1
|
Wawruszak A, Luszczki J, Bartuzi D, Kalafut J, Okon E, Czerwonka A, Stepulak A. Selisistat, a SIRT1 inhibitor, enhances paclitaxel activity in luminal and triple-negative breast cancer: in silico, in vitro, and in vivo studies. J Enzyme Inhib Med Chem 2025; 40:2458554. [PMID: 39935420 PMCID: PMC11823383 DOI: 10.1080/14756366.2025.2458554] [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: 03/15/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 02/13/2025] Open
Abstract
Sirtuins (SIRTs) are NAD+-dependent histone deacetylases, which play a key role in cancer progression; however, their prognostic values in breast cancer (BC) remain a subject of debate and controversy. Accumulative evidence suggests that each sirtuin possesses individual character, implicating its role in the regulation of multifaceted biological functions leading to BC initiation, progression and metastasis. Selisistat (EX527) is a potent, cell permeable, highly selective SIRT1 inhibitor. In the study, the tumour-suppressive effects of the SIRT1 inhibitor EX527 (selisistat) alone and in combination with paclitaxel (PAX) in different breast cancer cell lines and zebrafish xenograft models were investigated. The type of pharmacological drug-drug interaction between EX527 and PAX was determined using the isobolographic method. EX527 and PAX used individually inhibited proliferation, induced apoptosis and caused cell cycle arrest in G1 and subG1/G2 phases. Interestingly, the combination of these compounds used in the 1:1 dose-ratio augmented all these effects (IC50add 29.52 ± 3.29 - 38.45 ± 5.26). The co-treatment of EX527 with PAX generated desirable additive drug-drug interaction. The simultaneous application of EX527 and PAX induced a stronger inhibition of tumour growth compared to individual treatments in zebrafish xenografts. In silico analysis revealed a protein-protein interaction pathway (SIRT1-AKT-S1PR1-GNAI1/GNAO1-Tubulin) connecting molecular targets of both ligands. To summarise, the combination of EX527 and PAX more effectively impairs breast cancer cell growth compared to individual treatments. However, further investigations are required to clarify the specific targets and molecular mechanisms underlying the activity of EX527:PAX in other preclinical models.
Collapse
Affiliation(s)
- Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Jarogniew Luszczki
- Department of Occupational Medicine, Medical University of Lublin, Lublin, Poland
| | - Damian Bartuzi
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modelling Laboratory, Medical University of Lublin, Lublin, Poland
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Joanna Kalafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Arkadiusz Czerwonka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
2
|
Jessop E, Young N, Garcia-Del-Valle B, Crusher JT, Obara B, Karakesisoglou I. SIRT2 Inhibition by AGK2 Promotes Perinuclear Cytoskeletal Organisation and Reduces Invasiveness of MDA-MB-231 Triple-Negative Breast Cancer Cells in Confined In Vitro Models. Cells 2024; 13:2023. [PMID: 39682770 PMCID: PMC11639776 DOI: 10.3390/cells13232023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype characterised by the absence of targetable hormone receptors and increased metastatic rates. As nuclear softening strongly contributes to TNBC's enhanced metastatic capacity, increasing the nuclear stiffness of TNBC cells may present a promising therapeutic avenue. Previous evidence has demonstrated the ability of Sirtuin 2 (SIRT2) inhibition to induce cytoskeletal reorganisation, a key factor in regulating nuclear mechanics. Thus, our study aimed to investigate the effect of SIRT2 inhibition on the nuclear mechanics and migratory behaviour of TNBC cells. To achieve this, SIRT2 was pharmacologically inhibited in MDA-MB-231 cells using AGK2, a SIRT2-specific inhibitor. Although SIRT2 inhibition had no effect on LINC complex composition, the AGK2-treated MDA-MB-231 cells displayed more prominent perinuclear organisations of acetylated α-tubulin, vimentin, and F-actin. Additionally, the nuclei of the AGK2-treated MDA-MB-231 cells exhibited greater resistance to collapse under osmotic shock. Scratch-wound assays also revealed that SIRT2 inhibition led to polarity defects in the MDA-MB-231 cells, while in vitro space-restrictive invasion assays highlighted their reduced migratory capacity upon AGK2 treatment. Taken together, our findings suggest that SIRT2 inhibition promotes a perinuclear cytoskeletal organisation in MDA-MB-231 cells, which enhances their nuclear rigidity and impedes their invasion through confined spaces in vitro.
