1
|
Liu P, Chen Z, Guo Y, He Q, Pan C. Recent advances in small molecule inhibitors of deubiquitinating enzymes. Eur J Med Chem 2025; 287:117324. [PMID: 39908798 DOI: 10.1016/j.ejmech.2025.117324] [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: 10/15/2024] [Revised: 12/24/2024] [Accepted: 01/23/2025] [Indexed: 02/07/2025]
Abstract
Proteins play a pivotal role in maintaining cellular homeostasis. Their degradation primarily orchestrated through the ubiquitin-proteasome system (UPS) and cellular autophagy. Dysfunction of the UPS is associated with various human diseases, including cancer, autoimmune disorders, and neurodegenerative conditions. Consequently, the UPS has emerged as a promising therapeutic target. Deubiquitinases (DUBs) have garnered significant attention as potential targets for therapeutic intervention due to their role in modulating protein stability and function. This review focuses on recent advancements of DUBs, particularly their relevance in the UPS and their potential as drug targets. Notably, inhibitors targeting specific DUBs, such as USP1, USP7, USP14, and USP30 have shown promise in preclinical and clinical studies for cancer therapy. Additionally, DUB inhibitors have been involved in novel therapeutic approaches lately, including as targets for proteolysis-targeting chimeras (PROTACs) or as tools in deubiquitinase-targeting chimeras (DUBTACs).
Collapse
Affiliation(s)
- Pengwei Liu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China
| | - Zhengyang Chen
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China
| | - Yiting Guo
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China.
| | - Chenghao Pan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China.
| |
Collapse
|
2
|
Ziemniak M, Trzybiński D, Pawlędzio S, Filipowicz G, Pająk-Tarnacka B, Priebe W, Woźniak K. Crystal structure and polymorphic forms of auranofin revisited. RSC Adv 2025; 15:10378-10389. [PMID: 40182499 PMCID: PMC11967170 DOI: 10.1039/d5ra00196j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Auranofin, initially developed as a treatment for rheumatoid arthritis, is currently under extensive investigation as a potential drug for various conditions, including cancer, bacterial infections, and parasitic infections. The compound is a known inhibitor of thioredoxin reductase (TXNRD1) and related selenoproteins. Although preliminary studies on the auranofin crystal polymorphism exist, and a low-quality crystal structure has been reported, a comprehensive crystallographic characterization remains unexplored. Utilizing X-ray crystallography techniques, we conducted detailed structural analysis of auranofin and compared our findings with related organogold compounds. Implementation of Hirshfeld atom refinement (HAR) enabled a more accurate hydrogen atom positioning in the structure. The crystal packing reveals a layered arrangement stabilised by numerous weak hydrogen bonds and dispersive interactions. Notably, our attempts to reproduce the previously reported polymorphic form of auranofin, purportedly more water-soluble, were unsuccessful despite following published protocols. To our knowledge, this is the first study describing a "disappearing polymorph" phenomenon of any pharmaceutically relevant transition metal coordination compound. Our findings may have significant implications for medicinal chemistry and pharmacology of coordination complexes, suggesting the need for systematic revision of historical crystallographic data in this field.
Collapse
Affiliation(s)
- Marcin Ziemniak
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw Żwirki i Wigury 101 Warsaw 02-089 Poland
| | - Damian Trzybiński
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw Żwirki i Wigury 101 Warsaw 02-089 Poland
| | - Sylwia Pawlędzio
- Neutron Scattering Division, Oak Ridge National Laboratory Oak Ridge Tennessee 37831 USA
| | - Gabriela Filipowicz
- Pritzker School of Molecular Engineering, The University of Chicago Chicago Illinois 60637 USA
| | - Beata Pająk-Tarnacka
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw Żwirki i Wigury 101 Warsaw 02-089 Poland
- WPD Pharmaceuticals Inc Żwirki I Wigury 101 Warsaw 02-089 Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center 1901 East Rd. Houston TX 77054 USA
| | - Krzysztof Woźniak
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw Żwirki i Wigury 101 Warsaw 02-089 Poland
| |
Collapse
|
3
|
Chmelyuk N, Kordyukova M, Sorokina M, Sinyavskiy S, Meshcheryakova V, Belousov V, Abakumova T. Inhibition of Thioredoxin-Reductase by Auranofin as a Pro-Oxidant Anticancer Strategy for Glioblastoma: In Vitro and In Vivo Studies. Int J Mol Sci 2025; 26:2084. [PMID: 40076706 PMCID: PMC11900239 DOI: 10.3390/ijms26052084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Reactive oxygen species (ROS) play a key role in cancer progression and antitumor therapy. Glioblastoma is a highly heterogeneous tumor with different cell populations exhibiting various redox statuses. Elevated ROS levels in cancer cells promote tumor growth and simultaneously make them more sensitive to anticancer drugs, but further elevation leads to cell death and apoptosis. Meanwhile, various subsets of tumor cells, such a glioblastoma stem cells (GSC) or the cells in tumor microenvironment (TME), demonstrate adaptive mechanisms to excessive ROS production by developing effective antioxidant systems such as glutathione- and thioredoxin-dependent. GSCs demonstrate higher chemoresistance and lower ROS levels than other glioma cells, while TME cells create a pro-oxidative environment and have immunosuppressive effects. Both subpopulations have become an attractive target for developing therapies. Increased expression of thioredoxin reductase (TrxR) is often associated with tumor progression and poor patient survival. Various TrxR inhibitors have been investigated as potential anticancer therapies, including nitrosoureas, flavonoids and metallic complexes. Gold derivatives are irreversible inhibitors of TrxR. Among them, auranofin (AF), a selective TrxR inhibitor, has proven its effectiveness as a drug for the treatment of rheumatoid arthritis and its efficacy as an anticancer agent has been demonstrated in preclinical studies in vitro and in vivo. However, further clinical application of AF could be challenging due to the low solubility and insufficient delivery to glioblastoma. Different delivery strategies for hydrophobic drugs could be used to increase the concentration of AF in the brain. Combining different therapeutic approaches that affect the redox status of various glioma cell populations could become a new strategy for treating brain tumor diseases.
Collapse
Affiliation(s)
- Nelly Chmelyuk
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Laboratory of Biomedical nanomaterials, National Research Technological University “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
| | - Maria Kordyukova
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Maria Sorokina
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Semyon Sinyavskiy
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Valeriya Meshcheryakova
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod Belousov
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Tatiana Abakumova
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
4
|
Bakkar M, Khalil S, Bhayekar K, Kushwaha ND, Samarbakhsh A, Dorandish S, Edwards H, Dou QP, Ge Y, Gavande NS. Ubiquitin-Specific Protease Inhibitors for Cancer Therapy: Recent Advances and Future Prospects. Biomolecules 2025; 15:240. [PMID: 40001543 PMCID: PMC11853158 DOI: 10.3390/biom15020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer management has traditionally depended on chemotherapy as the mainstay of treatment; however, recent advancements in targeted therapies and immunotherapies have offered new options. Ubiquitin-specific proteases (USPs) have emerged as promising therapeutic targets in cancer treatment due to their crucial roles in regulating protein homeostasis and various essential cellular processes. This review covers the following: (1) the structural and functional characteristics of USPs, highlighting their involvement in key cancer-related pathways, and (2) the discovery, chemical structures, mechanisms of action, and potential clinical implications of USP inhibitors in cancer therapy. Particular attention is given to the role of USP inhibitors in enhancing cancer immunotherapy, e.g., modulation of the tumor microenvironment, effect on regulatory T cell function, and influence on immune checkpoint pathways. Furthermore, this review summarizes the current progress and challenges of clinical trials involving USP inhibitors as cancer therapy. We also discuss the complexities of achieving target selectivity, the ongoing efforts to develop more specific and potent USP inhibitors, and the potential of USP inhibitors to overcome drug resistance and synergize with existing cancer treatments. We finally provide a perspective on future directions in targeting USPs, including the potential for personalized medicine based on specific gene mutations, underscoring their significant potential for enhancing cancer treatment. By elucidating their mechanisms of action, clinical progress, and potential future applications, we hope that this review could serve as a useful resource for both basic scientists and clinicians in the field of cancer therapeutics.
Collapse
Affiliation(s)
- Mohamad Bakkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Division of Pediatric Hematology and Oncology, Children’s Hospital of Michigan, Detroit, MI 48201, USA
| | - Sara Khalil
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
| | - Komal Bhayekar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Narva Deshwar Kushwaha
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Amirreza Samarbakhsh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Sadaf Dorandish
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Q. Ping Dou
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Navnath S. Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
5
|
Arojojoye AS, Awuah SG. Functional utility of gold complexes with phosphorus donor ligands in biological systems. Coord Chem Rev 2025; 522:216208. [PMID: 39552640 PMCID: PMC11563041 DOI: 10.1016/j.ccr.2024.216208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Metallo-phosphines are ubiquitous in organometallic chemistry with widespread applications as catalysts in various chemical transformations, precursors for organic electronics, and chemotherapeutic agents or chemical probes. Here, we provide a comprehensive review of the exploration of the current biological applications of Au complexes bearing phosphine donor ligands. The goal is to deepen our understanding of the synthetic utility and reactivity of Au-phosphine complexes to provide insights that could lead to the design of new molecules and enhance the cross-application or repurposing of these complexes.
