1
|
Bruserud Ø, Selheim F, Hernandez-Valladares M, Reikvam H. Monocytic Differentiation in Acute Myeloid Leukemia Cells: Diagnostic Criteria, Biological Heterogeneity, Mitochondrial Metabolism, Resistance to and Induction by Targeted Therapies. Int J Mol Sci 2024; 25:6356. [PMID: 38928061 PMCID: PMC11203697 DOI: 10.3390/ijms25126356] [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: 05/05/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
We review the importance of monocytic differentiation and differentiation induction in non-APL (acute promyelocytic leukemia) variants of acute myeloid leukemia (AML), a malignancy characterized by proliferation of immature myeloid cells. Even though the cellular differentiation block is a fundamental characteristic, the AML cells can show limited signs of differentiation. According to the French-American-British (FAB-M4/M5 subset) and the World Health Organization (WHO) 2016 classifications, monocytic differentiation is characterized by morphological signs and the expression of specific molecular markers involved in cellular communication and adhesion. Furthermore, monocytic FAB-M4/M5 patients are heterogeneous with regards to cytogenetic and molecular genetic abnormalities, and monocytic differentiation does not have any major prognostic impact for these patients when receiving conventional intensive cytotoxic therapy. In contrast, FAB-M4/M5 patients have decreased susceptibility to the Bcl-2 inhibitor venetoclax, and this seems to be due to common molecular characteristics involving mitochondrial regulation of the cellular metabolism and survival, including decreased dependency on Bcl-2 compared to other AML patients. Thus, the susceptibility to Bcl-2 inhibition does not only depend on general resistance/susceptibility mechanisms known from conventional AML therapy but also specific mechanisms involving the molecular target itself or the molecular context of the target. AML cell differentiation status is also associated with susceptibility to other targeted therapies (e.g., CDK2/4/6 and bromodomain inhibition), and differentiation induction seems to be a part of the antileukemic effect for several targeted anti-AML therapies. Differentiation-associated molecular mechanisms may thus become important in the future implementation of targeted therapies in human AML.
Collapse
MESH Headings
- Humans
- Cell Differentiation
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Mitochondria/metabolism
- Monocytes/metabolism
- Monocytes/pathology
- Drug Resistance, Neoplasm/genetics
- Molecular Targeted Therapy
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
Collapse
Affiliation(s)
- Øystein Bruserud
- Acute Leukemia Research Group, Department of Clinical Science, University of Bergen, 5007 Bergen, Norway; (M.H.-V.); (H.R.)
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5009 Bergen, Norway
| | - Frode Selheim
- Proteomics Unit of University of Bergen (PROBE), University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway;
| | - Maria Hernandez-Valladares
- Acute Leukemia Research Group, Department of Clinical Science, University of Bergen, 5007 Bergen, Norway; (M.H.-V.); (H.R.)
- Department of Physical Chemistry, University of Granada, Avenida de la Fuente Nueva S/N, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Håkon Reikvam
- Acute Leukemia Research Group, Department of Clinical Science, University of Bergen, 5007 Bergen, Norway; (M.H.-V.); (H.R.)
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5009 Bergen, Norway
| |
Collapse
|
2
|
Prabhu KS, Kuttikrishnan S, Ahmad N, Habeeba U, Mariyam Z, Suleman M, Bhat AA, Uddin S. H2AX: A key player in DNA damage response and a promising target for cancer therapy. Biomed Pharmacother 2024; 175:116663. [PMID: 38688170 DOI: 10.1016/j.biopha.2024.116663] [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: 02/14/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
Cancer is caused by a complex interaction of factors that interrupt the normal growth and division of cells. At the center of this process is the intricate relationship between DNA damage and the cellular mechanisms responsible for maintaining genomic stability. When DNA damage is not repaired, it can cause genetic mutations that contribute to the initiation and progression of cancer. On the other hand, the DNA damage response system, which involves the phosphorylation of the histone variant H2AX (γH2AX), is crucial in preserving genomic integrity by signaling and facilitating the repair of DNA double-strand breaks. This review provides an explanation of the molecular dynamics of H2AX in the context of DNA damage response. It emphasizes the crucial role of H2AX in recruiting and localizing repair machinery at sites of chromatin damage. The review explains how H2AX phosphorylation, facilitated by the master kinases ATM and ATR, acts as a signal for DNA damage, triggering downstream pathways that govern cell cycle checkpoints, apoptosis, and the cellular fate decision between repair and cell death. The phosphorylation of H2AX is a critical regulatory point, ensuring cell survival by promoting repair or steering cells towards apoptosis in cases of catastrophic genomic damage. Moreover, we explore the therapeutic potential of targeting H2AX in cancer treatment, leveraging its dual function as a biomarker of DNA integrity and a therapeutic target. By delineating the pathways that lead to H2AX phosphorylation and its roles in apoptosis and cell cycle control, we highlight the significance of H2AX as both a prognostic tool and a focal point for therapeutic intervention, offering insights into its utility in enhancing the efficacy of cancer treatments.
Collapse
Affiliation(s)
- Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar.
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Nuha Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Ummu Habeeba
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Zahwa Mariyam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Muhammad Suleman
- Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
3
|
Egbuna C, Patrick‐Iwuanyanwu KC, Onyeike EN, Khan J, Palai S, Patel SB, Parmar VK, Kushwaha G, Singh O, Jeevanandam J, Kumarasamy S, Uche CZ, Narayanan M, Rudrapal M, Odoh U, Chikeokwu I, Găman M, Saravanan K, Ifemeje JC, Ezzat SM, Olisah MC, Chikwendu CJ, Adedokun KA, Imodoye SO, Bello IO, Twinomuhwezi H, Awuchi CG. Phytochemicals and bioactive compounds effective against acute myeloid leukemia: A systematic review. Food Sci Nutr 2023; 11:4191-4210. [PMID: 37457145 PMCID: PMC10345688 DOI: 10.1002/fsn3.3420] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 07/18/2023] Open
Abstract
This systematic review identified various bioactive compounds which have the potential to serve as novel drugs or leads against acute myeloid leukemia. Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy that arises from the dysregulation of cell differentiation, proliferation, and cell death. The risk factors associated with the onset of AML include long-term exposure to radiation and chemicals such as benzene, smoking, genetic disorders, blood disorders, advancement in age, and others. Although novel strategies to manage AML, including a refinement of the conventional chemotherapy regimens, hypomethylating agents, and molecular targeted drugs, have been developed in recent years, resistance and relapse remain the main clinical problems. In this study, three databases, PubMed/MEDLINE, ScienceDirect, and Google Scholar, were systematically searched to identify various bioactive compounds with antileukemic properties. A total of 518 articles were identified, out of which 59 were viewed as eligible for the current report. From the data extracted, over 60 bioactive compounds were identified and divided into five major groups: flavonoids, alkaloids, organosulfur compounds, terpenes, and terpenoids, and other known and emerging bioactive compounds. The mechanism of actions of the analyzed individual bioactive molecules differs remarkably and includes disrupting chromatin structure, upregulating the synthesis of certain DNA repair proteins, inducing cell cycle arrest and apoptosis, and inhibiting/regulating Hsp90 activities, DNA methyltransferase 1, and histone deacetylase 1.