Collapse
Affiliation(s)
- Emily Jessop
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Natalie Young
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Beatriz Garcia-Del-Valle
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Jack T. Crusher
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Boguslaw Obara
- School of Computing, Newcastle University, Newcastle upon Tyne NE4 5TG, UK;
| | - Iakowos Karakesisoglou
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| |
Collapse
|
3
|
Miller MJ, Akter D, Mahmud J, Chan GC. Human cytomegalovirus modulates mTORC1 to redirect mRNA translation within quiescently infected monocytes. J Virol 2024; 98:e0188823. [PMID: 38289104 PMCID: PMC10878035 DOI: 10.1128/jvi.01888-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 02/21/2024] Open
Abstract
Human cytomegalovirus (HCMV) utilizes peripheral blood monocytes as a means to systemically disseminate throughout the host. Following viral entry, HCMV stimulates non-canonical Akt signaling leading to the activation of mTORC1 and the subsequent translation of select antiapoptotic proteins within infected monocytes. However, the full extent to which the HCMV-initiated Akt/mTORC1 signaling axis reshapes the monocyte translatome is unclear. We found HCMV entry alone was able to stimulate widescale changes to mRNA translation levels and that inhibition of mTOR, a component of mTORC1, dramatically attenuated HCMV-induced protein synthesis. Although monocytes treated with normal myeloid growth factors also exhibited increased levels of translation, mTOR inhibition had no effect, suggesting HCMV activation of mTOR stimulates the acquisition of a unique translatome within infected monocytes. Indeed, polyribosomal profiling of HCMV-infected monocytes identified distinct prosurvival transcripts that were preferentially loaded with ribosomes when compared to growth factor-treated cells. Sirtuin 1 (SIRT1), a deacetylase that exerts prosurvival effects through regulation of the PI3K/Akt pathway, was found to be highly enriched following HCMV infection in an mTOR-dependent manner. Importantly, SIRT1 inhibition led to the death of HCMV-infected monocytes while having minimal effect on uninfected cells. SIRT1 also supported a positive feedback loop to sustain Akt/mTORC1 signaling following viral entry. Taken together, HCMV profoundly reshapes mRNA translation in an mTOR-dependent manner to enhance the synthesis of select factors necessary for the survival of infected monocytes.IMPORTANCEHuman cytomegalovirus (HCMV) infection is a significant cause of morbidity and mortality among the immunonaïve and immunocompromised. Peripheral blood monocytes are a major cell type responsible for disseminating the virus from the initial site of infection. In order for monocytes to mediate viral spread within the host, HCMV must subvert the naturally short lifespan of these cells. In this study, we performed polysomal profiling analysis, which demonstrated HCMV to globally redirect mRNA translation toward the synthesis of cellular prosurvival factors within infected monocytes. Specifically, HCMV entry into monocytes induced the translation of cellular SIRT1 to generate an antiapoptotic state. Defining the precise mechanisms through which HCMV stimulates survival will provide insight into novel anti-HCMV drugs able to target infected monocytes.
Collapse
Affiliation(s)
- Michael J. Miller
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Dilruba Akter
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jamil Mahmud
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Gary C. Chan
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
4
|
Lazaro-Pena MI, Ward ZC, Yang S, Strohm A, Merrill AK, Soto CA, Samuelson AV. HSF-1: Guardian of the Proteome Through Integration of Longevity Signals to the Proteostatic Network. FRONTIERS IN AGING 2022; 3:861686. [PMID: 35874276 PMCID: PMC9304931 DOI: 10.3389/fragi.2022.861686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/13/2022] [Indexed: 12/15/2022]
Abstract
Discoveries made in the nematode Caenorhabditis elegans revealed that aging is under genetic control. Since these transformative initial studies, C. elegans has become a premier model system for aging research. Critically, the genes, pathways, and processes that have fundamental roles in organismal aging are deeply conserved throughout evolution. This conservation has led to a wealth of knowledge regarding both the processes that influence aging and the identification of molecular and cellular hallmarks that play a causative role in the physiological decline of organisms. One key feature of age-associated decline is the failure of mechanisms that maintain proper function of the proteome (proteostasis). Here we highlight components of the proteostatic network that act to maintain the proteome and how this network integrates into major longevity signaling pathways. We focus in depth on the heat shock transcription factor 1 (HSF1), the central regulator of gene expression for proteins that maintain the cytosolic and nuclear proteomes, and a key effector of longevity signals.