Collapse
Affiliation(s)
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington KY 40506
- Center for Pharmaceutical Research and Innovation and Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington KY 40536
- Markey Cancer Centre, University of Kentucky, Lexington KY, 40536
- University of Kentucky Bioelectronics and Nanomedicine Research Center, Lexington, Kentucky 40506, United States
| |
Collapse
|
6
|
Gilpatrick ST, Obisesan OA, Parkin S, Awuah SG. Carbon-phosphorus stapled Au(I) anticancer agents via bisphosphine induced reductive elimination. Dalton Trans 2024; 53:18974-18982. [PMID: 39161271 PMCID: PMC11333934 DOI: 10.1039/d4dt01929f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Towards the goal of generating new stabilized gold complexes as potent anticancer agents, we report here a novel class of Au(I) agents from Au(III)-mediated Caryl-P bond formation captured within the same complex by reacting a C^N cyclometalated Au(III) complex with bisphosphines. Cyclometalated Au(III) complexes of the type [Au(C^N)Cl2], where C^N represent different aryl pyridine framework reacted with bis(2-diphenylphosphino)phenyl ether in refluxing methanol to access an unsymmetrical gold complex featuring C-P coupling and Au(I)-phosphine. The complexes were characterized by 1H-NMR, 13C-NMR, and 31P-NMR and mass spectrometry. The structures of the complexes were characterized by X-ray crystallography and purity ascertained by HPLC and elemental analysis. The complexes demonstrate promising anticancer activity in a broad panel of cancer cell lines of different tumor origin. Mechanistically, the complexes induce apoptosis, generate mitochondrial ROS, depolarize mitochondrial membrane potential and modulate mitochondrial respiration in cancer cells. Overall, we developed a new structural class of Au(I) complexes with promising anticancer potential with potential utility in other applications.
Collapse
Affiliation(s)
- Sean T Gilpatrick
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA.
| | | | - Sean Parkin
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA.
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA.
- Center for Pharmaceutical Research and Innovation and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Centre, University of Kentucky, Lexington, KY, 40536, USA
- Center for Bioelectronics and Nanomedicine, University of Kentucky, Lexington, KY, 40506, USA
| |
Collapse
|
7
|
Nogami A, Amemiya HJ, Fujiwara H, Umezawa Y, Tohda S, Nagao T. Targeting USP14/UCHL5: A Breakthrough Approach to Overcoming Treatment-Resistant FLT3-ITD-Positive AML. Int J Mol Sci 2024; 25:10372. [PMID: 39408703 PMCID: PMC11476563 DOI: 10.3390/ijms251910372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations in acute myeloid leukemia (AML) are associated with poor prognosis and therapy resistance. This study aimed to demonstrate that inhibiting the deubiquitinating enzymes ubiquitin-specific peptidase 14 (USP14) and ubiquitin C-terminal hydrolase L5 (UCHL5) (USP14/UCHL5) with b-AP15 or the organogold compound auranofin (AUR) induces apoptosis in the ITD-transformed human leukemia cell line MV4-11 and mononuclear leukocytes derived from patients with FLT3-ITD-positive AML. This study included patients diagnosed with AML at Tokyo Medical and Dental University Hospital between January 2018 and July 2024. Both treatments blocked downstream FLT3 pathway events, with the effects potentiated by USP14 knockdown. Both treatments inhibited FLT3 deubiquitination via K48 and disrupted translation initiation via 4EBP1, a downstream FLT3 target. FLT3 was downregulated in the leukemic cells, with the associated activation of stress-related MAP kinase pathways and increased NF-E2-related factor 2. Furthermore, the overexpression of B-cell lymphoma-extra-large and myeloid cell leukemia-1 prevented the cell death caused by b-AP15 and AUR. These results suggest that inhibiting USP14/UCHL5, which involves multiple regulatory mechanisms, is a promising target for novel therapies for treatment-resistant FLT3-ITD-positive AML.
Collapse
MESH Headings
- Humans
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Ubiquitin Thiolesterase/metabolism
- Ubiquitin Thiolesterase/genetics
- Ubiquitin Thiolesterase/antagonists & inhibitors
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Male
- Cell Line, Tumor
- Middle Aged
- Aged
- Apoptosis/drug effects
- Adult
- Mutation
Collapse
Affiliation(s)
- Ayako Nogami
- Department of Laboratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyoku, Tokyo 113-8510, Japan
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Hideki Jose Amemiya
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Hiroki Fujiwara
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Yoshihiro Umezawa
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Shuji Tohda
- Department of Laboratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyoku, Tokyo 113-8510, Japan
| | - Toshikage Nagao
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| |
Collapse
|
8
|
Rodríguez-Enríquez S, Robledo-Cadena DX, Pacheco-Velázquez SC, Vargas-Navarro JL, Padilla-Flores JA, Kaambre T, Moreno-Sánchez R. Repurposing auranofin and meclofenamic acid as energy-metabolism inhibitors and anti-cancer drugs. PLoS One 2024; 19:e0309331. [PMID: 39288141 PMCID: PMC11407620 DOI: 10.1371/journal.pone.0309331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE Cytotoxicity of the antirheumatic drug auranofin (Aur) and the non-steroidal anti-inflammatory drug meclofenamic acid (MA) on several cancer cell lines and isolated mitochondria was examined to assess whether these drugs behave as oxidative phosphorylation inhibitors. METHODS The effect of Aur or MA for 24 h was assayed on metastatic cancer and non-cancer cell proliferation, energy metabolism, mitophagy and metastasis; as well as on oxygen consumption rates of cancer and non-cancer mitochondria. RESULTS Aur doses in the low micromolar range were required to decrease proliferation of metastatic HeLa and MDA-MB-231 cells, whereas one or two orders of magnitude higher levels were required to affect proliferation of non-cancer cells. MA doses required to affect cancer cell growth were one order of magnitude higher than those of Aur. At the same doses, Aur impaired oxidative phosphorylation in isolated mitochondria and intact cells through mitophagy induction, as well as glycolysis. Consequently, cell migration and invasiveness were severely affected. The combination of Aur with very low cisplatin concentrations promoted that the effects on cellular functions were potentiated. CONCLUSION Aur surges as a highly promising anticancer drug, suggesting that efforts to establish this drug in the clinical treatment protocols are warranted and worthy to undertake.
Collapse
Affiliation(s)
- Sara Rodríguez-Enríquez
- Laboratorio de Control Metabólico, Carrera de Médico Cirujano de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
| | | | - Silvia Cecilia Pacheco-Velázquez
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jorge Luis Vargas-Navarro
- Laboratorio de Control Metabólico, Carrera de Médico Cirujano de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
- Laboratorio de Control Metabólico, Carrera de Biología de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
| | - Joaquín Alberto Padilla-Flores
- Laboratorio de Control Metabólico, Carrera de Médico Cirujano de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
- Laboratorio de Control Metabólico, Carrera de Biología de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Rafael Moreno-Sánchez
- Laboratorio de Control Metabólico, Carrera de Biología de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| |
Collapse
|
9
|
Hei Z, Yang S, Ouyang G, Hanna J, Lepoivre M, Huynh T, Aguinaga L, Cassinat B, Maslah N, Bourge M, Golinelli-Cohen MP, Guittet O, Vallières C, Vernis L, Fenaux P, Huang ME. Targeting the redox vulnerability of acute myeloid leukaemia cells with a combination of auranofin and vitamin C. Br J Haematol 2024; 205:1017-1030. [PMID: 39087522 DOI: 10.1111/bjh.19680] [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: 05/07/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by complex molecular and cytogenetic abnormalities. Pro-oxidant cellular redox status is a common hallmark of AML cells, providing a rationale for redox-based anticancer strategy. We previously discovered that auranofin (AUF), initially used for the treatment of rheumatoid arthritis and repositioned for its anticancer activity, can synergize with a pharmacological concentration of vitamin C (VC) against breast cancer cell line models. In this study, we observed that this drug combination synergistically and efficiently killed cells of leukaemic cell lines established from different myeloid subtypes. In addition to an induced elevation of reactive oxygen species and ATP depletion, a rapid dephosphorylation of 4E-BP1 and p70S6K, together with a strong inhibition of protein synthesis were early events in response to AUF/VC treatment, suggesting their implication in AUF/VC-induced cytotoxicity. Importantly, a study on 22 primary AML specimens from various AML subtypes showed that AUF/VC combinations at pharmacologically achievable concentrations were effective to eradicate primary leukaemic CD34+ cells from the majority of these samples, while being less toxic to normal cord blood CD34+ cells. Our findings indicate that targeting the redox vulnerability of AML with AUF/VC combinations could present a potential anti-AML therapeutic approach.
Collapse
Affiliation(s)
- Zhiliang Hei
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Shujun Yang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guifang Ouyang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jolimar Hanna
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Michel Lepoivre
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Tony Huynh
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Lorea Aguinaga
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Bruno Cassinat
- INSERM UMR 1131, Université Paris Cité, Hôpital Saint-Louis, IRSL, Paris, France
| | - Nabih Maslah
- INSERM UMR 1131, Université Paris Cité, Hôpital Saint-Louis, IRSL, Paris, France
| | - Mickaël Bourge
- Cytometry Facility, Imagerie-Gif, Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | | | - Olivier Guittet
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Cindy Vallières
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Laurence Vernis
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Pierre Fenaux
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Meng-Er Huang
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| |
Collapse
|
10
|
An X, Yu W, Liu J, Tang D, Yang L, Chen X. Oxidative cell death in cancer: mechanisms and therapeutic opportunities. Cell Death Dis 2024; 15:556. [PMID: 39090114 PMCID: PMC11294602 DOI: 10.1038/s41419-024-06939-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen-containing molecules generated as natural byproducts during cellular processes, including metabolism. Under normal conditions, ROS play crucial roles in diverse cellular functions, including cell signaling and immune responses. However, a disturbance in the balance between ROS production and cellular antioxidant defenses can lead to an excessive ROS buildup, causing oxidative stress. This stress damages essential cellular components, including lipids, proteins, and DNA, potentially culminating in oxidative cell death. This form of cell death can take various forms, such as ferroptosis, apoptosis, necroptosis, pyroptosis, paraptosis, parthanatos, and oxeiptosis, each displaying distinct genetic, biochemical, and signaling characteristics. The investigation of oxidative cell death holds promise for the development of pharmacological agents that are used to prevent tumorigenesis or treat established cancer. Specifically, targeting key antioxidant proteins, such as SLC7A11, GCLC, GPX4, TXN, and TXNRD, represents an emerging approach for inducing oxidative cell death in cancer cells. This review provides a comprehensive summary of recent progress, opportunities, and challenges in targeting oxidative cell death for cancer therapy.