Collapse
Affiliation(s)
- Chukwuebuka Egbuna
- Africa Centre of Excellence for Public Health and Toxicological Research (ACE‐PUTOR)University of Port HarcourtPort HarcourtNigeria
- Department of Biochemistry, Faculty of ScienceUniversity of Port HarcourtPort HarcourtNigeria
- Department of Biochemistry, Faculty of Natural SciencesChukwuemeka Odumegwu Ojukwu UniversityAnambraNigeria
| | - Kingsley C. Patrick‐Iwuanyanwu
- Africa Centre of Excellence for Public Health and Toxicological Research (ACE‐PUTOR)University of Port HarcourtPort HarcourtNigeria
- Department of Biochemistry, Faculty of ScienceUniversity of Port HarcourtPort HarcourtNigeria
| | - Eugene N. Onyeike
- Africa Centre of Excellence for Public Health and Toxicological Research (ACE‐PUTOR)University of Port HarcourtPort HarcourtNigeria
- Department of Biochemistry, Faculty of ScienceUniversity of Port HarcourtPort HarcourtNigeria
| | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAl MajmaahSaudi Arabia
| | - Santwana Palai
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary Science and Animal HusbandryOUATOdishaBhubaneswarIndia
| | - Sandip B. Patel
- Department of PharmacologyL.M. College of Pharmacy, NavrangpuraAhmedabadIndia
| | | | - Garima Kushwaha
- Department of BiotechnologyIndian Institute of TechnologyRoorkeeIndia
| | - Omkar Singh
- Department of Chemical EngineeringIndian Institute of Technology MadrasChennaiIndia
| | - Jaison Jeevanandam
- CQM—Centro de Química da MadeiraUniversidade da Madeira, Campus da PenteadaFunchalPortugal
| | | | - Chukwuemelie Zedech Uche
- Department of Medical Biochemistry and Molecular Biology, Faculty of Basic Medical SciencesUniversity of NigeriaEnuguNsukkaNigeria
| | - Mathiyazhagan Narayanan
- Division of Research and InnovationDepartment of Biotecnology, Saveetha School of Engineering SIMATSTamil NaduChennaiIndia
| | - Mithun Rudrapal
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical SciencesVignan’s Foundation for Science, Technology & ResearchGunturIndia
| | - Uchenna Odoh
- Department of Pharmacognosy and Environmental Medicines, Faculty of Pharmaceutical SciencesUniversity of NigeriaNsukkaNigeria
| | - Ikenna Chikeokwu
- Department of PharmacognosyEnugu State University of Science and Technology (ESUT)Agbani Enugu StateEnuguNigeria
| | - Mihnea‐Alexandru Găman
- Faculty of Medicine"Carol Davila" University of Medicine and PharmacyBucharestRomania
- Department of HematologyCenter of Hematology and Bone Marrow TransplantationBucharestRomania
| | - Kaliyaperumal Saravanan
- PG and Research Department of ZoologyNehru Memorial College (Autonomous), Puthanampatti (Affiliated to Bharathidasan University)Tamil NaduTiruchirappalliIndia
| | - Jonathan C. Ifemeje
- Department of Biochemistry, Faculty of Natural SciencesChukwuemeka Odumegwu Ojukwu UniversityAnambraNigeria
| | - Shahira M. Ezzat
- Department of Pharmacognosy, Faculty of PharmacyCairo UniversityCairoEgypt
- Department of Pharmacognosy, Faculty of PharmacyOctober University for Modern Sciences and Arts (MSA)GizaEgypt
| | - Michael C. Olisah
- Department of Medical Biochemistry, Faculty of Basic Medical SciencesChukwuemeka Odumegwu Ojukwu University, Uli CampusAnambraNigeria
| | - Chukwudi Jude Chikwendu
- Department of Biochemistry, Faculty of Natural SciencesChukwuemeka Odumegwu Ojukwu UniversityAnambraNigeria
| | - Kamoru A. Adedokun
- Department of ImmunologyRoswell Park Comprehensive Cancer CenterNew YorkBuffaloUSA
| | - Sikiru O. Imodoye
- Department of Oncological Sciences, Huntsman Cancer InstituteUniversity of UtahUtahSalt Lake CityUSA
| | - Ibrahim O. Bello
- Department of Biological SciencesSouthern Illinois University EdwardsvilleIllinoisEdwardsvilleUSA
| | - Hannington Twinomuhwezi
- Department of ChemistryKyambogo University, KyambogoKampalaUganda
- School of Natural and Applied SciencesKampala International UniversityKampalaUganda
| | | |
Collapse
|
4
|
Castro-Muñoz LJ, Vázquez Ulloa E, Sahlgren C, Lizano M, De La Cruz-Hernández E, Contreras-Paredes A. Modulating epigenetic modifications for cancer therapy (Review). Oncol Rep 2023; 49:59. [PMID: 36799181 PMCID: PMC9942256 DOI: 10.3892/or.2023.8496] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/08/2022] [Indexed: 02/12/2023] Open
Abstract
Cancer is a global public health concern. Alterations in epigenetic processes are among the earliest genomic aberrations occurring during cancer development and are closely related to progression. Unlike genetic mutations, aberrations in epigenetic processes are reversible, which opens the possibility for novel pharmacological treatments. Non‑coding RNAs (ncRNAs) represent an essential epigenetic mechanism, and emerging evidence links ncRNAs to carcinogenesis. Epigenetic drugs (epidrugs) are a group of promising target therapies for cancer treatment acting as coadjuvants to reverse drug resistance in cancer. The present review describes central epigenetic aberrations during malignant transformation and explains how epidrugs target DNA methylation, histone modifications and ncRNAs. Furthermore, clinical trials focused on evaluating the effect of these epidrugs alone or in combination with other anticancer therapies and other ncRNA‑based therapies are discussed. The use of epidrugs promises to be an effective tool for reversing drug resistance in some patients with cancer.
Collapse
Affiliation(s)
| | - Elenaé Vázquez Ulloa
- Faculty of Science and Engineering/Cell Biology, University of Turku and Åbo Akademi University, Turku 20500, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku 20500, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering/Cell Biology, University of Turku and Åbo Akademi University, Turku 20500, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku 20500, Finland
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Marcela Lizano
- Unidad de Investigacion Biomedica en Cancer, Instituto Nacional de Cancerología-Universidad Nacional Autonoma de Mexico, Ciudad de Mexico 14080, Mexico
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico, Mexico 04510, Mexico
| | - Erick De La Cruz-Hernández
- Laboratory of Research in Metabolic and Infectious Diseases, Multidisciplinary Academic Division of Comalcalco, Juarez Autonomous University of Tabasco, Comalcalco, Tabasco 86650, Mexico
| | - Adriana Contreras-Paredes
- Unidad de Investigacion Biomedica en Cancer, Instituto Nacional de Cancerología-Universidad Nacional Autonoma de Mexico, Ciudad de Mexico 14080, Mexico
| |
Collapse
|
5
|
Concept of Hybrid Drugs and Recent Advancements in Anticancer Hybrids. Pharmaceuticals (Basel) 2022; 15:ph15091071. [PMID: 36145292 PMCID: PMC9500727 DOI: 10.3390/ph15091071] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a complex disease, and its treatment is a big challenge, with variable efficacy of conventional anticancer drugs. A two-drug cocktail hybrid approach is a potential strategy in recent drug discovery that involves the combination of two drug pharmacophores into a single molecule. The hybrid molecule acts through distinct modes of action on several targets at a given time with more efficacy and less susceptibility to resistance. Thus, there is a huge scope for using hybrid compounds to tackle the present difficulties in cancer medicine. Recent work has applied this technique to uncover some interesting molecules with substantial anticancer properties. In this study, we report data on numerous promising hybrid anti-proliferative/anti-tumor agents developed over the previous 10 years (2011–2021). It includes quinazoline, indole, carbazole, pyrimidine, quinoline, quinone, imidazole, selenium, platinum, hydroxamic acid, ferrocene, curcumin, triazole, benzimidazole, isatin, pyrrolo benzodiazepine (PBD), chalcone, coumarin, nitrogen mustard, pyrazole, and pyridine-based anticancer hybrids produced via molecular hybridization techniques. Overall, this review offers a clear indication of the potential benefits of merging pharmacophoric subunits from multiple different known chemical prototypes to produce more potent and precise hybrid compounds. This provides valuable knowledge for researchers working on complex diseases such as cancer.
Collapse
|
6
|
Genome-wide CRISPR/Cas9 screening identifies determinant of panobinostat sensitivity in acute lymphoblastic leukemia. Blood Adv 2022; 6:2496-2509. [PMID: 35192680 PMCID: PMC9043932 DOI: 10.1182/bloodadvances.2021006152] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/08/2022] [Indexed: 11/29/2022] Open
Abstract
Genome-wide CRISPR/Cas9 screening in the ALL cell line identified mitochondrial activity as the driver of panobinostat resistance. SIRT1 expression sensitized ALL to panobinostat through activating mitochondrial activity and the mitochondria-related apoptosis pathway.
Epigenetic alterations, including histone acetylation, contribute to the malignant transformation of hematopoietic cells and disease progression, as well as the emergence of chemotherapy resistance. Targeting histone acetylation provides new strategies for the treatment of cancers. As a pan-histone deacetylase inhibitor, panobinostat has been approved by the US Food and Drug Administration for the treatment of multiple myeloma and has shown promising antileukemia effects in acute lymphoblastic leukemia (ALL). However, the underlying drug resistance mechanism in ALL remains largely unknown. Using genome-wide Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas)9 (CRISPR/Cas9) screening, we identified mitochondrial activity as the driver of panobinostat resistance in ALL. Mechanistically, ectopic SIRT1 expression activated mitochondrial activity and sensitized ALL to panobinostat through activating mitochondria-related apoptosis pathway. Meanwhile, the transcription level of SIRT1 was significantly associated with panobinostat sensitivity across diverse tumor types and thus could be a potential biomarker of panobinostat response in cancers. Our data suggest that patients with higher SIRT1 expression in cancer cells might benefit from panobinostat treatment, supporting the implementation of combinatorial therapy with SIRT1 or mitochondrial activators to overcome panobinostat resistance.
Collapse
|
7
|
Chandran A, Jayasankar V, Spagnuolo P, Subramanian J. Bioactive Compounds from Curcuma amada and Their Effect on Acute Myeloid Leukemia. Crit Rev Oncog 2022; 27:23-31. [PMID: 37183936 DOI: 10.1615/critrevoncog.2023047542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer with limited chemotherapy options and negative patient outcomes. Investigations with bioactive compounds from dietary sources against cancer have increased in the recent years, which highlight the need for novel therapeutic approaches and new anti-leukemic agents possessing higher efficacy and selectivity for AML cells and fewer negative side effects. Bioactive compounds demonstrated the ability to induce cell cycle blockage and apoptosis or autophagy in cancer cells, as well as inhibition of proliferation/migration and tumor progression, etc. Bioactive compounds isolated from dietary sources such as mango ginger show promise for AML treatment. Curcuma amada roots have been used in traditional medicine and showed antioxidant, antimicrobial and anticancer properties. Bioactive molecules isolated from C. amada showed effects on the mitochondrial metabolism and reduced the viability of multiple leukemic cell lines.