Collapse
Affiliation(s)
- Maria I. Lazaro-Pena
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Zachary C. Ward
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Sifan Yang
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Alexandra Strohm
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Toxicology Training Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Alyssa K. Merrill
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Toxicology Training Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Celia A. Soto
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
- Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Andrew V. Samuelson
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- *Correspondence: Andrew V. Samuelson,
| |
Collapse
|
5
|
Wawruszak A, Luszczki J, Czerwonka A, Okon E, Stepulak A. Assessment of Pharmacological Interactions between SIRT2 Inhibitor AGK2 and Paclitaxel in Different Molecular Subtypes of Breast Cancer Cells. Cells 2022; 11:1211. [PMID: 35406775 PMCID: PMC8998062 DOI: 10.3390/cells11071211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/20/2022] Open
Abstract
Breast carcinoma (BC) is the most commonly diagnosed type of cancer in women in the world. Although the advances in the treatment of BC patients are significant, numerous side effects, severe toxicity towards normal cells as well as the multidrug resistance (MDR) phenomenon restrict the effectiveness of the therapies used. Therefore, new active compounds which decrease the MDR, extend disease-free survival, thereby ameliorating the effectiveness of the current treatment regimens, are greatly needed. Histone deacetylase inhibitors (HDIs), including sirtuin inhibitors (SIRTi), are the epigenetic antitumor agents which induce a cytotoxic effect in different types of cancer cells, including BC cells. Currently, combined forms of therapy with two or even more chemotherapeutics are promising antineoplastic tools to obtain a better response to therapy and limit adverse effects. Thus, on the one hand, much more effective chemotherapeutics, e.g., sirtuin inhibitors (SIRTi), are in demand; on the other hand, combinations of accepted cytostatics are trialed. Thus, the aim of our research was to examine the combination effects of a renowned cytotoxic drug paclitaxel (PAX) and SIRT2 inhibitor AGK2 on the proliferation and viability of the T47D, MCF7, MDA-MB-231, MDA-MB-468, BT-549 and HCC1937 BC cells. Moreover, cell cycle arrest and apoptosis induction were explored. The type of pharmacological interactions between AGK2 and PAX in different molecular subtypes of BC cells was assessed using the advanced isobolographic method. Our findings demonstrated that the tested active agents singly inhibited viability and proliferation of BC cells as well as induced cell cycle arrest and apoptosis in the cell-dependent context. Additionally, AGK2 increased the antitumor effect of PAX in most BC cell lines. We observed that, depending on the BC cell lines, the combinations of tested drugs showed synergistic, additive or antagonistic pharmacological interaction. In conclusion, our studies demonstrated that the consolidated therapy with the use of AGK2 and PAX can be considered as a potential therapeutic regimen in the personalized cure of BC patients in the future.
Collapse
Affiliation(s)
- Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (A.C.); (E.O.); (A.S.)
| | - Jarogniew Luszczki
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Arkadiusz Czerwonka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (A.C.); (E.O.); (A.S.)
| | - Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (A.C.); (E.O.); (A.S.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (A.C.); (E.O.); (A.S.)
| |
Collapse
|
6
|
Chen X, Lu W, Wu D. Sirtuin 2 (SIRT2): Confusing Roles in the Pathophysiology of Neurological Disorders. Front Neurosci 2021; 15:614107. [PMID: 34108853 PMCID: PMC8180884 DOI: 10.3389/fnins.2021.614107] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 04/12/2021] [Indexed: 01/05/2023] Open
Abstract
As a type of nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases, sirtuin 2 (SIRT2) is predominantly found in the cytoplasm of cells in the central nervous system (CNS), suggesting its potential role in neurological disorders. Though SIRT2 is generally acknowledged to accelerate the development of neurological pathologies, it protects the brain from deterioration in certain circumstances. This review summarized the complex roles SIRT2 plays in the pathophysiology of diverse neurological disorders, compared and analyzed the discrete roles of SIRT2 in different conditions, and provided possible explanations for its paradoxical functions. In the future, the rapid growth in SIRT2 research may clarify its impacts on neurological disorders and develop therapeutic strategies targeting this protein.
Collapse
Affiliation(s)
- Xiuqi Chen
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wenmei Lu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Broussy S, Laaroussi H, Vidal M. Biochemical mechanism and biological effects of the inhibition of silent information regulator 1 (SIRT1) by EX-527 (SEN0014196 or selisistat). J Enzyme Inhib Med Chem 2021; 35:1124-1136. [PMID: 32366137 PMCID: PMC7241506 DOI: 10.1080/14756366.2020.1758691] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The human sirtuin silent information regulator 1 (SIRT1) is a NAD+-dependent deacetylase enzyme. It deacetylates many protein substrates, including histones and transcription factors, thereby controlling many physiological and pathological processes. Several synthetic inhibitors and activators of SIRT1 have been developed, and some therapeutic applications have been explored. The indole EX-527 and its derivatives are among the most potent and selective SIRT1 inhibitors. EX-527 has been often used as a pharmacological tool to explore the effect of SIRT1 inhibition in various cell types. Its therapeutic potential has, therefore, been evaluated in animal models for several pathologies, including cancer. It has also been tested in phase II clinical trial for the treatment of Huntington’s disease (HD). In this review, we will provide an overview of the literature on EX-527, including its mechanism of inhibition and biological studies.