Collapse
Affiliation(s)
- Xiaoqin An
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Yu
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Li Yang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China.
| | - Xin Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
11
|
Vitali V, Massai L, Messori L. Strategies for the design of analogs of auranofin endowed with anticancer potential. Expert Opin Drug Discov 2024; 19:855-867. [PMID: 38803122 DOI: 10.1080/17460441.2024.2355329] [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: 03/06/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Auranofin (AF) is a well-established, FDA-approved, antiarthritic gold drug that is currently being reevaluated for a variety of therapeutic indications through drug repurposing. AF has shown great promise as a potential anticancer agent and has been approved for a few clinical trials in cancer. The renewed interest in AF has led to extensive research into the design, preparation and biological evaluation of auranofin analogs, which may have an even better pharmacological profile than the parent drug. AREAS COVERED This article reviews the strategies for chemical modification of the AF scaffold. Several auranofin analogs have been prepared and characterized for medical application in the field of cancer treatment over the last 20 years. Some emerging structure-function relationships are proposed and discussed. EXPERT OPINION The chemical modification of the AF scaffold has been the subject of intense activity in recent years and this strategy has led to the preparation and evaluation of several AF analogs. The case of iodauranofin is a particularly promising example. The availability of homogeneous biological data for a group of AF derivatives allows some initial structure-function relationships to be proposed, which may inspire the design and synthesis of new and better AF analogs for cancer treatment.
Collapse
Affiliation(s)
- Valentina Vitali
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Lara Massai
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
12
|
Martinez-Jaramillo E, Jamali F, Abdalbari FH, Abdulkarim B, Jean-Claude BJ, Telleria CM, Sabri S. Pro-Oxidant Auranofin and Glutathione-Depleting Combination Unveils Synergistic Lethality in Glioblastoma Cells with Aberrant Epidermal Growth Factor Receptor Expression. Cancers (Basel) 2024; 16:2319. [PMID: 39001381 PMCID: PMC11240359 DOI: 10.3390/cancers16132319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Glioblastoma (GBM) is the most prevalent and advanced malignant primary brain tumor in adults. GBM frequently harbors epidermal growth factor receptor (EGFR) wild-type (EGFRwt) gene amplification and/or EGFRvIII activating mutation. EGFR-driven GBM relies on the thioredoxin (Trx) and/or glutathione (GSH) antioxidant systems to withstand the excessive production of reactive oxygen species (ROS). The impact of EGFRwt or EGFRvIII overexpression on the response to a Trx/GSH co-targeting strategy is unknown. In this study, we investigated Trx/GSH co-targeting in the context of EGFR overexpression in GBM. Auranofin is a thioredoxin reductase (TrxR) inhibitor, FDA-approved for rheumatoid arthritis. L-buthionine-sulfoximine (L-BSO) inhibits GSH synthesis by targeting the glutamate-cysteine ligase catalytic (GCLC) enzyme subunit. We analyzed the mechanisms of cytotoxicity of auranofin and the interaction between auranofin and L-BSO in U87MG, U87/EGFRwt, and U87/EGFRvIII GBM isogenic GBM cell lines. ROS-dependent effects were assessed using the antioxidant N-acetylsteine. We show that auranofin decreased TrxR1 activity and increased ROS. Auranofin decreased cell vitality and colony formation and increased protein polyubiquitination through ROS-dependent mechanisms, suggesting the role of ROS in auranofin-induced cytotoxicity in the three cell lines. ROS-dependent PARP-1 cleavage was associated with EGFRvIII downregulation in U87/EGFRvIII cells. Remarkably, the auranofin and L-BSO combination induced the significant depletion of intracellular GSH and synergistic cytotoxicity regardless of EGFR overexpression. Nevertheless, molecular mechanisms associated with cytotoxicity were modulated to a different extent among the three cell lines. U87/EGFRvIII exhibited the most prominent ROS increase, P-AKT(Ser-473), and AKT decrease along with drastic EGFRvIII downregulation. U87/EGFRwt and U87/EGFRvIII displayed lower basal intracellular GSH levels and synergistic ROS-dependent DNA damage compared to U87MG cells. Our study provides evidence for ROS-dependent synergistic cytotoxicity of auranofin and L-BSO combination in GBM in vitro. Unraveling the sensitivity of EGFR-overexpressing cells to auranofin alone, and synergistic auranofin and L-BSO combination, supports the rationale to repurpose this promising pro-oxidant treatment strategy in GBM.
Collapse
Affiliation(s)
- Elvis Martinez-Jaramillo
- Experimental Pathology Unit, McGill University, Montréal, QC H3A 2B4, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Fatemeh Jamali
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Farah H Abdalbari
- Experimental Pathology Unit, McGill University, Montréal, QC H3A 2B4, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Bassam Abdulkarim
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Department of Oncology, McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Bertrand J Jean-Claude
- Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H4A 3J1, Canada
- Cancer Drug Research Laboratory, Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Carlos M Telleria
- Experimental Pathology Unit, McGill University, Montréal, QC H3A 2B4, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Siham Sabri
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| |
Collapse
|
13
|
Shao X, Xing F, Zhang Y, Lok CN, Che CM. Integrative chemoproteomics reveals anticancer mechanisms of silver(i) targeting the proteasome regulatory complex. Chem Sci 2024; 15:5349-5359. [PMID: 38577372 PMCID: PMC10988589 DOI: 10.1039/d3sc04834a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
Abstract
Silver compounds have favorable properties as promising anticancer drug candidates, such as low side effects, anti-inflammatory properties, and high potential to overcome drug resistance. However, the exact mechanism by which Ag(i) confers anticancer activity remains unclear, which hinders further development of anticancer applications of silver compounds. Here, we combine thermal proteome profiling, cysteine profiling, and ubiquitome profiling to study the molecular mechanisms of silver(i) complexes supported by non-toxic thiourea (TU) ligands. Through the formation of AgTU complexes, TU ligands deliver Ag+ ions to cancer cells and tumour xenografts to elicit inhibitory potency. Our chemical proteomics studies show that AgTU acts on the ubiquitin-proteasome system (UPS) and disrupts protein homeostasis, which has been identified as a main anticancer mechanism. Specifically, Ag+ ions are released from AgTU in the cellular environment, directly target the 19S proteasome regulatory complex, and may oxidize its cysteine residues, thereby inhibiting proteasomal activity and accumulating ubiquitinated proteins. After AgTU treatment, proteasome subunits are massively ubiquitinated and aberrantly aggregated, leading to impaired protein homeostasis and paraptotic death of cancer cells. This work reveals the unique anticancer mechanism of Ag(i) targeting the 19S proteasome regulatory complex and opens up new avenues for optimizing silver-based anticancer efficacy.
Collapse
Affiliation(s)
- Xiaojian Shao
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Fangrong Xing
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Yiwei Zhang
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Chun-Nam Lok
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Chi-Ming Che
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| |
Collapse
|
14
|
Lobas AA, Saei AA, Lyu H, Zubarev RA, Gorshkov MV. Chemical Proteomics Reveals that the Anticancer Drug Everolimus Affects the Ubiquitin-Proteasome System. ACS Pharmacol Transl Sci 2024; 7:787-796. [PMID: 38481686 PMCID: PMC10928898 DOI: 10.1021/acsptsci.3c00316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2025]
Abstract
Rapamycin is a natural antifungal, immunosuppressive, and antiproliferative compound that allosterically inhibits mTOR complex 1. The ubiquitin-proteasome system (UPS) responsible for protein turnover is usually not listed among the pathways affected by mTOR signaling. However, some previous studies have indicated the interplay between the UPS and mTOR. It has also been reported that rapamycin and its analogs can allosterically inhibit the proteasome itself. In this work, we studied the molecular effect of rapamycin and its analogs (rapalogs), everolimus and temsirolimus, on the A549 cell line by expression proteomics. The analysis of differentially expressed proteins showed that the cellular response to everolimus treatment is strikingly different from that to rapamycin and temsirolimus. In the cluster analysis, the effect of everolimus was similar to that of bortezomib, a well-established proteasome inhibitor. UPS-related pathways were enriched in the cluster of proteins specifically upregulated upon everolimus and bortezomib treatments, suggesting that both compounds have similar proteasome inhibition effects. In particular, the total amount of ubiquitin was significantly elevated in the samples treated with everolimus and bortezomib, and analysis of the polyubiquitination patterns revealed elevated intensities of the ubiquitin peptide with a GG modification at the K48 residue, consistent with a bottleneck in proteasomal protein degradation. Moreover, the everolimus treatment resulted in both ubiquitin phosphorylation and generation of a significant amount of semitryptic peptides, illustrating the increase in the protease activity. These observations suggest that everolimus affects the UPS in a unique way, and its mechanism of action is different from that of its close chemical analogs, rapamycin and temsirolimus.