Collapse
Affiliation(s)
- Ajila Chandran
- Department of Chemistry, Eastern Kentucky University, Richmond, KY, USA
| | - Varsha Jayasankar
- Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul Spagnuolo
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
8
|
Abstract
Background: Acute lymphoblastic leukemia (ALL) is a malignant disease characterized by an excessive number of immature lymphocytes, including immature precursors of both B- and T cells. ALL affects children more often than adults. Immature lymphocytes lead to arrested differentiation and proliferation of cells. Its conventional treatments involve medication with dexamethasone, vincristine, and other anticancer drugs. Although the current first-line drugs can achieve effective treatment, they still cannot prevent the recurrence of some patients with ALL. Treatments have high risk of recurrence especially after the first remission. Currently, novel therapies to treat ALL are in need. Autophagy and apoptosis play important roles in regulating cancer development. Autophagy involves degradation of proteins and organelles, and apoptosis leads to cell death. These phenomena are crucial in cancer progression. Past studies reported that many potential anticancer agents regulate intracellular signaling pathways. Methods: The authors discuss the recent research findings on the role of autophagy and apoptosis in ALL. Results: The autophagy and apoptosis are widely used in the treatment of ALL. Most studies showed that many agents regulate autophagy and apoptosis in ALL cell models, clinical trials, and ALL animal models. Conclusions: In summary, activating autophagy and apoptosis pathways are the main strategies for ALL treatments. For ALL, combining new drugs with traditional chemotherapy and glucocorticoids treatments can achieve the greatest therapeutic effect by activating autophagy and apoptosis.
Collapse
Affiliation(s)
- Fang-Liang Huang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC.,Department of Physical Therapy, Hungkuang University, Taichung, Taiwan, ROC.,Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan, ROC
| | - Sheng-Jie Yu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Chia-Ling Li
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| |
Collapse
|
9
|
The Anti-Leukemic Activity of Natural Compounds. Molecules 2021; 26:molecules26092709. [PMID: 34063044 PMCID: PMC8124534 DOI: 10.3390/molecules26092709] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 12/24/2022] Open
Abstract
The use of biologically active compounds has become a realistic option for the treatment of malignant tumors due to their cost-effectiveness and safety. In this review, we aimed to highlight the main natural biocompounds that target leukemic cells, assessed by in vitro and in vivo experiments or clinical studies, in order to explore their therapeutic potential in the treatment of leukemia: acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia (CLL). It provides a basis for researchers and hematologists in improving basic and clinical research on the development of new alternative therapies in the fight against leukemia, a harmful hematological cancer and the leading cause of death among patients.
Collapse
|
10
|
Panina SB, Pei J, Kirienko NV. Mitochondrial metabolism as a target for acute myeloid leukemia treatment. Cancer Metab 2021; 9:17. [PMID: 33883040 PMCID: PMC8058979 DOI: 10.1186/s40170-021-00253-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemias (AML) are a group of aggressive hematologic malignancies resulting from acquired genetic mutations in hematopoietic stem cells that affect patients of all ages. Despite decades of research, standard chemotherapy still remains ineffective for some AML subtypes and is often inappropriate for older patients or those with comorbidities. Recently, a number of studies have identified unique mitochondrial alterations that lead to metabolic vulnerabilities in AML cells that may present viable treatment targets. These include mtDNA, dependency on oxidative phosphorylation, mitochondrial metabolism, and pro-survival signaling, as well as reactive oxygen species generation and mitochondrial dynamics. Moreover, some mitochondria-targeting chemotherapeutics and their combinations with other compounds have been FDA-approved for AML treatment. Here, we review recent studies that illuminate the effects of drugs and synergistic drug combinations that target diverse biomolecules and metabolic pathways related to mitochondria and their promise in experimental studies, clinical trials, and existing chemotherapeutic regimens.
Collapse
Affiliation(s)
| | - Jingqi Pei
- Department of BioSciences, Rice University, Houston, TX, USA
| | | |
Collapse
|
11
|
Shalini, Kumar V. Have molecular hybrids delivered effective anti-cancer treatments and what should future drug discovery focus on? Expert Opin Drug Discov 2020; 16:335-363. [PMID: 33305635 DOI: 10.1080/17460441.2021.1850686] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Cancer continues to be a big threat and its treatment is a huge challenge among the medical fraternity. Conventional anti-cancer agents are losing their efficiency which highlights the need to introduce new anti-cancer entities for treating this complex disease. A hybrid molecule has a tendency to act through varied modes of action on multiple targets at a given time. Thus, there is the significant scope with hybrid compounds to tackle the existing limitations of cancer chemotherapy. AREA COVERED This perspective describes the most significant hybrids that spring hope in the field of cancer chemotherapy. Several hybrids with anti-proliferative/anti-tumor properties currently approved or in clinical development are outlined, along with a description of their mechanism of action and identified drug targets. EXPERT OPINION The success of molecular hybridization in cancer chemotherapy is quite evident by the number of molecules entering into clinical trials and/or have entered the drug market over the past decade. Indeed, the recent advancements and co-ordinations in the interface between chemistry, biology, and pharmacology will help further the advancement of hybrid chemotherapeutics in the future.List of abbreviations: Deoxyribonucleic acid, DNA; national cancer institute, NCI; peripheral blood mononuclear cells, PBMC; food and drug administration, FDA; histone deacetylase, HDAC; epidermal growth factor receptor, EGFR; vascular endothelial growth factor receptor, VEGFR; suberoylanilide hydroxamic acid, SAHA; farnesyltransferase inhibitor, FTI; adenosine triphosphate, ATP; Tamoxifen, TAM; selective estrogen receptor modulator, SERM; structure activity relationship, SAR; estrogen receptor, ER; lethal dose, LD; half maximal growth inhibitory concentration, GI50; half maximal inhibitory concentration, IC50.
Collapse
Affiliation(s)
- Shalini
- Department of Chemistry, Guru Nanak Dev University, Amritsar-India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar-India
| |
Collapse
|
12
|
Oxidative resistance of leukemic stem cells and oxidative damage to hematopoietic stem cells under pro-oxidative therapy. Cell Death Dis 2020; 11:291. [PMID: 32341354 PMCID: PMC7184730 DOI: 10.1038/s41419-020-2488-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
Leukemic stem cells (LSCs) and hematopoietic stem cells (HSCs) are both dependent on the hypoxic bone marrow (BM) microenvironment (also known as the BM niche). There is always fierce competition between the two types of cells, and the former exhibits a greater competitive advantage than the latter via multiple mechanisms. Under hypoxia, the dynamic balance between the generation and clearing of intracellular reactive oxygen species (ROS) is conducive to maintaining a quiescent state of cells. Quiescent LSCs can reside well in the BM niche, avoiding attack by chemotherapeutic agents, which is the cause of chemotherapeutic resistance and relapse in leukemia. HSCs acquire energy mainly through anaerobic glycolysis, whereas LSCs achieve energy metabolism largely through mitochondrial oxidative respiration. Mitochondria are the primary site of ROS generation. Thus, in theory, mitochondria-mediated respiration will cause an increase in ROS generation in LSCs and a higher intracellular oxidative stress level. The sensitivity of the cells to pro-oxidant drugs increases as well, which allows for the selective clearing of LSCs by pro-oxidative therapy. However, HSCs are also highly sensitive to changes in ROS levels, and the toxic effects of pro-oxidant drugs on HSCs poses a major challenge to pro-oxidative therapy in leukemia. Given the above facts, we reviewed studies on the oxidative resistance of LSCs and the oxidative damage to HSCs under pro-oxidative therapy. An in-depth investigation into the oxidative stress status and regulatory mechanisms of LSCs and HSCs in hypoxic environments will promote our understanding of the survival strategy employed by LSCs and the mechanism of the oxidative damage to HSCs in the BM niche, thus facilitating individualized treatment of leukemia patients and helping eliminate LSCs without disturbing normal hematopoietic cells.