Collapse
Affiliation(s)
- Sylvain Broussy
- Université de Paris, Faculté de Pharmacie de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, Paris, France
| | - Hanna Laaroussi
- Université de Paris, Faculté de Pharmacie de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, Paris, France
| | - Michel Vidal
- Université de Paris, Faculté de Pharmacie de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, Paris, France.,Service biologie du médicament, toxicologie, AP-HP, Hôpital Cochin, Paris, France
| |
Collapse
|
8
|
Philips BJ, Kumar A, Burki S, Ryan JP, Noda K, D'Cunha J. Triptolide-induced apoptosis in non-small cell lung cancer via a novel miR204-5p/Caveolin-1/Akt-mediated pathway. Oncotarget 2020; 11:2793-2806. [PMID: 32733649 PMCID: PMC7367654 DOI: 10.18632/oncotarget.27672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is one of the most prevalent malignancies world-wide with non-small cell lung cancer (NSCLC) comprising nearly 80% of all cases. Unfortunately, many lung cancer patients are diagnosed at advanced stages of the disease with an associated poor prognosis. Recently, the Chinese herb root extract Triptolide/Minnelide (TL) has shown significant promise as a therapeutic agent for NSCLC treatment both in vitro and in vivo. The aim of this study was to investigate the underlying mechanism(s) of action regarding TL-induced cytotoxicity in NSCLC. We demonstrate that triptolide treatment of A549 and H460 NSCLC cells decreases Caveolin-1 (CAV-1) mRNA/protein expression, resulting in activation of the Akt/Bcl-2-mediated mitochondrial apoptosis pathway. CAV-1 down-regulation was triggered by Micro-RNA 204-5p (miR204-5p) up-regulation and could be significantly blocked by pre-treatment with both Sirt-1/Sirt-3 specific siRNA and SIRT-1/SIRT-3 enzyme inhibitors, EX-527 and nicotinamide. Overall, our results provide evidence for a novel mechanism by which TL exerts its cytotoxic effects on NSCLC via CAV-1 down-regulation. Furthermore, these findings demonstrate a pivotal role for TL induction of the Akt/Bax pathway in apoptosis of human lung cancer.
Collapse
Affiliation(s)
- Brian J Philips
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ajay Kumar
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah Burki
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - John P Ryan
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kentaro Noda
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan D'Cunha
- Department of Cardiothoracic Surgery, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
9
|
Kozako T, Itoh Y, Honda SI, Suzuki T. Epigenetic Control Using Small Molecules in Cancer. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/978-3-030-32857-3_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
10
|
Carafa V, Poziello A, Della Torre L, Giovannelli P, Di Donato M, Safadeh E, Yu Z, Baldi A, Castoria G, Tomaselli D, Mai A, Rotili D, Nebbioso A, Altucci L. Enzymatic and Biological Characterization of Novel Sirtuin Modulators against Cancer. Int J Mol Sci 2019; 20:ijms20225654. [PMID: 31726691 PMCID: PMC6888689 DOI: 10.3390/ijms20225654] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/05/2019] [Accepted: 11/08/2019] [Indexed: 01/13/2023] Open
Abstract
Sirtuins, a family of nicotinamide adenine dinucleotide (NAD+)-dependent lysine deacetylases, are promising targets for anticancer treatment. Recently, we characterized a novel pan-sirtuin (SIRT) inhibitor, MC2494, displaying antiproliferative effects and able to induce death pathways in several human cancer cell lines and decrease tumor growth in vivo. Based on the chemical scaffold of MC2494, and by applying a structure–activity relationship approach, we developed a small library of derivative compounds and extensively analyzed their enzymatic action at cellular level as well as their ability to induce cell death. We also investigated the effect of MC2494 on regulation of cell cycle progression in different cancer cell lines. Our investigations indicated that chemical substitutions applied to MC2494 scaffold did not confer higher efficacy in terms of biological activity and SIRT1 inhibition, but carbethoxy-containing derivatives showed higher SIRT2 specificity. The carbethoxy derivative of MC2494 and its 2-methyl analog displayed the strongest enzymatic activity. Applied chemical modifications improved the enzymatic selectivity of these SIRT inhibitors. Additionally, the observed activity of MC2494 via cell cycle and apoptotic regulation and inhibition of cell migration supports the potential role of SIRTs as targets in tumorigenesis and makes SIRT-targeting molecules good candidates for novel pharmacological approaches in personalized medicine.
Collapse
Affiliation(s)
- Vincenzo Carafa
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Angelita Poziello
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Laura Della Torre
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Pia Giovannelli
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Marzia Di Donato
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Elham Safadeh
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Zhijun Yu
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Alfonso Baldi
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Gabriella Castoria
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
| | - Daniela Tomaselli
- Dipartimento di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Roma, Italy;
| | - Antonello Mai
- Dipartimento di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Roma, Italy;
- Correspondence: (A.M.); (D.R.); (A.N.); (L.A.); Tel.: +39-0649-913-392 (A.M.); +39-0649-913-891 (D.R.); +39-0815-665-682 (A.N.); +39-0815-667-569 (L.A.); Fax: +39-064-9693-268 (A.M. & D.R.); +39-081-450-169 (A.N. & L.A.)