Collapse
Affiliation(s)
- Anna A. Lobas
- V.
L. Talrose Institute for Energy Problems of Chemical Physics, Federal
Research Center for Chemical Physics, Russian
Academy of Sciences, 119334 Moscow, Russia
| | - Amir Ata Saei
- Division
of Physiological Chemistry I, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
- Biozentrum, University of Basel, 4056 Basel, Switzerland
- Center
for Translational Microbiome Research, Department of Microbiology,
Tumor and Cell Biology, Karolinska Institutet, SE-17 177 Stockholm, Sweden
| | - Hezheng Lyu
- Division
of Physiological Chemistry I, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
| | - Roman A. Zubarev
- Division
of Physiological Chemistry I, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
- The
National Medical Research Center for Endocrinology, 115478 Moscow, Russia
| | - Mikhail V. Gorshkov
- V.
L. Talrose Institute for Energy Problems of Chemical Physics, Federal
Research Center for Chemical Physics, Russian
Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
15
|
Komorowski L, Dabkowska A, Madzio J, Pastorczak A, Szczygiel K, Janowska M, Fidyt K, Bielecki M, Hunia J, Bajor M, Stoklosa T, Winiarska M, Patkowska E, Firczuk M. Concomitant inhibition of the thioredoxin system and nonhomologous DNA repair potently sensitizes Philadelphia-positive lymphoid leukemia to tyrosine kinase inhibitors. Hemasphere 2024; 8:e56. [PMID: 38486859 PMCID: PMC10938465 DOI: 10.1002/hem3.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024] Open
Abstract
Breakpoint cluster region-Abelson (BCR::ABL1) gene fusion is an essential oncogene in both chronic myeloid leukemia (CML) and Philadelphia-positive (Ph+) B-cell acute lymphoblastic leukemia (B-ALL). While tyrosine kinase inhibitors (TKIs) are effective in up to 95% of CML patients, 50% of Ph+ B-ALL cases do not respond to treatment or relapse. This calls for new therapeutic approaches for Ph+ B-ALL. Previous studies have shown that inhibitors of the thioredoxin (TXN) system exert antileukemic activity against B-ALL cells, particularly in combination with other drugs. Here, we present that peroxiredoxin-1 (PRDX1), one of the enzymes of the TXN system, is upregulated in Ph+ lymphoid as compared to Ph+ myeloid cells. PRDX1 knockout negatively affects the viability of Ph+ B-ALL cells and sensitizes them to TKIs. Analysis of global gene expression changes in imatinib-treated, PRDX1-deficient cells revealed that the nonhomologous end-joining (NHEJ) DNA repair is a novel vulnerability of Ph+ B-ALL cells. Accordingly, PRDX1-deficient Ph+ B-ALL cells were susceptible to NHEJ inhibitors. Finally, we demonstrated the potent efficacy of a novel combination of TKIs, TXN inhibitors, and NHEJ inhibitors against Ph+ B-ALL cell lines and primary cells, which can be further investigated as a potential therapeutic approach for the treatment of Ph+ B-ALL.
Collapse
Affiliation(s)
- Lukasz Komorowski
- Department of ImmunologyMedical University of WarsawWarsawPoland
- Postgraduate School of Molecular MedicineMedical University of WarsawWarsawPoland
| | - Agnieszka Dabkowska
- Department of ImmunologyMedical University of WarsawWarsawPoland
- Laboratory of Immunology, Mossakowski Medical Research InstitutePolish Academy of SciencesWarsawPoland
| | - Joanna Madzio
- Department of Pediatrics, Oncology and HematologyMedical University of LodzLodzPoland
| | - Agata Pastorczak
- Department of Pediatrics, Oncology and HematologyMedical University of LodzLodzPoland
| | - Kacper Szczygiel
- Department of ImmunologyMedical University of WarsawWarsawPoland
- Polpharma Biologics SAGdańskPoland
| | - Martyna Janowska
- Laboratory of Immunology, Mossakowski Medical Research InstitutePolish Academy of SciencesWarsawPoland
| | - Klaudyna Fidyt
- Department of ImmunologyMedical University of WarsawWarsawPoland
| | - Maksymilian Bielecki
- Department of PsychologySWPS University of Social Sciences and HumanitiesWarsawPoland
| | - Jaromir Hunia
- Department of ImmunologyMedical University of WarsawWarsawPoland
| | - Malgorzata Bajor
- Laboratory of Immunology, Mossakowski Medical Research InstitutePolish Academy of SciencesWarsawPoland
| | - Tomasz Stoklosa
- Department of Tumor Biology and GeneticsMedical University of WarsawWarsawPoland
| | - Magdalena Winiarska
- Department of ImmunologyMedical University of WarsawWarsawPoland
- Laboratory of Immunology, Mossakowski Medical Research InstitutePolish Academy of SciencesWarsawPoland
| | | | - Malgorzata Firczuk
- Department of ImmunologyMedical University of WarsawWarsawPoland
- Laboratory of Immunology, Mossakowski Medical Research InstitutePolish Academy of SciencesWarsawPoland
| |
Collapse
|
16
|
Ahad A, K. Saeed H, del Solar V, López-Hernández JE, Michel A, Mathew J, Lewis JS, Contel M. Shifting the Antibody-Drug Conjugate Paradigm: A Trastuzumab-Gold-Based Conjugate Demonstrates High Efficacy against Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer Mouse Model. ACS Pharmacol Transl Sci 2023; 6:1972-1986. [PMID: 38093840 PMCID: PMC10714425 DOI: 10.1021/acsptsci.3c00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 01/17/2024]
Abstract
Antibody-drug conjugates (ADCs) combine the selectivity of monoclonal antibodies (mAbs) with the efficacy of chemotherapeutics to target cancers without toxicity to normal tissue. Clinically, most chemotherapeutic ADCs are based on complex organic molecules, while the conjugation of metallodrugs to mAbs has been overlooked, despite the resurgent interest in metal-based drugs as cancer chemotherapeutics. In 2019, we described the first gold ADCs containing gold-triphenylphosphane fragments as a proof of concept. The ADCs (based on the antibody trastuzumab) were selective and highly active against HER2-positive breast cancer cells. In this study, we developed site-specific ADCs (Thio-1b and Thio-2b) using the cysteine-engineered trastuzumab derivative THIOMAB antibody technology with gold(I)-containing phosphanes and a maleimide-based linker amenable to bioconjugation (1b and 2b). In addition, we developed lysine-directed ADCs with gold payloads based on phosphanes and N-heterocyclic carbenes featuring an activated ester moiety (2c and 5c) with trastuzumab (Tras-2c and Tras-5c) and another anti-HER2 antibody, pertuzumab (Per-2c and Per-5c). Both sets of ADCs demonstrated significant anticancer potency in vitro assays. Based on these results, one ADC (Tras-2c), containing the [Au(PEt3)] fragment present in FDA-approved auranofin, was selected for an in vivo antitumor efficacy study. Immunocompromised mice xenografted with the HER2-positive human cancer cell line SKBR-3 exhibited almost complete tumor reduction and low toxicity with intravenous administration of Tras-2c. With this highly selective targeting system, we demonstrated that a subnanomolar cytotoxicity profile in cells is not required for an impressive antitumor effect in a mouse xenograft model.
Collapse
Affiliation(s)
- Afruja Ahad
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Hiwa K. Saeed
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Virginia del Solar
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Javier E. López-Hernández
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| | - Alexa Michel
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Joshua Mathew
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Maria Contel
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Chemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| |
Collapse
|
17
|
Ren J, Yu P, Liu S, Li R, Niu X, Chen Y, Zhang Z, Zhou F, Zhang L. Deubiquitylating Enzymes in Cancer and Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303807. [PMID: 37888853 PMCID: PMC10754134 DOI: 10.1002/advs.202303807] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/30/2023] [Indexed: 10/28/2023]
Abstract
Deubiquitylating enzymes (DUBs) maintain relative homeostasis of the cellular ubiquitome by removing the post-translational modification ubiquitin moiety from substrates. Numerous DUBs have been demonstrated specificity for cleaving a certain type of ubiquitin linkage or positions within ubiquitin chains. Moreover, several DUBs perform functions through specific protein-protein interactions in a catalytically independent manner, which further expands the versatility and complexity of DUBs' functions. Dysregulation of DUBs disrupts the dynamic equilibrium of ubiquitome and causes various diseases, especially cancer and immune disorders. This review summarizes the Janus-faced roles of DUBs in cancer including proteasomal degradation, DNA repair, apoptosis, and tumor metastasis, as well as in immunity involving innate immune receptor signaling and inflammatory and autoimmune disorders. The prospects and challenges for the clinical development of DUB inhibitors are further discussed. The review provides a comprehensive understanding of the multi-faced roles of DUBs in cancer and immunity.