Collapse
|
13
|
El‐Naggar AM, Somasekharan SP, Wang Y, Cheng H, Negri GL, Pan M, Wang XQ, Delaidelli A, Rafn B, Cran J, Zhang F, Zhang H, Colborne S, Gleave M, Mandinova A, Kedersha N, Hughes CS, Surdez D, Delattre O, Wang Y, Huntsman DG, Morin GB, Sorensen PH. Class I HDAC inhibitors enhance YB-1 acetylation and oxidative stress to block sarcoma metastasis. EMBO Rep 2019; 20:e48375. [PMID: 31668005 PMCID: PMC6893361 DOI: 10.15252/embr.201948375] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/29/2019] [Accepted: 10/06/2019] [Indexed: 12/13/2022] Open
Abstract
Outcomes for metastatic Ewing sarcoma and osteosarcoma are dismal and have not changed for decades. Oxidative stress attenuates melanoma metastasis, and melanoma cells must reduce oxidative stress to metastasize. We explored this in sarcomas by screening for oxidative stress sensitizers, which identified the class I HDAC inhibitor MS-275 as enhancing vulnerability to reactive oxygen species (ROS) in sarcoma cells. Mechanistically, MS-275 inhibits YB-1 deacetylation, decreasing its binding to 5'-UTRs of NFE2L2 encoding the antioxidant factor NRF2, thereby reducing NFE2L2 translation and synthesis of NRF2 to increase cellular ROS. By global acetylomics, MS-275 promotes rapid acetylation of the YB-1 RNA-binding protein at lysine-81, blocking binding and translational activation of NFE2L2, as well as known YB-1 mRNA targets, HIF1A, and the stress granule nucleator, G3BP1. MS-275 dramatically reduces sarcoma metastasis in vivo, but an MS-275-resistant YB-1K81-to-alanine mutant restores metastatic capacity and NRF2, HIF1α, and G3BP1 synthesis in MS-275-treated mice. These studies describe a novel function for MS-275 through enhanced YB-1 acetylation, thus inhibiting YB-1 translational control of key cytoprotective factors and its pro-metastatic activity.
Collapse
Affiliation(s)
- Amal M El‐Naggar
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
- Department of PathologyFaculty of MedicineMenoufia UniversityShibin El KomEgypt
| | | | - Yemin Wang
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | | | | | - Melvin Pan
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Xue Qi Wang
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Alberto Delaidelli
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Bo Rafn
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Jordan Cran
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Fan Zhang
- Vancouver Prostate CentreVancouverBCCanada
| | - Haifeng Zhang
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | | | | | - Anna Mandinova
- Brigham and Women's HospitalHarvard UniversityBostonMAUSA
| | - Nancy Kedersha
- Massachusetts General HospitalHarvard UniversityBostonMAUSA
| | - Christopher S Hughes
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | | | | | | | - David G Huntsman
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Gregg B Morin
- Michael Smith Genome Sciences CentreVancouverBCCanada
| | - Poul H Sorensen
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| |
Collapse
|
14
|
Peterson JA, Crowther CM, Andrus MB, Kenealey JD. Resveratrol derivatives increase cytosolic calcium by inhibiting plasma membrane ATPase and inducing calcium release from the endoplasmic reticulum in prostate cancer cells. Biochem Biophys Rep 2019; 19:100667. [PMID: 31463373 PMCID: PMC6709415 DOI: 10.1016/j.bbrep.2019.100667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022] Open
Abstract
Resveratrol (RES) is a putative chemotherapeutic naturally found in grapes, peanuts, and Japanese knotweed. Previous studies demonstrate that RES modulates calcium signaling as part of its chemotherapeutic activity. In this study, we determined the chemotherapeutic activity of three RES esters that have been modified at the 4' hydroxyl by the addition of pivalate, butyrate, and isobutyrate. All of the RES derivatives disrupted the calcium signaling in prostate cancer cells more than the parent compound, RES. Further, we demonstrate that the RES derivatives may disrupt the calcium homeostasis by activating calcium release from the endoplasmic reticulum and inhibiting plasma membrane Ca2+-ATPase. The pivalated and butyrated RES derivatives decreased cell viability significantly more than RES. Because pivalated and butyrated RES are more effective than RES at targeting calcium signaling pathways, pivalated and butyrated RES may serve as more effective chemotherapeutics.
Collapse
Key Words
- 2-APB, 2-Aminoethyl diphenylborinate
- AUC, area under the curve
- BuRV, 4′-butyrate resveratrol
- Calcium signaling
- DMEM, Dulbecco's modified Eagle medium
- DMSO, dimethyl sulfoxide
- ER, endoplasmic reticulum
- FBS, fetal bovine serum
- Fura-2
- Fura-2, Fura-2-Acetoxymethyl ester
- HBSS, Ca2+- and Mg2+-free Hank's Balanced Salt Solution
- IP3, inositol triphosphate
- IP3R, inositol triphosphate receptor
- IsoRV, 4′-isobutyrate resveratrol
- MTT, 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- PBS, phosphate-buffered saline
- PIP2, phosphatidylinositol biphosphate
- PIV, 4′-pivalate resveratrol
- PLC, phospholipase C
- PMCA, plasma membrane Ca2+-ATPase
- Plasma membrane Ca2+-ATPase
- Prostate cancer
- RES, resveratrol
- Resveratrol
- SERCA, sarcoendoplasmic reticular Ca2+-ATPase
- TG, thapsigargin
- [Ca2+]i, cytosolic calcium concentration
Collapse
Affiliation(s)
- Joshua A. Peterson
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT, USA
| | - Colton M. Crowther
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Merritt B. Andrus
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Jason D. Kenealey
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT, USA
| |
Collapse
|
15
|
Farhan M, Ullah MF, Faisal M, Farooqi AA, Sabitaliyevich UY, Biersack B, Ahmad A. Differential Methylation and Acetylation as the Epigenetic Basis of Resveratrol's Anticancer Activity. MEDICINES 2019; 6:medicines6010024. [PMID: 30781847 PMCID: PMC6473688 DOI: 10.3390/medicines6010024] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 01/15/2023]
Abstract
Numerous studies support the potent anticancer activity of resveratrol and its regulation of key oncogenic signaling pathways. Additionally, the activation of sirtuin 1, a deacetylase, by resveratrol has been known for many years, making resveratrol perhaps one of the earliest nutraceuticals with associated epigenetic activity. Such epigenetic regulation by resveratrol, and the mechanism thereof, has attracted much attention in the past decade. Focusing on methylation and acetylation, the two classical epigenetic regulations, we showcase the potential of resveratrol as an effective anticancer agent by virtue of its ability to induce differential epigenetic changes. We discuss the de-repression of tumor suppressors such as BRCA-1, nuclear factor erythroid 2-related factor 2 (NRF2) and Ras Associated Domain family-1α (RASSF-1α) by methylation, PAX1 by acetylation and the phosphatase and tensin homologue (PTEN) by both methylation and acetylation, in addition to the epigenetic regulation of oncogenic NF-κB and STAT3 signaling by resveratrol. Further, we evaluate the literature supporting the potentiation of HDAC inhibitors and the inhibition of DNMTs by resveratrol in different human cancers. This discussion underlines a robust epigenetic activity of resveratrol that warrants further evaluation, particularly in clinical settings.
Collapse
Affiliation(s)
- Mohd Farhan
- College of Basic Sciences, King Faisal University, Hofuf 400-Al Ahsa-31982, Saudi Arabia.
| | - Mohammad Fahad Ullah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia.
| | - Mohd Faisal
- Department of Psychiatry, University Hospital Limerick, Limerick V94 T9PX, Ireland.
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan.
| | | | - Bernhard Biersack
- Organic Chemistry Laboratory, Department of Chemistry, University of Bayreuth, Universitaetsstrasse 30, 95447 Bayreuth, Germany.
| | - Aamir Ahmad
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| |
Collapse
|
16
|
Rivera-Del Valle N, Cheng T, Irwin ME, Donnella H, Singh MM, Chandra J. Combinatorial effects of histone deacetylase inhibitors (HDACi), vorinostat and entinostat, and adaphostin are characterized by distinct redox alterations. Cancer Chemother Pharmacol 2018; 81:483-495. [PMID: 29313067 DOI: 10.1007/s00280-017-3509-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE Amongst the epigenetically targeted therapies, targeting of the histone deacetylases (HDACs) has yielded numerous drugs for clinical use in hematological malignancies, but none as yet for acute lymphocytic leukemia (ALL). Single agent activity of HDAC inhibitors (HDACi) has been elusive in ALL, and has prompted study of combinatorial strategies. Because several HDACi raise levels of intracellular oxidative stress, we evaluated combinations of two structurally distinct HDACi with the redox active compound adaphostin in ALL. METHODS The HDACi vorinostat and entinostat were tested in combination with adaphostin in human ALL cell lines. DNA fragmentation, caspase activation, mitochondrial disruption and levels of intracellular peroxides, superoxide and glutathione were measured in cells treated with the HDACi/adaphostin combinations. Antioxidant blockade of cell death induction and gene expression profiling of cells treated with vorinostat/adaphostin versus entinostat/adaphostin combinations were evaluated. RESULTS Both combinations synergistically induced apoptotic DNA fragmentation, which was preceded by an increase in superoxide levels, a reduction in mitochondrial membrane potential, and an increase in caspase-9 activation. The antioxidant N-acetylcysteine (NAC) blocked superoxide generation and prevented reduction of mitochondrial membrane potential. NAC decreased DNA fragmentation and caspase activity in cells treated with adaphostin and vorinostat, but not in those treated with adaphostin and entinostat. Gene expression arrays revealed differential regulation of several redox genes prior to cell death induction. CONCLUSIONS A redox modulatory agent, adaphostin, enhances efficacy of two HDACi, vorinostat or entinostat, but via different mechanisms indicating a point of divergence in the mechanisms of synergy between the two distinct HDACi and adaphostin.