| | - Dante Rotili
- Dipartimento di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Roma, Italy;
- Correspondence: (A.M.); (D.R.); (A.N.); (L.A.); Tel.: +39-0649-913-392 (A.M.); +39-0649-913-891 (D.R.); +39-0815-665-682 (A.N.); +39-0815-667-569 (L.A.); Fax: +39-064-9693-268 (A.M. & D.R.); +39-081-450-169 (A.N. & L.A.)
| | - Angela Nebbioso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
- Correspondence: (A.M.); (D.R.); (A.N.); (L.A.); Tel.: +39-0649-913-392 (A.M.); +39-0649-913-891 (D.R.); +39-0815-665-682 (A.N.); +39-0815-667-569 (L.A.); Fax: +39-064-9693-268 (A.M. & D.R.); +39-081-450-169 (A.N. & L.A.)
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.C.); (A.P.); (L.D.T.); (P.G.); (M.D.D.); (E.S.); (Z.Y.); (G.C.)
- Correspondence: (A.M.); (D.R.); (A.N.); (L.A.); Tel.: +39-0649-913-392 (A.M.); +39-0649-913-891 (D.R.); +39-0815-665-682 (A.N.); +39-0815-667-569 (L.A.); Fax: +39-064-9693-268 (A.M. & D.R.); +39-081-450-169 (A.N. & L.A.)
| |
Collapse
|
11
|
Wang T, Xu Z, Lu Y, Shi J, Liu W, Zhang C, Jiang Z, Qi B, Bai L. Recent Progress on the Discovery of Sirt2 Inhibitors for the Treatment of Various Cancers. Curr Top Med Chem 2019; 19:1051-1058. [PMID: 31074370 DOI: 10.2174/1568026619666190510103416] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/27/2019] [Accepted: 04/06/2019] [Indexed: 01/14/2023]
Abstract
Sirtuins family is a class of NDA+ dependent protein deacetylases that play a key role in the
regulation of several aspects of biological processes, such as cell cycle regulation, autophagy, immune
and inflammatory response. Many studies have shown that sirtuins2 as a key player in the cancer pathway
is of great significance in tumorigenesis. This review summarizes the newly discovered, in recent
years, some SIRT2 inhibitors for cancer target structure, action mechanism, biological activity, substrate
specificity, and signaling pathways.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhuyu Xu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yongping Lu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenbo Liu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, China
| | - Chengchen Zhang
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, Florida, United States
| | - Baowen Qi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
12
|
Zhu J, Wang R, Xu T, Zhang S, Zhao Y, Li Z, Wang C, Zhou J, Gao D, Hu Y, Tian X, Yao J. Salvianolic Acid A Attenuates Endoplasmic Reticulum Stress and Protects Against Cholestasis-Induced Liver Fibrosis via the SIRT1/HSF1 Pathway. Front Pharmacol 2018; 9:1277. [PMID: 30455644 PMCID: PMC6230567 DOI: 10.3389/fphar.2018.01277] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/18/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Endoplasmic reticulum stress (ER stress) plays a critical role in the pathogenesis of liver fibrosis; thus, it can be a potential therapeutic target of fibrosis. However, the mechanism of ER stress regulation in fibrosis, particularly through sirtuin 1 (SIRT1), remains unclear. The objective of this study was to investigate the effect of SIRT1-mediated inhibition of ER stress in bile duct ligation (BDL)-induced liver fibrosis, and to explore the effect of salvianolic acid A (SalA) on BDL-induced liver fibrosis through SIRT1/heat shock factor 1 (HSF1) signaling. Materials and Methods: We explored the effects of SalA on liver fibrosis and ER stress in BDL-induced liver fibrosis in rats and the human hepatic stellate cell line LX2 cells. The LX2 cells were treated with 20 ng of platelet-derived growth factor-BB homodimer (PDGF-BB) for 24 h, and then incubated in the absence or presence of SalA (25 μM) for 24 h. Results: In vivo, SalA treatment alleviated BDL-induced liver injury and ER stress. Importantly, SalA treatment increased HSF1 expression and activity using a SIRT1-dependent mechanism. In LX2 cells, PDGF-BB induced ER stress and fibrosis were blocked by HSF1 overexpression. Furthermore, SIRT1 siRNA abrogated the SalA-mediated promotion of HSF1 deacetylation and expression, suggesting that SalA-mediated protection occurs by SIRT1 targeting HSF1 for deacetylation. Conclusion: This is the first study to identify the SIRT1/HSF1 pathway as a key therapeutic target for controlling BDL-induced liver fibrosis and to show that SalA confers protection against BDL- and PDGF-BB-induced hepatic fibrosis and ER stress through SIRT1-mediated HSF1 deacetylation.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Ruiwen Wang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Ting Xu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Shuai Zhang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Zhenlu Li
- Department of General Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chao Wang
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Junjun Zhou
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Dongyan Gao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yan Hu
- Department of Pharmacy, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaofeng Tian
- Department of General Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| |
Collapse
|
13
|
Anticancer activities of a benzimidazole compound through sirtuin inhibition in colorectal cancer. Future Med Chem 2018; 10:2039-2057. [DOI: 10.4155/fmc-2018-0052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: This study aims to investigate the mode of action of a novel sirtuin inhibitor (BZD9L1) and its associated molecular pathways in colorectal cancer (CRC) cells. Materials & methods: BZD9L1 was tested against metastatic CRC cell lines to evaluate cytotoxicity, cell cycle and apoptosis, senescence, apoptosis related genes and protein expressions, as well as effect against major cancer signaling pathways. Results & conclusion: BZD9L1 reduced the viability, cell migration and colony forming ability of both HCT 116 and HT-29 metastatic CRC cell lines through apoptosis. BZD9L1 regulated major cancer pathways differently in CRC with different mutation profiles. BZD9L1 exhibited anticancer activities as a cytotoxic drug in CRC and as a promising therapeutic strategy in CRC treatment.