Collapse
Affiliation(s)
- Jiang Ren
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Peng Yu
- Zhongshan Institute for Drug DiscoveryShanghai Institute of Materia MedicaChinese Academy of SciencesZhongshanGuangdongP. R. China
| | - Sijia Liu
- International Biomed‐X Research CenterSecond Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhouP. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhou310058China
| | - Ran Li
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Xin Niu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Yan Chen
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Zhenyu Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Long Zhang
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
- International Biomed‐X Research CenterSecond Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhouP. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| |
Collapse
|
18
|
Mrkvicová A, Peterová E, Nemec I, Křikavová R, Muthná D, Havelek R, Kazimírová P, Řezáčová M, Štarha P. Rh(III) and Ru(II) complexes with phosphanyl-alkylamines: inhibition of DNA synthesis induced by anticancer Rh complex. Future Med Chem 2023; 15:1583-1602. [PMID: 37750220 DOI: 10.4155/fmc-2023-0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
Aim: This investigation was designed to synthesize half-sandwich Rh(III) and Ru(II) complexes and study their antiproliferative activity in human cancer cell lines. Materials & methods: Nine compounds were prepared and tested by various assays for their anticancer activity and mechanism of action. Results: Hit Rh(III) complex 6 showed low-micromolar potency in cisplatin-sensitive (A2780) and -resistant (A2780cis) ovarian carcinoma cell lines, promising selectivity toward these cancer cells over normal lung fibroblasts and an unprecedented mechanism of action in the treated cells. DNA synthesis was decreased and CDKN1A expression was upregulated, but p21 expression was not induced. Conclusion: Rh complex 6 showed high antiproliferative activity, which is induced through a p21-independent mechanism of action.
Collapse
Affiliation(s)
- Alena Mrkvicová
- Department of Medical Biochemistry, Charles University, Šimkova 870, 500 03, Hradec Králové, Czech Republic
| | - Eva Peterová
- Department of Medical Biochemistry, Charles University, Šimkova 870, 500 03, Hradec Králové, Czech Republic
| | - Ivan Nemec
- Department of Inorganic Chemistry, Palacký University Olomouc, 17. listopadu 12, 771 46, Olomouc, Czech Republic
| | - Radka Křikavová
- Department of Inorganic Chemistry, Palacký University Olomouc, 17. listopadu 12, 771 46, Olomouc, Czech Republic
| | - Darina Muthná
- Department of Medical Biochemistry, Charles University, Šimkova 870, 500 03, Hradec Králové, Czech Republic
| | - Radim Havelek
- Department of Medical Biochemistry, Charles University, Šimkova 870, 500 03, Hradec Králové, Czech Republic
| | - Petra Kazimírová
- Department of Medical Biochemistry, Charles University, Šimkova 870, 500 03, Hradec Králové, Czech Republic
| | - Martina Řezáčová
- Department of Medical Biochemistry, Charles University, Šimkova 870, 500 03, Hradec Králové, Czech Republic
| | - Pavel Štarha
- Department of Inorganic Chemistry, Palacký University Olomouc, 17. listopadu 12, 771 46, Olomouc, Czech Republic
| |
Collapse
|
19
|
Yan W, Zhong Y, Hu X, Xu T, Zhang Y, Kales S, Qu Y, Talley DC, Baljinnyam B, LeClair CA, Simeonov A, Polster BM, Huang R, Ye Y, Rai G, Henderson MJ, Tao D, Fang S. Auranofin targets UBA1 and enhances UBA1 activity by facilitating ubiquitin trans-thioesterification to E2 ubiquitin-conjugating enzymes. Nat Commun 2023; 14:4798. [PMID: 37558718 PMCID: PMC10412574 DOI: 10.1038/s41467-023-40537-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023] Open
Abstract
UBA1 is the primary E1 ubiquitin-activating enzyme responsible for generation of activated ubiquitin required for ubiquitination, a process that regulates stability and function of numerous proteins. Decreased or insufficient ubiquitination can cause or drive aging and many diseases. Therefore, a small-molecule enhancing UBA1 activity could have broad therapeutic potential. Here we report that auranofin, a drug approved for the treatment of rheumatoid arthritis, is a potent UBA1 activity enhancer. Auranofin binds to the UBA1's ubiquitin fold domain and conjugates to Cys1039 residue. The binding enhances UBA1 interactions with at least 20 different E2 ubiquitin-conjugating enzymes, facilitating ubiquitin charging to E2 and increasing the activities of seven representative E3s in vitro. Auranofin promotes ubiquitination and degradation of misfolded ER proteins during ER-associated degradation in cells at low nanomolar concentrations. It also facilitates outer mitochondrial membrane-associated degradation. These findings suggest that auranofin can serve as a much-needed tool for UBA1 research and therapeutic exploration.
Collapse
Affiliation(s)
- Wenjing Yan
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yongwang Zhong
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Xin Hu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Tuan Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yinghua Zhang
- Center for Innovative Biomedical Resources, Biosensor Core, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Stephen Kales
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yanyan Qu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Daniel C Talley
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Bolormaa Baljinnyam
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA.
| | - Shengyun Fang
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Program in Oncology, UM Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
20
|
O’Shaughnessy M, Sheils O, Baird AM. The Lung Microbiome in COPD and Lung Cancer: Exploring the Potential of Metal-Based Drugs. Int J Mol Sci 2023; 24:12296. [PMID: 37569672 PMCID: PMC10419288 DOI: 10.3390/ijms241512296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer 17 are two of the most prevalent and debilitating respiratory diseases worldwide, both associated with high morbidity and mortality rates. As major global health concerns, they impose a substantial burden on patients, healthcare systems, and society at large. Despite their distinct aetiologies, lung cancer and COPD share common risk factors, clinical features, and pathological pathways, which have spurred increasing research interest in their co-occurrence. One area of particular interest is the role of the lung microbiome in the development and progression of these diseases, including the transition from COPD to lung cancer. Exploring novel therapeutic strategies, such as metal-based drugs, offers a potential avenue for targeting the microbiome in these diseases to improve patient outcomes. This review aims to provide an overview of the current understanding of the lung microbiome, with a particular emphasis on COPD and lung cancer, and to discuss the potential of metal-based drugs as a therapeutic strategy for these conditions, specifically concerning targeting the microbiome.
Collapse
Affiliation(s)
- Megan O’Shaughnessy
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, St. James’s Hospital, D08 RX0X Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| |
Collapse
|
21
|
Zhong Z, Zhang C, Ni S, Ma M, Zhang X, Sang W, Lv T, Qian Z, Yi C, Yu B. NFATc1-mediated expression of SLC7A11 drives sensitivity to TXNRD1 inhibitors in osteoclast precursors. Redox Biol 2023; 63:102711. [PMID: 37148740 PMCID: PMC10184050 DOI: 10.1016/j.redox.2023.102711] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023] Open
Abstract
Excess osteoclast activity is found in many bone metabolic diseases, and inhibiting osteoclast differentiation has proven to be an effective strategy. Here, we revealed that osteoclast precursors (pre-OCs) were more susceptible to thioredoxin reductase 1 (TXNRD1) inhibitors than bone marrow-derived monocytes (BMDMs) during receptor activator of nuclear factor kappa B ligand (RANKL)-mediated osteoclastogenesis. Mechanistically, we found that nuclear factor of activated T-cells 1 (NFATc1) upregulated solute carrier family 7 member 11 (SLC7A11) expression through transcriptional regulation during RANKL-induced osteoclastogenesis. During TXNRD1 inhibition, the rate of intracellular disulfide reduction is significantly reduced. Increased cystine transport leads to increased cystine accumulation, which leads to increased cellular disulfide stress and disulfidptosis. We further demonstrated that SLC7A11 inhibitors and treatments that prevent disulphide accumulation could rescue this type of cell death, but not the ferroptosis inhibitors (DFO, Ferro-1), the ROS scavengers (Trolox, Tempol), the apoptosis inhibitor (Z-VAD), the necroptosis inhibitor (Nec-1), or the autophagy inhibitor (CQ). An in vivo study indicated that TXNRD1 inhibitors increased bone cystine content, reduced the number of osteoclasts, and alleviated bone loss in an ovariectomized (OVX) mouse model. Together, our findings demonstrate that NFATc1-mediated upregulation of SLC7A11 induces targetable metabolic sensitivity to TXNRD1 inhibitors during osteoclast differentiation. Moreover, we innovatively suggest that TXNRD1 inhibitors, a classic drug for osteoclast-related diseases, selectively kill pre-OCs by inducing intracellular cystine accumulation and subsequent disulfidptosis.
Collapse
Affiliation(s)
- Zeyuan Zhong
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China; Shanghai Medical College, Fudan University, Shanghai, China
| | - Chongjing Zhang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China; Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuo Ni
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Miao Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaomeng Zhang
- Renal Medicine and Baxter Novum, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Weicong Sang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Tao Lv
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Zhi Qian
- Institution of Orthopaedic Diseases, Zhangye People's Hospital Affiliated to Hexi University, Zhangye, China.
| | - Chengqing Yi
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| | - Baoqing Yu
- Department of Orthopedics, Shanghai Pudong New Area People's Hospital, Shanghai, China.
| |
Collapse
|
22
|
Schleser SW, Ghosh H, Hörner G, Seib J, Bhattacharyya S, Weber B, Schobert R, Dandawate P, Biersack B. New 4,5-Diarylimidazol-2-ylidene-iodidogold(I) Complexes with High Activity against Esophageal Adenocarcinoma Cells. Int J Mol Sci 2023; 24:5738. [PMID: 36982817 PMCID: PMC10052191 DOI: 10.3390/ijms24065738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Inspired by the vascular-disrupting agent combretastatin A-4 and recently published anticancer active N-heterocyclic carbene (NHC) complexes of Au(I), a series of new iodidogold(I)-NHC complexes was synthesized and characterized. The iodidogold(I) complexes were synthesized by a route involving van Leusen imidazole formation and N-alkylation, followed by complexation with Ag2O, transmetalation with chloro(dimethylsulfide)gold(I) [Au(DMS)Cl], and anion exchange with KI. The target complexes were characterized by IR spectroscopy, 1H and 13C NMR spectroscopy, and mass spectrometry. The structure of 6c was validated via single-crystal X-ray diffraction. A preliminary anticancer screening of the complexes using two esophageal adenocarcinoma cell lines showed promising nanomolar activities for certain iodidogold(I) complexes accompanied with apoptosis induction, as well as c-Myc and cyclin D1 suppression in esophageal adenocarcinoma cells treated with the most promising derivative 6b.