Collapse
Affiliation(s)
- Nilsa Rivera-Del Valle
- Department of Pediatrics Research, Children's Cancer Hospital, The University of Texas (UT) M. D. Anderson Cancer Center, 1515 Holcombe Blvd. Unit 853, Houston, TX, 77030, USA.,Center for Cancer Epigenetics, The University of Texas (UT) M. D. Anderson Cancer Center, Houston, TX, 77030, USA.,MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Tiewei Cheng
- Department of Pediatrics Research, Children's Cancer Hospital, The University of Texas (UT) M. D. Anderson Cancer Center, 1515 Holcombe Blvd. Unit 853, Houston, TX, 77030, USA.,Center for Cancer Epigenetics, The University of Texas (UT) M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mary E Irwin
- Department of Pediatrics Research, Children's Cancer Hospital, The University of Texas (UT) M. D. Anderson Cancer Center, 1515 Holcombe Blvd. Unit 853, Houston, TX, 77030, USA.,Center for Cancer Epigenetics, The University of Texas (UT) M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hayley Donnella
- Department of Pediatrics Research, Children's Cancer Hospital, The University of Texas (UT) M. D. Anderson Cancer Center, 1515 Holcombe Blvd. Unit 853, Houston, TX, 77030, USA.,Center for Cancer Epigenetics, The University of Texas (UT) M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Melissa M Singh
- Department of Pediatrics Research, Children's Cancer Hospital, The University of Texas (UT) M. D. Anderson Cancer Center, 1515 Holcombe Blvd. Unit 853, Houston, TX, 77030, USA.,Center for Cancer Epigenetics, The University of Texas (UT) M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Joya Chandra
- Department of Pediatrics Research, Children's Cancer Hospital, The University of Texas (UT) M. D. Anderson Cancer Center, 1515 Holcombe Blvd. Unit 853, Houston, TX, 77030, USA. .,Center for Cancer Epigenetics, The University of Texas (UT) M. D. Anderson Cancer Center, Houston, TX, 77030, USA. .,MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Zabkiewicz J, Gilmour M, Hills R, Vyas P, Bone E, Davidson A, Burnett A, Knapper S. The targeted histone deacetylase inhibitor tefinostat (CHR-2845) shows selective in vitro efficacy in monocytoid-lineage leukaemias. Oncotarget 2017; 7:16650-62. [PMID: 26934551 PMCID: PMC4941341 DOI: 10.18632/oncotarget.7692] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/17/2016] [Indexed: 01/18/2023] Open
Abstract
Tefinostat (CHR-2845) is a novel monocyte/macrophage-targeted histone deacetylase (HDAC) inhibitor which is cleaved into its active acid by the intracellular esterase human carboxylesterase-1 (hCE-1). The in vitro efficacy of tefinostat was characterised in cell lines and in a cohort of 73 primary AML and CMML samples. Dose-dependent induction of apoptosis and significant growth inhibitory effects were seen in myelomonocytic (M4), monocytic/monoblastic (M5) and CMML samples in comparison to non-monocytoid AML sub-types (p = 0.007). Importantly, no growth inhibitory effects were seen in normal bone marrow CD34+ cells exposed to AML-toxic doses of tefinostat in clonogenic assays. Expression of hCE-1 was measured by intracellular flow cytometry and immunoblotting across the cohort, with highest levels seen in M5 AML patients. hCE-1 levels correlated with significantly increased tefinostat sensitivity (low EC50) as measured by growth inhibition assays (p = 0.001) and concomitant elevation of the mature monocytoid marker CD14+. Strong induction of intracellular histone protein acetylation was observed in tefinostat-responsive samples, as were high levels of the DNA damage sensor γ-H2A.X, highlighting potential biomarkers of patient responsiveness. Synergistic interaction between tefinostat and the current standard treatment cytarabine was demonstrated in dose response and clonogenic assays using simultaneous drug addition in primary samples (median Combination Index value = 0.51). These data provide a strong rationale for the further clinical evaluation of tefinostat in monocytoid-lineage haematological neoplasms including CMML and monocyte-lineage AMLs.
Collapse
Affiliation(s)
- Joanna Zabkiewicz
- Department of Haematology, Experimental Cancer Medicine Centre (ECMC), Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Marie Gilmour
- Department of Haematology, Experimental Cancer Medicine Centre (ECMC), Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Robert Hills
- Department of Haematology, Experimental Cancer Medicine Centre (ECMC), Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Pares Vyas
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | | - Alan Burnett
- Department of Haematology, Experimental Cancer Medicine Centre (ECMC), Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Steven Knapper
- Department of Haematology, Experimental Cancer Medicine Centre (ECMC), Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
18
|
Chen G, Xu Z, Chang G, Hou J, Hu L, Zhang Y, Yu D, Li B, Chang S, Xie Y, Zhang Y, Wei R, Wu H, Xiao W, Sun X, Tao Y, Gao L, Dai B, Shi J, Zhu W. The blueberry component pterostilbene has potent anti-myeloma activity in bortezomib-resistant cells. Oncol Rep 2017; 38:488-496. [PMID: 28560392 DOI: 10.3892/or.2017.5675] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/17/2017] [Indexed: 11/05/2022] Open
Abstract
Multiple myeloma (MM) is an incurable hematologic malignancy because of its drug resistance. Pterostilbene (Pter) is found mainly in blueberries and grapes. The effects of Pter and its exact pharmacologic mechanisms on chemoresistant myeloma are not known. Herein, we investigated the anti-myeloma activity of Pter in bortezomib-resistant cell line H929R and explored the related mechanism of action for the first time. We found that Pter inhibited proliferation of H929R cells and promoted apoptosis of the cells through a caspase-dependent pathway, loss of mitochondrial membrane potential, and activation of Akt and p38 mitogen-activated protein kinase (MAPK) signaling pathways. DNA damage and S-phase arrest might be involved in Pter-related toxicity in H929R cells. Pter and the histone deacetylase inhibitors panobinostat or vorinostat inhibited proliferation of H929R cells in a synergistic manner. These data supported that Pter might be a promising natural compound for relapsed/refractory myeloma therapy, especially against myeloma resistant to bortezomib chemotherapy.
Collapse
Affiliation(s)
- Gege Chen
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Gaomei Chang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jun Hou
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Liangning Hu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yiwen Zhang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Dandan Yu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Bo Li
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Shuaikang Chang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yongsheng Xie
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yong Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Rong Wei
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Huiqun Wu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Wenqin Xiao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xi Sun
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yi Tao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lu Gao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Bojie Dai
- College of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Jumei Shi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| |
Collapse
|
19
|
Li J, Qiu M, Chen L, Liu L, Tan G, Liu J. Resveratrol promotes regression of renal carcinoma cells via a renin-angiotensin system suppression-dependent mechanism. Oncol Lett 2017; 13:613-620. [PMID: 28356937 PMCID: PMC5351218 DOI: 10.3892/ol.2016.5519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/11/2016] [Indexed: 11/10/2022] Open
Abstract
The aim of the present study was to investigate the effect of resveratrol on renal carcinoma cells and explore possible renin-angiotensin system-associated mechanisms. Subsequent to resveratrol treatment, the cell viability, apoptosis rate, cytotoxicity levels, caspase 3/7 activity and the levels of angiotensin II (AngII), AngII type 1 receptor (AT1R), vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2) were evaluated in renal carcinoma cells. The effects of AngII, AT1R, VEGF and COX-2 on resveratrol-induced cell growth inhibition and apoptosis were also examined. The results indicated that resveratrol treatment may suppress growth, induce apoptosis, and decrease AngII, AT1R, VEGF and COX-2 levels in renal carcinoma ACHN and A498 cells. In addition, resveratrol-induced cell growth suppression and apoptosis were reversed when co-culturing with AT1R or VEGF. Thus, resveratrol may suppress renal carcinoma cell proliferation and induce apoptosis via an AT1R/VEGF pathway.