Collapse
|
14
|
Yuan Q, Zhang D, Liu C, Zhang C, Yuan D. Chikusetsusaponin V Inhibits LPS-Activated Inflammatory Responses via SIRT1/NF-κB Signaling Pathway in RAW264.7 Cells. Inflammation 2018; 41:2149-2159. [DOI: 10.1007/s10753-018-0858-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
15
|
Ma W, Zhao X, Wang K, Liu J, Huang G. Dichloroacetic acid (DCA) synergizes with the SIRT2 inhibitor Sirtinol and AGK2 to enhance anti-tumor efficacy in non-small cell lung cancer. Cancer Biol Ther 2018; 19:835-846. [PMID: 30067423 DOI: 10.1080/15384047.2018.1480281] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Combination chemotherapy is a potentially promising approach to enhance anticancer activity, overcome drug resistance, and improve disease-free and overall survival. The current study investigates the antitumor activity of sodium dichloroacetic acid (DCA) in combination with SIRT2 inhibitor Sirtinol and AGK2. We found that combining DCA with Sirtinol produced a synergistic therapeutic benefit in A549 and H1299 NSCLC cells in vitro and in a mouse A549 xenograft model. Synergistic potentiation of oxidative phosphorylation (OXPHOS) was observed, including decreased glucose consumption, decreased lactate production, increased OCR and increased ROS generation, possibly via co-targeting pyruvate dehydrogenase alpha 1(PDHA1). Mechanically, AGK2 and Sirtinol were found to increase the lysine-acetylation and decrease the serine-phosphorylation of PDHA1, which enabled the two inhibitors to synergize with DCA to further activate PDHA1. Besides, a AMPKα-ROS feed-forward loop was notably activated after the combined treatments compared with mono-therapy. Our results indicate that the combination of DCA and SIRT2 inhibitor may provide a promising therapeutic strategy to effectively kill cancer cells.
Collapse
Affiliation(s)
- Wenjing Ma
- a The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences , Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine , Shanghai , China.,c Shanghai Key Laboratory for Molecular Imaging , Shanghai University of Medicine and Health Sciences , Shanghai , China
| | - Xiaoping Zhao
- b Department of Nuclear Medicine, Renji Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Kaiying Wang
- a The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences , Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Jianjun Liu
- b Department of Nuclear Medicine, Renji Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Gang Huang
- a The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences , Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine , Shanghai , China.,b Department of Nuclear Medicine, Renji Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China.,c Shanghai Key Laboratory for Molecular Imaging , Shanghai University of Medicine and Health Sciences , Shanghai , China
| |
Collapse
|
16
|
Inhibition of sirtuins 1 and 2 impairs cell survival and migration and modulates the expression of P-glycoprotein and MRP3 in hepatocellular carcinoma cell lines. Toxicol Lett 2018; 289:63-74. [PMID: 29545174 DOI: 10.1016/j.toxlet.2018.03.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 02/26/2018] [Accepted: 03/10/2018] [Indexed: 01/15/2023]
Abstract
Sirtuins (SIRTs) 1 and 2 deacetylases are overexpressed in hepatocellular carcinoma (HCC) and are associated with tumoral progression and multidrug resistance (MDR). In this study we analyzed whether SIRTs 1 and 2 activities blockage was able to affect cellular survival and migration and to modulate p53 and FoxO1 acetylation in HepG2 and Huh7 cells. Moreover, we analyzed ABC transporters P-glycoprotein (P-gp) and multidrug resistance-associated protein 3 (MRP3) expression. We used cambinol and EX-527 as SIRTs inhibitors. Both drugs reduced cellular viability, number of colonies and cellular migration and augmented apoptosis. In 3D cultures, SIRTs inhibitors diminished spheroid growth and viability. 3D culture was less sensitive to drugs than 2D culture. The levels of acetylated p53 and FoxO1 increased after treatments. Drugs induced a decrease in ABC transporters mRNA and protein levels in HepG2 cells; however, only EX-527 was able to reduce MRP3 mRNA and protein levels in Huh7 cells. This is the first work demonstrating the regulation of MRP3 by SIRTs. In conclusion, both drugs decreased HCC cells survival and migration, suggesting SIRTs 1 and 2 activities blockage could be beneficial during HCC therapy. Downregulation of the expression of P-gp and MRP3 supports the potential application of SIRTs 1 and 2 inhibitions in combination with conventional chemotherapy.