Collapse
Affiliation(s)
- Sebastian W. Schleser
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Hindole Ghosh
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Gerald Hörner
- Inorganic Chemistry IV, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Jonathan Seib
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Sangita Bhattacharyya
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Birgit Weber
- Inorganic Chemistry IV, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Rainer Schobert
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Prasad Dandawate
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Bernhard Biersack
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| |
Collapse
|
23
|
Seo MJ, Kim IY, Lee DM, Park YJ, Cho MY, Jin HJ, Choi KS. Dual inhibition of thioredoxin reductase and proteasome is required for auranofin-induced paraptosis in breast cancer cells. Cell Death Dis 2023; 14:42. [PMID: 36658130 PMCID: PMC9852458 DOI: 10.1038/s41419-023-05586-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
Auranofin (AF), a gold (I)-containing phosphine compound, is being investigated for oncological application as a repurposed drug. We show here that 4~5 µM AF induces paraptosis, a non-apoptotic cell death mode characterized by dilation of the endoplasmic reticulum (ER) and mitochondria, in breast cancer cells. Although the covalent inhibition of thioredoxin reductase (TrxR), an enzyme that critically controls intracellular redox homeostasis, is considered the primary mechanism of AF's anticancer activity, knockdown of TrxR1 did not induce paraptosis. Instead, both TrxR1 knockdown plus the proteasome inhibitor (PI), bortezomib (Bz), and 2 μM AF plus Bz induced paraptosis, thereby mimicking the effect of 5 μM AF. These results suggest that the paraptosis induced by 5 μM AF requires the inhibition of both TrxR1 and proteasome. We found that TrxR1 knockdown/Bz or subtoxic doses of AF and Bz induced paraptosis selectively in breast cancer cells, sparing non-transformed MCF10A cells, whereas 4~5 μM AF killed both cancer and MCF10A cells. GSH depletion was found to be more critical than ROS generation for the paraptosis induced by dual TrxR1/proteasome inhibition. In this process, the ATF4/CHAC1 (glutathione-specific gamma-glutamylcyclotransferase 1) axis leads to GSH degradation, contributing to proteotoxic stress possibly due to the accumulation of misfolded thiol-containing proteins. These results suggest that the paraptosis-inducing strategy of AF plus a PI may provide an effective therapeutic strategy against pro-apoptotic therapy-resistant cancers and reduce the potential side effects associated with high-dose AF.
Collapse
Affiliation(s)
- Min Ji Seo
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
| | - In Young Kim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
- Nano-safety Team, Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Korea
| | - Dong Min Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
| | - Yeon Jung Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
| | - Mi-Young Cho
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Hyo Joon Jin
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Ajou University School of Medicine, Suwon, 16499, Korea
| | - Kyeong Sook Choi
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea.
| |
Collapse
|
24
|
Optimization of the Solvent and In Vivo Administration Route of Auranofin in a Syngeneic Non-Small Cell Lung Cancer and Glioblastoma Mouse Model. Pharmaceutics 2022; 14:pharmaceutics14122761. [PMID: 36559255 PMCID: PMC9783082 DOI: 10.3390/pharmaceutics14122761] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
The antineoplastic activity of the thioredoxin reductase 1 (TrxR) inhibitor, auranofin (AF), has already been investigated in various cancer mouse models as a single drug, or in combination with other molecules. However, there are inconsistencies in the literature on the solvent, dose and administration route of AF treatment in vivo. Therefore, we investigated the solvent and administration route of AF in a syngeneic SB28 glioblastoma (GBM) C57BL/6J and a 344SQ non-small cell lung cancer 129S2/SvPasCrl (129) mouse model. Compared to daily intraperitoneal injections and subcutaneous delivery of AF via osmotic minipumps, oral gavage for 14 days was the most suitable administration route for high doses of AF (10-15 mg/kg) in both mouse models, showing no measurable weight loss or signs of toxicity. A solvent comprising 50% DMSO, 40% PEG300 and 10% ethanol improved the solubility of AF for oral administration in mice. In addition, we confirmed that AF was a potent TrxR inhibitor in SB28 GBM tumors at high doses. Taken together, our results and results in the literature indicate the therapeutic value of AF in several in vivo cancer models, and provide relevant information about AF's optimal administration route and solvent in two syngeneic cancer mouse models.
Collapse
|
25
|
Speciation Analysis Highlights the Interactions of Auranofin with the Cytoskeleton Proteins of Lung Cancer Cells. Pharmaceuticals (Basel) 2022; 15:ph15101285. [PMID: 36297397 PMCID: PMC9610265 DOI: 10.3390/ph15101285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 12/01/2022] Open
Abstract
Two types of lung cells (epithelial cancer lung cells, A-549 and lung fibroblasts MRC-5) were exposed to the clinically established gold drug auranofin at concentrations close to the half-maximal inhibitory drug concentrations (IC50). Collected cells were subjected to speciation analysis using inductively coupled plasma mass spectrometry (ICP-MS). Auranofin showed better affinity toward proteins than DNA, RNA, and hydrophilic small molecular weight compounds. It can bind to proteins that vary in size (~20 kDa, ~75 kDa, and ≥200 kDa) and pI. However, the possibility of dimerization and protein–protein complex formation should also be taken into account. µRPLC/CZE-ESI-MS/MS studies on trypsinized proteins allowed the indication of 76 peptides for which signal intensity was influenced by auranofin presence in cells. Based on it, identity was proposed for 20 proteins. Except for thioredoxin reductase (TrxR), which is directly targeted by gold complex, the proteins were found to be transformed. Five indicated proteins: myosin, plectin, talin, two annexins, and kinase M3K5, are responsible for cell–cell, cell–protein interactions, and cell motility. A wound healing test confirmed their regulation by auranofin as cell migration decreased by 40% while the cell cycle was not interrupted.
Collapse
|
26
|
Zhang J, Li Y, Fang R, Wei W, Wang Y, Jin J, Yang F, Chen J. Organometallic gold(I) and gold(III) complexes for lung cancer treatment. Front Pharmacol 2022; 13:979951. [PMID: 36176441 PMCID: PMC9513137 DOI: 10.3389/fphar.2022.979951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Metal compounds, especially gold complexes, have recently gained increasing attention as possible lung cancer therapeutics. Some gold complexes display not only excellent activity in cisplatin-sensitive lung cancer but also in cisplatin-resistant lung cancer, revealing promising prospects in the development of novel treatments for lung cancer. This review summarizes examples of anticancer gold(I) and gold (III) complexes for lung cancer treatment, including mechanisms of action and approaches adopted to improve their efficiency. Several excellent examples of gold complexes against lung cancer are highlighted.
Collapse
Affiliation(s)
- Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Yanping Li
- School of Public Health, Guilin Medical University, Guilin, China
| | - Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Wei Wei
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Yong Wang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
- *Correspondence: Feng Yang, mailto:, Jian Chen, mailto:
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
- *Correspondence: Feng Yang, mailto:, Jian Chen, mailto:
| |
Collapse
|
27
|
Sachdeva H, Khaturia S, Saquib M, Khatik N, Khandelwal AR, Meena R, Sharma K. Oxygen- and Sulphur-Containing Heterocyclic Compounds as Potential Anticancer Agents. Appl Biochem Biotechnol 2022; 194:6438-6467. [PMID: 35900713 DOI: 10.1007/s12010-022-04099-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 11/28/2022]
Abstract
Oxygen- and sulphur-based heterocycles form the core structure of many biologically active molecules as well as U.S. FDA-approved drugs. Moreover, they possess broad range of biological activities, viz. anticancer, antiinflammatory, antioxidant, antitumour, antibacterial, antiviral, antidiabetic, anticonvulsant, anti-tubercular, analgesic, anti-leishmanial, antimalarial, antifungal, and anti-histaminic, Hence, O- and S-based heterocycles are gaining more attention in recent years on the road to the discovery of innovative anticancer drugs after the extensive investigation of nitrogen-based heterocycles as anticancer agents. Several attempts have been made to synthesize fused oxygen- and sulphur-based heterocyclic derivatives as joining one heterocyclic moiety with another may lead to improvement in the biological profile of a molecule. Humans have been cursed with cancer since long time. Despite the development of several heterocyclic anticancer medications such as 5-fluorouracil, doxorubicin, methotrexate, and daunorubicin, cure of cancer is difficult. Hence, researchers are trying to synthesize new fused/spiro heterocyclic molecules to discover novel anticancer drugs which may show promising anticancer effects with fewer side effects. Furthermore, fused heterocycles behave as DNA intercalating agents which have the ability to interact with DNA, leading to cell death thereby exerting anticancer effect. This review article highlights the synthesis and anticancer potentiality of oxygen- and sulphur-containing heterocyclic compounds covering the period from 2011 to 2021.