Collapse
Affiliation(s)
- Jianchang Li
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Mingning Qiu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Lieqian Chen
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Lei Liu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Guobin Tan
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Jianjun Liu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
20
|
Chen YF, Liu H, Luo XJ, Zhao Z, Zou ZY, Li J, Lin XJ, Liang Y. The roles of reactive oxygen species (ROS) and autophagy in the survival and death of leukemia cells. Crit Rev Oncol Hematol 2017; 112:21-30. [PMID: 28325262 DOI: 10.1016/j.critrevonc.2017.02.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 11/27/2016] [Accepted: 02/06/2017] [Indexed: 02/07/2023] Open
Abstract
As a clonal disease of hematopoietic stem cells (HSCs), the etiology and pathogenesis of leukemia is not fully understood. Recent studies suggest that cellular homeostasis plays an essential role in maintaining the function of HSCs because dysregulation of cellular homeostasis is one of the major factors underlying the malignant transformation of HSCs. Reactive oxygen species (ROS) and autophagy, key factors regulating cellular homeostasis, are commonly observed in the human body. Autophagy can be induced by ROS through a variety of signaling pathways, and conversely inhibits ROS-induced damage to cells and tissues. ROS and autophagy coordinate to maintain cellular homeostasis. Previous studies have demonstrated that both of ROS and autophagy play important roles in the development of leukemia and are closely involved in drug resistance in leukemia. Interference with cellular homeostasis by promoting programmed leukemia cell death via ROS and autophagy has been verified to be an efficient technique in the treatment of leukemia. However, the critical roles of ROS and autophagy in the development of leukemia are largely unknown. In this review, we summarize the roles of ROS and autophagy in the pathogenesis of leukemia, which may allow the identification of novel targets and drugs for the treatment of leukemia based on the regulation of HSCs homeostasis through ROS and autophagy.
Collapse
Affiliation(s)
- Yong-Feng Chen
- Department of Basic Medical Sciences, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China; Institute of Tumor, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China.
| | - Hao Liu
- School of Pharmacy, Shanghai Jiaotong University, Shanghai 200240, China
| | - Xin-Jing Luo
- Department of Basic Medical Sciences, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China; Institute of Tumor, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China
| | - Zhiqiang Zhao
- Department of Basic Medical Sciences, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China; Institute of Tumor, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China
| | - Zhen-You Zou
- Department of Basic Medical Sciences, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China; Institute of Tumor, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China; Biochemistry Department of Purdue University, West Lafayette, IN 47906, USA
| | - Jing Li
- Department of Histology and Embryology, North SiChuan Medical College, Nanchong 637000, Sichuan, China
| | - Xiao-Jing Lin
- Department of Hematology, the Affiliated Hospital of Guiyang Medical College, Guiyang 550004, China
| | - Yong Liang
- Institute of Tumor, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China.
| |
Collapse
|
21
|
Mannarino L, Paracchini L, Craparotta I, Romano M, Marchini S, Gatta R, Erba E, Clivio L, Romualdi C, D'Incalci M, Beltrame L, Pattini L. A systems biology approach to investigate the mechanism of action of trabectedin in a model of myelomonocytic leukemia. THE PHARMACOGENOMICS JOURNAL 2016; 18:56-63. [PMID: 27958379 PMCID: PMC5817395 DOI: 10.1038/tpj.2016.76] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/31/2016] [Accepted: 08/25/2016] [Indexed: 12/20/2022]
Abstract
This study was designed to investigate the mode of action of trabectedin in myelomonocytic leukemia cells by applying systems biology approaches to mine gene expression profiling data and pharmacological assessment of the cellular effects. Significant enrichment was found in regulons of target genes inferred for specific transcription factors, among which MAFB was the most upregulated after treatment and was central in the transcriptional network likely to be relevant for the specific therapeutic effects of trabectedin against myelomonocytic cells. Using the Connectivity Map, similarity among transcriptional signatures elicited by treatment with different compounds was investigated, showing a high degree of similarity between transcriptional signatures of trabectedin and those of the topoisomerase I inhibitor, irinotecan, and an anti-dopaminergic antagonist, thioridazine. The study highlights the potential importance of systems biology approaches to generate new hypotheses that are experimentally testable to define the specificity of the mechanism of action of drugs.
Collapse
Affiliation(s)
- L Mannarino
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - L Paracchini
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - I Craparotta
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - M Romano
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - S Marchini
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - R Gatta
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.,Department of Bioscience, University of Milan, Milan, Italy
| | - E Erba
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - L Clivio
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - C Romualdi
- Department of Biology, University of Padua, Padua, Italy
| | - M D'Incalci
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - L Beltrame
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - L Pattini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
22
|
Park MH, Kim JH, Chung YH, Lee SH. Bakuchiol sensitizes cancer cells to TRAIL through ROS- and JNK-mediated upregulation of death receptors and downregulation of survival proteins. Biochem Biophys Res Commun 2016; 473:586-92. [DOI: 10.1016/j.bbrc.2016.03.127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/27/2016] [Indexed: 10/22/2022]
|
23
|
Sonnemann J, Kahl M, Siranjeevi PM, Blumrich A, Blümel L, Becker S, Wittig S, Winkler R, Krämer OH, Beck JF. Reverse chemomodulatory effects of the SIRT1 activators resveratrol and SRT1720 in Ewing's sarcoma cells: resveratrol suppresses and SRT1720 enhances etoposide- and vincristine-induced anticancer activity. J Cancer Res Clin Oncol 2015; 142:17-26. [PMID: 26055805 DOI: 10.1007/s00432-015-1994-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/01/2015] [Indexed: 11/27/2022]
Abstract
PURPOSE SIRT1-activating compounds (STACs) may have potential in the management of cancer. However, the best-studied STAC, the naturally occurring compound resveratrol, is reported to have contradictory effects in combination chemotherapy regimens: It has been shown both to increase and to decrease the action of anticancer agents. To shed more light on this issue, we comparatively investigated the impact of resveratrol and the synthetic STAC SRT1720 on the responsiveness of Ewing's sarcoma (ES) cells to the chemotherapeutic drugs etoposide and vincristine. METHODS Because the effects of STACs can depend on the functionality of the tumor suppressor protein p53, we used three ES cell lines differing in their p53 status, i.e., wild-type p53 WE-68 cells, mutant p53 SK-ES-1 cells and p53 null SK-N-MC cells. Single agent and combination therapy effects were assessed by flow cytometric analyses of propidium iodide uptake and mitochondrial depolarization, by measuring caspase 3/7 activity and by gene expression profiling. RESULTS When applied as single agents, both STACs were effective in ES cells irrespective of their p53 status. Strikingly, however, when applied in conjunction with cytostatic agents, the STACs displayed reverse effects: SRT1720 largely enhanced etoposide- and vincristine-induced cell death, while resveratrol inhibited it. Combination index analyses validated the antipodal impact of the STACs on the effectiveness of the chemotherapeutics. CONCLUSION These findings suggest that the synthetic STAC SRT1720 may be useful to enhance the efficacy of anticancer therapy in ES. But they also suggest that the dietary intake of the natural STAC resveratrol may be detrimental during chemotherapy of ES.
Collapse
Affiliation(s)
- Jürgen Sonnemann
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany.
- Klinik für Kinder- und Jugendmedizin, Friedrich-Schiller-Universität Jena, Kochstr. 2, 07745, Jena, Germany.
| | - Melanie Kahl
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Priyanka M Siranjeevi
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Annelie Blumrich
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Lisa Blümel
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Sabine Becker
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Susan Wittig
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - René Winkler
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| | - James F Beck
- Children's Clinic, Department of Pediatric Hematology and Oncology, Jena University Hospital, Jena, Germany
| |
Collapse
|
24
|
Gu S, Chen C, Jiang X, Zhang Z. Resveratrol synergistically triggers apoptotic cell death with arsenic trioxide via oxidative stress in human lung adenocarcinoma A549 cells. Biol Trace Elem Res 2015; 163:112-23. [PMID: 25431299 DOI: 10.1007/s12011-014-0186-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
Arsenic trioxide (As2O3) is a potent anticancer drug for the treatment of acute promyelocytic leukemia. However, the clinical applications of the agent to treat solid tumors are largely compromised by the drug resistance. Our previous study demonstrated that resveratrol, a plant-derived natural product, could potentiate the toxicity of arsenite in lung adenocarcinoma A549 cells at relatively high concentration, indicating that combination of resveratrol and As2O3 may be a helpful strategy to solve the drug resistance of As2O3 in tumor cells. To test this possibility, in the present study, we determined the combined effects of resveratrol and As2O3 in cultured A549 cells. Our results showed that co-treatment of resveratrol with As2O3 resulted in a synergistic augmentation of cytotoxicity and apoptosis in cells at the tested concentration. To further reveal the detailed mechanism of this synergistic effect on cytotoxicity and apoptosis, apoptosis-related proteins, DNA and chromosomal damage, and the level of oxidative stress were also evaluated. Our data revealed that co-treatment with resveratrol and As2O3 caused more genotoxicity and serious oxidative stress in A549 cells than that of single agent treatment. Moreover, resveratrol and As2O3 could also corporately enhance the release of cytochrome c and the expressions of death receptor Fas and FasL. Together, our results suggest that resveratrol and As2O3 synergistically increase the apoptotic cell death in A549 cells through induction of oxidative stress, indicating that the combination of resveratrol with As2O3 may be a promising strategy to increase the clinical efficacy of As2O3 in the treatment of lung tumor.