Collapse
|
17
|
The Fungal Metabolite Eurochevalierine, a Sequiterpene Alkaloid, Displays Anti-Cancer Properties through Selective Sirtuin 1/2 Inhibition. Molecules 2018; 23:molecules23020333. [PMID: 29401749 PMCID: PMC6017873 DOI: 10.3390/molecules23020333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 01/19/2023] Open
Abstract
NAD+-dependent histone deacetylases (sirtuins) are implicated in cellular processes such as proliferation, DNA repair, and apoptosis by regulating gene expression and the functions of numerous proteins. Due to their key role in cells, the discovery of small molecule sirtuin modulators has been of significant interest for diverse therapeutic applications. In particular, it has been shown that inhibition of sirtuin 1 and 2 activities is beneficial for cancer treatment. Here, we demonstrate that the fungal metabolite eurochevalierine from the fungus Neosartorya pseudofischeri inhibits sirtuin 1 and 2 activities (IC50 about 10 µM) without affecting sirtuin 3 activity. The binding modes of the eurochevalierine for sirtuin 1 and 2 have been identified through computational docking analyses. Accordingly, this sequiterpene alkaloid induces histone H4 and α-tubulin acetylation in various cancer cell models in which it induces strong cytostatic effects without affecting significantly the viability of healthy PBMCs. Importantly, eurochevalierine targets preferentially cancer cell proliferation (selectivity factor ≫ 7), as normal human primary CD34+ stem/progenitor cells were less affected by the treatment. Finally, eurochevalierine displays suitable drug-likeness parameters and therefore represent a promising scaffold for lead molecule optimization to study the mechanism and biological roles of sirtuins and potentially a basis for development into therapeutics.
Collapse
|
18
|
Huang C, Wu J, Xu L, Wang J, Chen Z, Yang R. Regulation of HSF1 protein stabilization: An updated review. Eur J Pharmacol 2018; 822:69-77. [PMID: 29341886 DOI: 10.1016/j.ejphar.2018.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/11/2017] [Accepted: 01/09/2018] [Indexed: 12/12/2022]
Abstract
Heat shock factor 1 (HSF1) is a transcriptional factor that determines the efficiency of heat shock responses (HSRs) in the cell. Given its function has been extensively studied in recent years, HSF1 is considered a potential target for the treatment of disorders associated with protein aggregation. The activity of HSF1 is traditionally regulated at the transcriptional level in which the transactivation domain of HSF1 is modified by extensive array of pos-translational modifications, such as phosphorylation, sumoylation, and acetylation. Recently, HSF1 is also reported to be regulated at the monomeric level. For example, in neurodegenerative disorders such as Huntington's disease and Alzheimer's disease the expression levels of the monomeric HSF1 are found to be reduced markedly. Methylene blue (MB) and riluzole, two clinical available drugs, increase the amount of the monomeric HSF1 in both cells and animals. Since the monomeric HSF1 not only determines the efficiency of HSRs, but exerts protective effects in a trimerization-independent manner, increasing the amount of the monomeric HSF1 via stabilization of HSF1 may be an alternative strategy for the amplification of HSR. However, to date we have no outlined knowledges about HSF1 protein stabilization, though studies regarding the regulation of the monomeric HSF1 have been documented in recent years. Here, we summarize the regulation of the monomeric HSF1 by some previously reported factors, such as synuclein, Huntingtin (Htt), TDP-43, unfolded protein response (UPR), MB and doxorubicin (DOX), as well as their possible mechanisms, aiming to push the understanding about HSF1 protein stabilization.
Collapse
Affiliation(s)
- Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Li Xu
- Department of Ultrasound, Danyang People's Hospital, #2 Xinmin Western Road, Danyang 212300, Jiangsu, China
| | - Jili Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, # 6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20Xisi Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
19
|
Qian Y, Xin Z, Lv Y, Wang Z, Zuo L, Huang X, Li Y, Xin HB. Asiatic acid suppresses neuroinflammation in BV2 microgliaviamodulation of the Sirt1/NF-κB signaling pathway. Food Funct 2018; 9:1048-1057. [DOI: 10.1039/c7fo01442b] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Asiatic acid promotes Sirt1 expression and inhibits NF-κB-induced microglia activation.