Collapse
Affiliation(s)
- Harshita Sachdeva
- Department of Chemistry, University of Rajasthan, 302004, Jaipur, Rajasthan, India.
| | - Sarita Khaturia
- Department of Chemistry, School of Liberal Arts and Sciences, Mody University of Science and Technology, Lakshmangarh (Sikar), Rajasthan, India
| | - Mohammad Saquib
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Narsingh Khatik
- Department of Chemistry, University of Rajasthan, 302004, Jaipur, Rajasthan, India
| | | | - Ravina Meena
- Department of Chemistry, University of Rajasthan, 302004, Jaipur, Rajasthan, India
| | - Khushboo Sharma
- Department of Chemistry, University of Rajasthan, 302004, Jaipur, Rajasthan, India
| |
Collapse
|
28
|
El-Lateef HMA, El-Dabea T, Khalaf MM, Abu-Dief AM. Development of Metal Complexes for Treatment of Coronaviruses. Int J Mol Sci 2022; 23:6418. [PMID: 35742870 PMCID: PMC9223400 DOI: 10.3390/ijms23126418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/29/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Coronavirus disease (SARS-CoV-2) is a global epidemic. This pandemic, which has been linked to high rates of death, has forced some countries throughout the world to implement complete lockdowns in order to contain the spread of infection. Because of the advent of new coronavirus variants, it is critical to find effective treatments and vaccines to prevent the virus's rapid spread over the world. In this regard, metal complexes have attained immense interest as antibody modifiers and antiviral therapies, and they have a lot of promise towards SARS-CoV-2 and their suggested mechanisms of action are discussed, i.e., a new series of metal complexes' medicinal vital role in treatment of specific proteins or SARS-CoV-2 are described. The structures of the obtained metal complexes were fully elucidated by different analytical and spectroscopic techniques also. Molecular docking and pharmacophore studies presented that most of complexes studied influenced good binding affinity to the main protease SARS-CoV-2, which also was attained as from the RCSB pdb (Protein Data Bank) data PDB ID: 6 W41, to expect the action of metal complexes in contradiction of COVID-19. Experimental research is required to determine the pharmacokinetics of most of the complexes analyzed for the treatment of SARS-CoV-2-related disease. Finally, the toxicity of a metal-containing inorganic complex will thus be discussed by its capability to transfer metals which may bind with targeted site.
Collapse
Affiliation(s)
- Hany M. Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Tarek El-Dabea
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Mai M. Khalaf
- Department of Chemistry, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Ahmed M. Abu-Dief
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
- Chemistry Department, College of Science, Taibah University, Madinah 344, Saudi Arabia
| |
Collapse
|
29
|
Cheng XF, Wang N, Jiang Z, Chen Z, Niu Y, Tong L, Yu T, Tang B. Quantitative Chemoproteomic Profiling of Targets of Au(I) Complexes by Competitive Activity-Based Protein Profiling. Bioconjug Chem 2022; 33:1131-1137. [PMID: 35576584 DOI: 10.1021/acs.bioconjchem.2c00080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Owing to the encouraging pharmacological action and acceptable toxicity profile, Au(I) complexes have attracted growing interest in the application of disease treatment. In order to investigate their potential target proteins and related bioinformation, herein, we screened four Au(I) complexes and explored the binding proteins utilizing a competitive activity-based protein profiling (ABPP) strategy, including identification experiments and reactivity classification experiments, which offers a simple and robust method to identify the target proteins of Au(I) complexes. We quantified the target proteins of the four Au(I) complexes and found that most of proteins were associated with cancer. In addition, the newly Au(I)-binding proteins and biological gold-protein interaction pathways were exhibited. Furthermore, we estimated the correlation between target proteins of Au(I) complexes and various cancers, which will promote the development of the gold anticancer drugs.
Collapse
Affiliation(s)
- Xiu-Fen Cheng
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Nan Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Zhongyao Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Zhenzhen Chen
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Yaxin Niu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Lili Tong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Ting Yu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| |
Collapse
|
30
|
Gamberi T, Chiappetta G, Fiaschi T, Modesti A, Sorbi F, Magherini F. Upgrade of an old drug: Auranofin in innovative cancer therapies to overcome drug resistance and to increase drug effectiveness. Med Res Rev 2022; 42:1111-1146. [PMID: 34850406 PMCID: PMC9299597 DOI: 10.1002/med.21872] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022]
Abstract
Auranofin is an oral gold(I) compound, initially developed for the treatment of rheumatoid arthritis. Currently, Auranofin is under investigation for oncological application within a drug repurposing plan due to the relevant antineoplastic activity observed both in vitro and in vivo tumor models. In this review, we analysed studies in which Auranofin was used as a single drug or in combination with other molecules to enhance their anticancer activity or to overcome chemoresistance. The analysis of different targets/pathways affected by this drug in different cancer types has allowed us to highlight several interesting targets and effects of Auranofin besides the already well-known inhibition of thioredoxin reductase. Among these targets, inhibitory-κB kinase, deubiquitinates, protein kinase C iota have been frequently suggested. To rationalize the effects of Auranofin by a system biology-like approach, we exploited transcriptomic data obtained from a wide range of cell models, extrapolating the data deposited in the Connectivity Maps website and we attempted to provide a general conclusion and discussed the major points that need further investigation.
Collapse
Affiliation(s)
- Tania Gamberi
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics GroupPlasticité du Cerveau UMR 8249 CNRSParisESPCI Paris‐PSLFrance
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Flavia Sorbi
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| |
Collapse
|
31
|
Ferraro MG, Piccolo M, Misso G, Santamaria R, Irace C. Bioactivity and Development of Small Non-Platinum Metal-Based Chemotherapeutics. Pharmaceutics 2022; 14:pharmaceutics14050954. [PMID: 35631543 PMCID: PMC9147010 DOI: 10.3390/pharmaceutics14050954] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Countless expectations converge in the multidisciplinary endeavour for the search and development of effective and safe drugs in fighting cancer. Although they still embody a minority of the pharmacological agents currently in clinical use, metal-based complexes have great yet unexplored potential, which probably hides forthcoming anticancer drugs. Following the historical success of cisplatin and congeners, but also taking advantage of conventional chemotherapy limitations that emerged with applications in the clinic, the design and development of non-platinum metal-based chemotherapeutics, either as drugs or prodrugs, represents a rapidly evolving field wherein candidate compounds can be fine-tuned to access interactions with druggable biological targets. Moving in this direction, over the last few decades platinum family metals, e.g., ruthenium and palladium, have been largely proposed. Indeed, transition metals and molecular platforms where they originate are endowed with unique chemical and biological features based on, but not limited to, redox activity and coordination geometries, as well as ligand selection (including their inherent reactivity and bioactivity). Herein, current applications and progress in metal-based chemoth are reviewed. Converging on the recent literature, new attractive chemotherapeutics based on transition metals other than platinum—and their bioactivity and mechanisms of action—are examined and discussed. A special focus is committed to anticancer agents based on ruthenium, palladium, rhodium, and iridium, but also to gold derivatives, for which more experimental data are nowadays available. Next to platinum-based agents, ruthenium-based candidate drugs were the first to reach the stage of clinical evaluation in humans, opening new scenarios for the development of alternative chemotherapeutic options to treat cancer.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Marialuisa Piccolo
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Gabriella Misso
- Department of Precision Medicine, School of Medicine and Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: (G.M.); (C.I.)
| | - Rita Santamaria
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Carlo Irace
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
- Correspondence: (G.M.); (C.I.)
| |
Collapse
|
32
|
Tu R, Ma J, Zhang P, Kang Y, Xiong X, Zhu J, Li M, Zhang C. The emerging role of deubiquitylating enzymes as therapeutic targets in cancer metabolism. Cancer Cell Int 2022; 22:130. [PMID: 35307036 PMCID: PMC8935717 DOI: 10.1186/s12935-022-02524-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 02/14/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractCancer cells must rewire cellular metabolism to satisfy the unbridled proliferation, and metabolic reprogramming provides not only the advantage for cancer cell proliferation but also new targets for cancer treatment. However, the plasticity of the metabolic pathways makes them very difficult to target. Deubiquitylating enzymes (DUBs) are proteases that cleave ubiquitin from the substrate proteins and process ubiquitin precursors. While the molecular mechanisms are not fully understood, many DUBs have been shown to be involved in tumorigenesis and progression via controlling the dysregulated cancer metabolism, and consequently recognized as potential drug targets for cancer treatment. In this article, we summarized the significant progress in understanding the key roles of DUBs in cancer cell metabolic rewiring and the opportunities for the application of DUBs inhibitors in cancer treatment, intending to provide potential implications for both research purpose and clinical applications.
Collapse
|
33
|
Potent, p53-independent induction of NOXA sensitizes MLL-rearranged B-cell acute lymphoblastic leukemia cells to venetoclax. Oncogene 2022; 41:1600-1609. [PMID: 35091682 PMCID: PMC8913358 DOI: 10.1038/s41388-022-02196-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 01/02/2022] [Accepted: 01/18/2022] [Indexed: 12/11/2022]
Abstract
The prognosis for B-cell precursor acute lymphoblastic leukemia patients with Mixed-Lineage Leukemia (MLL) gene rearrangements (MLLr BCP-ALL) is still extremely poor. Inhibition of anti-apoptotic protein BCL-2 with venetoclax emerged as a promising strategy for this subtype of BCP-ALL, however, lack of sufficient responses in preclinical models and the possibility of developing resistance exclude using venetoclax as monotherapy. Herein, we aimed to uncover potential mechanisms responsible for limited venetoclax activity in MLLr BCP-ALL and to identify drugs that could be used in combination therapy. Using RNA-seq, we observed that long-term exposure to venetoclax in vivo in a patient-derived xenograft model leads to downregulation of several tumor protein 53 (TP53)-related genes. Interestingly, auranofin, a thioredoxin reductase inhibitor, sensitized MLLr BCP-ALL to venetoclax in various in vitro and in vivo models, independently of the p53 pathway functionality. Synergistic activity of these drugs resulted from auranofin-mediated upregulation of NOXA pro-apoptotic protein and potent induction of apoptotic cell death. More specifically, we observed that auranofin orchestrates upregulation of the NOXA-encoding gene Phorbol-12-Myristate-13-Acetate-Induced Protein 1 (PMAIP1) associated with chromatin remodeling and increased transcriptional accessibility. Altogether, these results present an efficacious drug combination that could be considered for the treatment of MLLr BCP-ALL patients, including those with TP53 mutations.