Collapse
Affiliation(s)
- Shiyan Gu
- Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, People's Republic of China
| | | | | | | |
Collapse
|
25
|
Summerlin N, Soo E, Thakur S, Qu Z, Jambhrunkar S, Popat A. Resveratrol nanoformulations: challenges and opportunities. Int J Pharm 2015; 479:282-90. [PMID: 25572692 DOI: 10.1016/j.ijpharm.2015.01.003] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 12/29/2014] [Accepted: 01/02/2015] [Indexed: 12/28/2022]
Abstract
Resveratrol, a naturally occurring polyphenol and phytoalexin, has received significant attention in recent years due to its vast therapeutic effects including anticancer, antioxidant and anti-inflammatory effects. However, poor pharmacokinetic properties such as low aqueous solubility, low photostability and extensive first pass metabolism result in poor bioavailability, hindering its immense potential. Conventional dosage forms such as dry powder capsules and injections have met with limited success, demonstrating challenges faced in developing an effective formulation. Recently, nanotechnology-based formulations (nanoformulations) are being looked upon as a novel method for improving the pharmacokinetic properties, as well as enhancing targetability and bioavailability of resveratrol. This review outlines the therapeutic potential of resveratrol, explores its mechanisms of action and pharmacokinetic limitations, and discusses the success and challenges of resveratrol-encapsulated nanoparticles in the last decade. Potential techniques to improve encapsulation of the drug within nanoparticles, thereby enhancing its clinical potential are highlighted.
Collapse
Affiliation(s)
- Natalie Summerlin
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Ernest Soo
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Sachin Thakur
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Zhi Qu
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Siddharth Jambhrunkar
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mucosal Diseases Group, Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent St., Woolloongabba, Queensland 4102, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mucosal Diseases Group, Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent St., Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
26
|
Han HY, Kim H, Son YH, Lee G, Jeong SH, Ryu MH. Anti-cancer effects of Kochia scoparia fruit in human breast cancer cells. Pharmacogn Mag 2014; 10:S661-7. [PMID: 25298688 PMCID: PMC4189286 DOI: 10.4103/0973-1296.139812] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/15/2013] [Accepted: 08/30/2014] [Indexed: 01/01/2023] Open
Abstract
Background: The fruit of Kochia scoparia Scharder is widely used as a medicinal ingredient for the treatment of dysuria and skin diseases in China, Japan and Korea. Especially, K. scoparia had been used for breast masses and chest and flank pain. Objective: To investigate the anti-cancer effect of K. scoparia on breast cancer. Materials and Methods: We investigated the anti-cancer effects of K. scoparia, methanol extract (MEKS) in vitro. We examined the effects of MEKS on the proliferation rate, cell cycle arrest, reactive oxygen species (ROS) generation and activation of apoptosis-associated proteins in MDA-MB-231, human breast cancer cells. Results: MTT assay results demonstrated that MEKS decreased the proliferation rates of MDA-MB-231 cells in a dose-dependent manner with an IC50 value of 36.2 μg/ml. MEKS at 25 μg/ml significantly increased the sub-G1 DNA contents of MDA-MB-231 cells to 44.7%, versus untreated cells. In addition, MEKS induced apoptosis by increasing the levels of apoptosis-associated proteins such as cleaved caspase 3, cleaved caspase 8, cleaved caspase 9 and cleaved Poly (ADP-ribose) polymerase (PARP). Conclusion: These results suggest that MEKS inhibits cell proliferation and induces apoptosis in breast cancer cells and that MEKS may have potential chemotherapeutic value for the treatment of human breast cancer.
Collapse
Affiliation(s)
- Hye-Yeon Han
- Department of Oral Pathology, School of Dentistry, Institute of Translational Dental Sciences, Gyeongnam, Korea
| | - Hyungwoo Kim
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Gyeongnam, Korea
| | - Yong Hae Son
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Gyeongnam, Korea
| | - Guemsan Lee
- Department of Herbology, College of Korean Medicine, Wonkwang University, Jeonbuk, Korea
| | - Sung-Hee Jeong
- Department of Oral Medicine, School of Dentistry, Dental Research Institute, Pusan National University, Gyeongnam, Korea
| | - Mi Heon Ryu
- Department of Oral Pathology, School of Dentistry, Institute of Translational Dental Sciences, Gyeongnam, Korea
| |
Collapse
|
27
|
Zhang J, Zhong Q. Histone deacetylase inhibitors and cell death. Cell Mol Life Sci 2014; 71:3885-901. [PMID: 24898083 PMCID: PMC4414051 DOI: 10.1007/s00018-014-1656-6] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 04/23/2014] [Accepted: 05/20/2014] [Indexed: 12/14/2022]
Abstract
Histone deacetylases (HDACs) are a vast family of enzymes involved in chromatin remodeling and have crucial roles in numerous biological processes, largely through their repressive influence on transcription. In addition to modifying histones, HDACs also target many other non-histone protein substrates to regulate gene expression. Recently, HDACs have gained growing attention as HDAC-inhibiting compounds are being developed as promising cancer therapeutics. Histone deacetylase inhibitors (HDACi) have been shown to induce differentiation, cell cycle arrest, apoptosis, autophagy and necrosis in a variety of transformed cell lines. In this review, we mainly discuss how HDACi may elicit a therapeutic response to human cancers through different cell death pathways, in particular, apoptosis and autophagy.
Collapse
Affiliation(s)
- Jing Zhang
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Qing Zhong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
28
|
Bose P, Dai Y, Grant S. Histone deacetylase inhibitor (HDACI) mechanisms of action: emerging insights. Pharmacol Ther 2014; 143:323-36. [PMID: 24769080 PMCID: PMC4117710 DOI: 10.1016/j.pharmthera.2014.04.004] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 04/10/2014] [Indexed: 02/05/2023]
Abstract
Initially regarded as "epigenetic modifiers" acting predominantly through chromatin remodeling via histone acetylation, HDACIs, alternatively referred to as lysine deacetylase or simply deacetylase inhibitors, have since been recognized to exert multiple cytotoxic actions in cancer cells, often through acetylation of non-histone proteins. Some well-recognized mechanisms of HDACI lethality include, in addition to relaxation of DNA and de-repression of gene transcription, interference with chaperone protein function, free radical generation, induction of DNA damage, up-regulation of endogenous inhibitors of cell cycle progression, e.g., p21, and promotion of apoptosis. Intriguingly, this class of agents is relatively selective for transformed cells, at least in pre-clinical studies. In recent years, additional mechanisms of action of these agents have been uncovered. For example, HDACIs interfere with multiple DNA repair processes, as well as disrupt cell cycle checkpoints, critical to the maintenance of genomic integrity in the face of diverse genotoxic insults. Despite their pre-clinical potential, the clinical use of HDACIs remains restricted to certain subsets of T-cell lymphoma. Currently, it appears likely that the ultimate role of these agents will lie in rational combinations, only a few of which have been pursued in the clinic to date. This review focuses on relatively recently identified mechanisms of action of HDACIs, with particular emphasis on those that relate to the DNA damage response (DDR), and discusses synergistic strategies combining HDACIs with several novel targeted agents that disrupt the DDR or antagonize anti-apoptotic proteins that could have implications for the future use of HDACIs in patients with cancer.
Collapse
Affiliation(s)
- Prithviraj Bose
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yun Dai
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA; Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA; Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
29
|
Al-Hussaini M, DiPersio JF. Small molecule inhibitors in acute myeloid leukemia: from the bench to the clinic. Expert Rev Hematol 2014; 7:439-64. [PMID: 25025370 PMCID: PMC4283573 DOI: 10.1586/17474086.2014.932687] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many patients with acute myeloid leukemia will eventually develop refractory or relapsed disease. In the absence of standard therapy for this population, there is currently an urgent unmet need for novel therapeutic agents. Targeted therapy with small molecule inhibitors represents a new therapeutic intervention that has been successful for the treatment of multiple tumors (e.g., gastrointestinal stromal tumors, chronic myelogenous leukemia). Hence, there has been great interest in generating selective small molecule inhibitors targeting critical pathways of proliferation and survival in acute myeloid leukemia. This review highlights a selective group of intriguing therapeutic agents and their presumed targets in both preclinical models and in early human clinical trials.
Collapse
Affiliation(s)
- Muneera Al-Hussaini
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis Missouri
| | - John F. DiPersio
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis Missouri
- Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St Louis Missouri
| |
Collapse
|
30
|
Vargas JE, Filippi-Chiela EC, Suhre T, Kipper FC, Bonatto D, Lenz G. Inhibition of HDAC increases the senescence induced by natural polyphenols in glioma cells. Biochem Cell Biol 2014; 92:297-304. [PMID: 25070040 DOI: 10.1139/bcb-2014-0022] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cellular senescence is an irreversible block of cellular division, and induction of senescence is being considered for treatment of many cancer types, mainly those resistant to classical pro-apoptotic therapies. Resveratrol (Rsv) and quercetin (Quer), two natural polyphenols, are able to induce senescence in different cancer models, including gliomas, the most common and aggressive primary brain tumor. These polyphenols modulate the activity of several proteins involved in cell growth and death in cancer cells, including histone deacetylases (HDAC), but the role of HDAC in senescence induced by Rsv and Quer is unclear. The HDAC inhibitor sodium butyrate (NaB) potentiated the pro-senescent effect of Rsv and Quer in human and rat glioma cell lines but not in normal rat astrocytes. Furthermore, the increment of Quer-induced senescence by NaB was accompanied by an increase of reactive oxygen species levels and an increment of the number of cells with nuclear abnormalities. Altogether, these data support a positive role of HDAC inhibition on the senescence induced by these polyphenols, and therefore co-treatment of HDAC inhibitors and polyphenols emerges as a potential alternative for gliomas.