Collapse
Affiliation(s)
- Yisong Qian
- Institute of Translational Medicine
- Nanchang University
- Nanchang 330031
- PR China
| | - Zhaochen Xin
- Institute of Translational Medicine
- Nanchang University
- Nanchang 330031
- PR China
| | - Yanni Lv
- Department of Pharmacy
- The First Affiliated Hospital of Nanchang University
- Nanchang 330006
- China
| | - Ziwei Wang
- Institute of Translational Medicine
- Nanchang University
- Nanchang 330031
- PR China
| | - Li Zuo
- Institute of Translational Medicine
- Nanchang University
- Nanchang 330031
- PR China
| | - Xiang Huang
- Institute of Translational Medicine
- Nanchang University
- Nanchang 330031
- PR China
| | - Yunman Li
- Department of Physiology
- China Pharmaceutical University
- Nanjing 210009
- PR China
| | - Hong-Bo Xin
- Institute of Translational Medicine
- Nanchang University
- Nanchang 330031
- PR China
| |
Collapse
|
20
|
Schnekenburger M, Goffin E, Lee JY, Jang JY, Mazumder A, Ji S, Rogister B, Bouider N, Lefranc F, Miklos W, Mathieu V, de Tullio P, Kim KW, Dicato M, Berger W, Han BW, Kiss R, Pirotte B, Diederich M. Discovery and Characterization of R/S-N-3-Cyanophenyl-N'-(6-tert-butoxycarbonylamino-3,4-dihydro-2,2-dimethyl-2H-1-benzopyran-4-yl)urea, a New Histone Deacetylase Class III Inhibitor Exerting Antiproliferative Activity against Cancer Cell Lines. J Med Chem 2017; 60:4714-4733. [PMID: 28475330 DOI: 10.1021/acs.jmedchem.7b00533] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A new series of N-aryl-N'-3,4-dihydro-2,2-dimethyl-2H-1-benzopyran-4-yl)ureas bearing an alkoxycarbonylamino group at the 6-position were synthesized and examined as putative anticancer agents targeting sirtuins in glioma cells. On the basis of computational docking combined to in vitro sirtuin 1/2 inhibition assays, we selected compound 18 [R/S-N-3-cyanophenyl-N'-(6-tert-butoxycarbonylamino-3,4-dihydro-2,2-dimethyl-2H-1-benzopyran-4-yl)urea] which displays a potent antiproliferative activity on various glioma cell types, assessed by quantitative videomicroscopy, eventually triggering senescence. The impact on normal glial cells was lower with a selectivity index of >10. Furthermore, human U373 and Hs683 glioblastoma cell lines served to demonstrate the inhibitory activity of 18 against histone deacetylase (HDAC) class III sirtuins 1 and 2 (SIRT1/2) by quantifying acetylation levels of histone and non-histone proteins. The translational potential of 18 was validated by an NCI-60 cell line screen and validation of growth inhibition of drug resistant cancer cell models. Eventually, the anticancer potential of 18 was validated in 3D glioblastoma spheroids and in vivo by zebrafish xenografts. In summary, compound 18 is the first representative of a new class of SIRT inhibitors opening new perspectives in the medicinal chemistry of HDAC inhibitors.
Collapse
Affiliation(s)
- Michael Schnekenburger
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg , 9, Rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Eric Goffin
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège , 4000 Liège, Belgium
| | - Jin-Young Lee
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | - Jun Young Jang
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | - Aloran Mazumder
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | - Seungwon Ji
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | - Bernard Rogister
- Nervous System Diseases and Treatment, GIGA-Neurosciences, University of Liège , 4000 Liège, Belgium
| | - Nafila Bouider
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège , 4000 Liège, Belgium
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Walter Miklos
- Department of Medicine I, Comprehensive Cancer Center and Institute of Cancer Research, Medical University of Vienna , 1090 Vienna, Austria
| | - Véronique Mathieu
- Laboratoire de Cancérologie et de Toxicologie Expérimentale, Faculté de Pharmacie, Université Libre de Bruxelles , 1050 Brussels, Belgium
| | - Pascal de Tullio
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège , 4000 Liège, Belgium
| | - Kyu-Won Kim
- SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University , Seoul 151-742, Korea
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg , 9, Rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Walter Berger
- Department of Medicine I, Comprehensive Cancer Center and Institute of Cancer Research, Medical University of Vienna , 1090 Vienna, Austria
| | - Byung Woo Han
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | - Robert Kiss
- Laboratoire de Cancérologie et de Toxicologie Expérimentale, Faculté de Pharmacie, Université Libre de Bruxelles , 1050 Brussels, Belgium
| | - Bernard Pirotte
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège , 4000 Liège, Belgium
| | - Marc Diederich
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| |
Collapse
|