Bcl-2 overexpression, in accordance with a Bax dependency. Early loss of the mitochondrial membrane potential, release of cytochrome c and Smac (second mitochondria-derived activator of caspases) clearly indicated the activation of mitochondrial apoptosis pathways. Of note, BMS-345541 alone resulted in an early Bax activation, seen by conformational changes and by Bax translocation. The synergistic effects can be explained by Bid activation through TRAIL, which inhibits Bcl-2, and the activation of Bax through BMS-345541. The critical roles of XIAP (X-chromosome-linked inhibitor of apoptosis protein), Smac and Bid were clearly proven by overexpression and siRNA knockdown, respectively. The way of Bax activation by BMS-345541 was unraveled by establishing new assays for Bax activation. These showed reduction of the inactivating Bax phosphorylation at serine-184, while the activating Bax phosphorylation at threonine-167 was enhanced. Thus, modulation of Bax phosphorylation appeared as tightly related to TRAIL sensitivity/resistance in melanoma cells, and therapeutic strategies may be considered.
Collapse
Affiliation(s)
- A Berger
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - S-A Quast
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - M Plötz
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - A Kammermeier
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - J Eberle
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| |
Collapse
|
31
|
Rodust PM, Fecker LF, Stockfleth E, Eberle J. Activation of mitochondrial apoptosis pathways in cutaneous squamous cell carcinoma cells by diclofenac/hyaluronic acid is related to upregulation of Bad as well as downregulation of Mcl-1 and Bcl-w. Exp Dermatol 2012; 21:520-5. [PMID: 22716247 DOI: 10.1111/j.1600-0625.2012.01516.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Actinic keratosis (AK) is characterized by high prevalence and the risk to proceed to squamous cell carcinoma (SCC). Cyclooxygenase-2 (COX-2)-mediated prostaglandin E2 (PGE (2) ) synthesis has been reported in AK and SCC, and the COX inhibitor diclofenac in hyaluronic acid (diclofenac/HA) was approved for AK therapy. Its mode of action, however, remained to be unravelled. In the present study, diclofenac resulted in reduced PGE (2) levels in apoptosis-sensitive cutaneous SCC cell lines (SCL-II, SCC-12, SCC-13) whereas no PGE (2) and no COX-2 expression was detectable in a SCC cell line resistant to apoptosis induction (SCL-I). Activation of mitochondrial apoptosis pathways was evident in SCC cells owing to loss of the mitochondrial membrane potential and release of the mitochondrial factors cytochrome c and apoptosis-inducing factor. Characteristic proapoptotic changes at the level of Bcl-2 proteins occurred in sensitive cells, as upregulation of Bad and downregulation of Mcl-1 and Bcl-w. In contrast, Bad was already high, and Mcl-1 and Bcl-w were already low in resistant SCL-I, even without treatment, which may be explained by the lack of PGE (2) . An antiapoptotic downregulation of proapoptotic Bcl-2 proteins Noxa and Puma was, however, also seen in SCL-I, suggesting here pathways independent of COX-2. The regulations of Mcl-1 and Bad were also reproduced in SCC cells by the more selective COX-2 inhibitor celecoxib, thus further underlining the specific role of COX-2. The findings illuminate the mode of action of diclofenac/HA in SCC cells as well as principles of their resistance, which may allow further adaptation and improvement of the new therapy.
Collapse
Affiliation(s)
- Paul M Rodust
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | | | | | | |
Collapse
|
32
|
Al-Yacoub N, Fecker LF, Möbs M, Plötz M, Braun FK, Sterry W, Eberle J. Apoptosis induction by SAHA in cutaneous T-cell lymphoma cells is related to downregulation of c-FLIP and enhanced TRAIL signaling. J Invest Dermatol 2012; 132:2263-74. [PMID: 22551975 DOI: 10.1038/jid.2012.125] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Suberoylanilide hydroxamic acid (SAHA) has been approved for the treatment of cutaneous T-cell lymphoma (CTCL), but its mode of action remained largely elusive. As shown here in four CTCL cell lines, loss of cell viability correlated with significant time- and dose-dependent induction of apoptosis, whereas cytotoxicity was less pronounced. Both extrinsic and intrinsic apoptosis pathways were activated, as seen by processing of initiator caspases 8 and 9, loss of mitochondrial membrane potential, and cytochrome c release. Characteristically, antiapoptotic mediators such as Mcl-1, XIAP, survivin, and c-FLIP were downregulated. Consistent with its critical function, c-FLIP overexpression resulted in a significant decrease of SAHA-mediated apoptosis. Enhanced sensitivity to TRAIL (TNF-related apoptosis-inducing ligand) and enhanced TRAIL signaling was seen in CTCL cell lines with high sensitivity, whereas cell lines with moderate response were characterized by downregulation of TRAIL-R2 and weaker TRAIL expression. Comparable proapoptotic responses to SAHA and to the combination with TRAIL were seen in ex vivo tumor T cells of CTCL patients. Thus, activation of extrinsic apoptosis pathways, related to c-FLIP downregulation and enhanced TRAIL signaling, appeared as characteristic for CTCL cell responsiveness to SAHA. An improved understanding of the pathways may facilitate its targeted use and the selection of suitable combinations.
Collapse
Affiliation(s)
- Nadya Al-Yacoub
- Department of Dermatology and Allergy, Skin Cancer Center Charité (HTCC), Charité-University Medical Center Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|