Collapse
Affiliation(s)
- José E Vargas
- a Department of Biophysics and Center of Biotechnology, IB, Federal University of Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, Porto Alegre, CEP 91501-970 RS, Brazil
| | | | | | | | | | | |
Collapse
|
31
|
Yuan H, Li AJ, Ma SL, Cui LJ, Wu B, Yin L, Wu MC. Inhibition of autophagy significantly enhances combination therapy with sorafenib and HDAC inhibitors for human hepatoma cells. World J Gastroenterol 2014; 20:4953-4962. [PMID: 24833845 PMCID: PMC4009527 DOI: 10.3748/wjg.v20.i17.4953] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 11/13/2013] [Accepted: 01/02/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To clarify whether histone deacetylase inhibitors histone deacetylase inhibitors (HDACIs) can sensitize hepatocellular carcinoma (HCC) cells to sorafenib treatment.
METHODS: Bax, Bcl-2, ATG5-ATG12, p21, and p27 protein levels in Hep3B, HepG2, and PLC/PRF/5 cells were examined by Western blot. CCK8 and a fluorometric caspase-3 assay were used to examine cellular viability and apoptosis levels. The effect of Beclin-1 on sensitization of HCC cells to sorafenib was examined by transfecting Beclin-1 siRNA into Hep3B, HepG2, and PLC/PRF/5 cells.
RESULTS: Autophagy inhibition enhances the inhibitory effects of vorinostat and sorafenib alone or in combination on HCC cell growth. Vorinostat and sorafenib synergistically induced apoptosis and cell cycle alterations. Western blot data indicated that HDACIs and Beclin-1 knockdown increased the p53 acetylation level. The knockdown of Beclin-1 enhanced the synergistic effect of the combination of vorinostat with sorafenib.
CONCLUSION: HDACIs can sensitize HCC cells to sorafenib treatment by regulating the acetylation level of Beclin-1.
Collapse
|
32
|
Bergadà L, Yeramian A, Sorolla A, Matias-Guiu X, Dolcet X. Antioxidants impair anti-tumoral effects of Vorinostat, but not anti-neoplastic effects of Vorinostat and caspase-8 downregulation. PLoS One 2014; 9:e92764. [PMID: 24651472 PMCID: PMC3961419 DOI: 10.1371/journal.pone.0092764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 02/26/2014] [Indexed: 11/18/2022] Open
Abstract
We have recently demonstrated that histone deacetylase inhibitor, Vorinostat, applied as a single therapy or in combination with caspase-8 downregulation exhibits high anti-tumoral activity on endometrial carcinoma cell lines. In the present study, we have assessed the signalling processes underlying anti-tumoral effects of Vorinostat. Increasing evidence suggests that reactive oxygen species are responsible for histone deacetylase inhibitor-induced cell killing. We have found that Vorinostat induces formation of reactive oxygen species and DNA damage. To investigate the role of oxidative stress as anti-neoplastic mechanism, we have evaluated the effects of different antioxidants (Bha, Nac and Tiron) on endometrial carcinoma cell line Ishikawa treated with Vorinostat. We show that Bha, Nac and Tiron markedly inhibited the cytotoxic effects of Vorinostat, increasing cell viability in vitro. We found that all three antioxidants did not inhibited accumulation of acetyl Histone H4, so that antioxidants did not inhibit Vorinostat activity. Finally, we have evaluated the effects of antioxidants on anti-tumoral activity of Vorinostat as monotherapy or in combination with caspase-8 downregulation in vivo. Interestingly, antioxidants blocked the reduction of tumour growth caused by Vorinostat, but they were unable to inhibit anti-tumoral activity of Vorinostat plus caspase-8 inhibition.
Collapse
Affiliation(s)
- Laura Bergadà
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Andree Yeramian
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Annabel Sorolla
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Xavier Dolcet
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
- * E-mail:
| |
Collapse
|
33
|
Wu ML, Li H, Yu LJ, Chen XY, Kong QY, Song X, Shu XH, Liu J. Short-term resveratrol exposure causes in vitro and in vivo growth inhibition and apoptosis of bladder cancer cells. PLoS One 2014; 9:e89806. [PMID: 24587049 PMCID: PMC3934942 DOI: 10.1371/journal.pone.0089806] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/24/2014] [Indexed: 12/31/2022] Open
Abstract
Conventional adjuvant chemotherapies for bladder transitional cell carcinomas (TCCs) may cause strong systemic toxicity and local irritation. Non-toxic resveratrol inhibits TCC cell growth but its feasibility in clinical management of TCCs remains obscure. This study aimed to evaluate the safety and anti-TCC efficacy of resveratrol, using the experimental models closer to the clinical treatment condition. Human TCC EJ cells were exposed to 100 µM, 150 µM and 200 µM resveratrol respectively for 1 hour and 2 hours to mimic intravesical drug instillation and the cell responses were analyzed by multiple experimental approaches. An orthotopic TCC nude mouse model was established by injecting EJ cells into the sub-urothelial layer and used for short-term intravesical resveratrol instillation. The safety of resveratrol instillation was evaluated and compared with that of MCC. The results revealed that 2 h 150 µM or 200 µM resveratrol treatment leaded to remarkable S phase arrest and apoptosis at 72 h time-point, accompanied with attenuated phosphorylation, nuclear translocation and transcription of STAT3, down-regulation of STAT3 downstream genes (survivin, cyclinD1, c-Myc and VEGF) and nuclear translocations of Sirt1 and p53. The importance of STAT3 signaling in cell growth was confirmed by treating EJ cells with JAK2 inhibitor tyrphostin AG490. The efficacy and safety of resveratrol instillation were proved by the findings from nude mouse orthotopic xenograft models, because this treatment caused growth suppression, distinctive apoptosis and STAT3 inactivation of the transplanted tumors without affecting normal urothelium. Our results thus suggest for the first time the practical values of resveratrol as a safe and effective agent in the post-operative treatment of TCCs.
Collapse
Affiliation(s)
- Mo-Li Wu
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Hong Li
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Li-Jun Yu
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiao-Yan Chen
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Qing-You Kong
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xue Song
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiao-Hong Shu
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jia Liu
- Liaoning Laboratory of Cancer Genetics and Epigenomics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- * E-mail:
| |
Collapse
|
34
|
Wang JH, Shih KS, Wu YW, Wang AW, Yang CR. Histone deacetylase inhibitors increase microRNA-146a expression and enhance negative regulation of interleukin-1β signaling in osteoarthritis fibroblast-like synoviocytes. Osteoarthritis Cartilage 2013; 21:1987-96. [PMID: 24107356 DOI: 10.1016/j.joca.2013.09.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/14/2013] [Accepted: 09/18/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE MiR-146a exerts negative control on inflammatory responses by suppressing cytokine-induced expression of interleukin-1 receptor-associated kinase-1 (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6) by impairing NF-κB activity and inhibiting the expression of target genes. Recent study suggests that histone deacetylases (HDACs) are involved in the regulation of microRNA (miRNA) expression. Therefore, we determined whether HDAC inhibitors can increase miR-146a expression, thereby inhibiting interleukin-1β (IL-1β)-induced signaling in osteoarthritis fibroblast-like synoviocytes (OA-FLS). METHOD MiRNA expression was analyzed using real-time PCR. IL-1β-induced downstream signals and cytokine expression were evaluated using Western blotting and ELISA. Transcription factors regulating promoter activation were identified using chromatin immunoprecipitation assays. RESULTS IL-1β treatment of OA-FLS induced a mild (1.7-fold) increase in miR-146a expression that was unable to appropriately downregulate IRAK1 and TRAF6 expression. HDAC inhibitors, SAHA (vorinostat), and LBH589 (panobinostat) significantly (6.1- and 5.4-fold) elevated miR-146a expression by increasing the binding of the transcription factor NF-κB to the miR-146a promoter, and negatively regulated IL-1β-induced IKK/IκB/p65 phosphorylation signaling and IL-6 secretion. The increase in miR-146a expression induced by the HDAC inhibitors was prevented by transfection of miR-146a inhibitor or HDAC1 (class I HDAC), HDAC4 (class IIa HDAC), and HDAC6 (class IIb HDAC) overexpression, suggesting that they were due to inhibition of HDAC activity. CONCLUSIONS Our study demonstrated that HDAC inhibitor treatment in OA-FLS significantly increased miR-146a expression and mediated markedly negative regulation to inhibit IL-1β-induced signaling and cytokine secretion. Our results indicate the potential rationale of anti-inflammatory effects for HDAC inhibitors.
Collapse
Affiliation(s)
- J H Wang
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei 10051, Taiwan
| | | | | | | | | |
Collapse
|