Collapse
|
34
|
Abstract
INTRODUCTION High-risk HPV infections are related to several epithelial cancers. Despite the availability of prophylactic vaccines, HPV infections are still responsible for about 5% of all human malignancies worldwide. While therapeutic vaccines are ongoing clinical trials, genotoxic agents and surgical interventions represent current clinical treatments, with no specific anti-HPV drugs yet available in the clinics. AREAS COVERED We offer a comprehensive report of small molecules in preclinical studies proposed as potential anticancer agents against HPV-driven tumors. Given the importance of HPV oncoproteins for cancer maintenance, particularly E6 and E7, we present a classification of both non-targeted and targeted agents, with a further subdivision of the latter into two categories according to their either direct or indirect activity against viral protein functions. EXPERT OPINION Prophylactic vaccines can prevent the insurgence of HPV-related cancers, but have no effect against pre-existing infections. Moreover, their high cost, genotype-restricted effect and the growing worldwide distrust for vaccines make the availability of a specific drug an unmet medical need. Different viral early proteins emerge as ideal candidates for drug development. We highlight the most promising strategies and address future challenges in this field to herald the prospect of a specific therapeutic regimen against HPV-related cancers.
Collapse
Affiliation(s)
- Lorenzo Messa
- Department of Molecular Medicine, University of Padua, Padua, 35121, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, 35121, Italy.,Clinical Microbiology and Virology Unit, Padua University Hospital, Padua, Italy
| |
Collapse
|
35
|
Lin Z, Lin Z, Zhao Y, Cheng N, Zhang D, Lin J, Zhang H, Lin D. Auranofin and ICG-001 Emerge Synergistic Anti-tumor Effect on Canine Breast Cancer by Inducing Apoptosis via Mitochondrial Pathway. Front Vet Sci 2021; 8:772687. [PMID: 34977210 PMCID: PMC8714754 DOI: 10.3389/fvets.2021.772687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Canine breast cancer (CBC) is the most common spontaneous tumor in intact female dogs, especially in developing countries. The effective anti-tumor agents or therapies for the clinical treatment of CBC are still in need. Auranofin (AF) is a gold complex that has been attested by FDA for treating human rheumatism, which has been found as a great anti-tumor agent in recent years. ICG-001 is a small molecule inhibitor of Wnt/β-catenin pathway. In the present study, we demonstrated that a combination of AF and ICG-001 could synergistically suppress the proliferation of CBC in vitro and in vivo. Moreover, the synergistical effect was related with apoptosis caused by mitochondrial damage and ROS production. In conclusion, combination of AF and ICG-001 could synergistically suppress the growth of CBC in vitro and in vivo by leading apoptosis via mitochondrial signaling pathway and might provide a novel potential choice for the clinical treatment of CBC.
Collapse
Affiliation(s)
- Zhaoyan Lin
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zixiang Lin
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ying Zhao
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Nan Cheng
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Di Zhang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiahao Lin
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hong Zhang
- College of Animal Science and Technology, Hainan University, Haikou, China
- *Correspondence: Hong Zhang
| | - Degui Lin
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Degui Lin
| |
Collapse
|
36
|
Parihar N, Bhatt LK. Deubiquitylating enzymes: potential target in autoimmune diseases. Inflammopharmacology 2021; 29:1683-1699. [PMID: 34792672 DOI: 10.1007/s10787-021-00890-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022]
Abstract
The ubiquitin-proteasome pathway is responsible for the turnover of different cellular proteins, such as transport proteins, presentation of antigens to the immune system, control of the cell cycle, and activities that promote cancer. The enzymes which remove ubiquitin, deubiquitylating enzymes (DUBs), play a critical role in central and peripheral immune tolerance to prevent the development of autoimmune diseases and thus present a potential therapeutic target for the treatment of autoimmune diseases. DUBs function by removing ubiquitin(s) from target protein and block ubiquitin chain elongation. The addition and removal of ubiquitin molecules have a significant impact on immune responses. DUBs and E3 ligases both specifically cleave target protein and modulate protein activity and expression. The balance between ubiquitylation and deubiquitylation modulates protein levels and also protein interactions. Dysregulation of the ubiquitin-proteasome pathway results in the development of various autoimmune diseases such as inflammatory bowel diseases (IBD), psoriasis, multiple sclerosis (MS), systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). This review summarizes the current understanding of ubiquitination in autoimmune diseases and focuses on various DUBs responsible for the progression of autoimmune diseases.
Collapse
Affiliation(s)
- Niraj Parihar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
37
|
|
38
|
Abdalbari FH, Telleria CM. The gold complex auranofin: new perspectives for cancer therapy. Discov Oncol 2021; 12:42. [PMID: 35201489 PMCID: PMC8777575 DOI: 10.1007/s12672-021-00439-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced stages of cancer are highly associated with short overall survival in patients due to the lack of long-term treatment options following the standard form of care. New options for cancer therapy are needed to improve the survival of cancer patients without disease recurrence. Auranofin is a clinically approved agent against rheumatoid arthritis that is currently enrolled in clinical trials for potential repurposing against cancer. Auranofin mainly targets the anti-oxidative system catalyzed by thioredoxin reductase (TrxR), which protects the cell from oxidative stress and death in the cytoplasm and the mitochondria. TrxR is over-expressed in many cancers as an adaptive mechanism for cancer cell proliferation, rendering it an attractive target for cancer therapy, and auranofin as a potential therapeutic agent for cancer. Inhibiting TrxR dysregulates the intracellular redox state causing increased intracellular reactive oxygen species levels, and stimulates cellular demise. An alternate mechanism of action of auranofin is to mimic proteasomal inhibition by blocking the ubiquitin-proteasome system (UPS), which is critically important in cancer cells to prevent cell death when compared to non-cancer cells, because of its role on cell cycle regulation, protein degradation, gene expression, and DNA repair. This article provides new perspectives on the potential mechanisms used by auranofin alone, in combination with diverse other compounds, or in combination with platinating agents and/or immune checkpoint inhibitors to combat cancer cells, while assessing the feasibility for its repurposing in the clinical setting.
Collapse
Affiliation(s)
- Farah H Abdalbari
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Carlos M Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
39
|
Lei H, Wang J, Hu J, Zhu Q, Wu Y. Deubiquitinases in hematological malignancies. Biomark Res 2021; 9:66. [PMID: 34454635 PMCID: PMC8401176 DOI: 10.1186/s40364-021-00320-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Deubiquitinases (DUBs) are enzymes that control the stability, interactions or localization of most cellular proteins by removing their ubiquitin modification. In recent years, some DUBs, such as USP7, USP9X and USP10, have been identified as promising therapeutic targets in hematological malignancies. Importantly, some potent inhibitors targeting the oncogenic DUBs have been developed, showing promising inhibitory efficacy in preclinical models, and some have even undergone clinical trials. Different DUBs perform distinct function in diverse hematological malignancies, such as oncogenic, tumor suppressor or context-dependent effects. Therefore, exploring the biological roles of DUBs and their downstream effectors will provide new insights and therapeutic targets for the occurrence and development of hematological malignancies. We summarize the DUBs involved in different categories of hematological malignancies including leukemia, multiple myeloma and lymphoma. We also present the recent development of DUB inhibitors and their applications in hematological malignancies. Together, we demonstrate DUBs as potential therapeutic drug targets in hematological malignancies.
Collapse
Affiliation(s)
- Hu Lei
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jiaqi Wang
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiacheng Hu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Zhu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingli Wu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
40
|
Yan D, Li X, Yang Q, Huang Q, Yao L, Zhang P, Sun W, Lin S, Dou QP, Liu J, Chen X. Regulation of Bax-dependent apoptosis by mitochondrial deubiquitinase USP30. Cell Death Discov 2021; 7:211. [PMID: 34381024 PMCID: PMC8357812 DOI: 10.1038/s41420-021-00599-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/13/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022] Open
Abstract
Deubiquitinates (DUBs) have been suggested as novel promising targets for cancer therapies. Accumulating experimental evidence suggests that some metal compounds have the potential to induce cancer cell death via inhibition of DUBs. We previously reported that auranofin, a gold(I)-containing agent used for the treatment of rheumatoid arthritis in clinics, can induce cell death by inhibiting proteasomal DUBs in a series of cancer cell lines. Unfortunately, currently available gold compounds are not potent in inhibiting DUBs. Here, we report that: (i) aumdubin, a synthetic derivative of auranofin, exhibited stronger DUB-inhibiting and apoptosis-inducing activities than auranofin in lung cancer cells; (ii) aumdubin shows high affinity for mitochondrial DUB USP30; (iii) aumdubin induces apoptosis by increasing the ubiquitination and mitochondrial location of Bax protein; and (iv) USP30 inhibition may contribute to Bax-dependent apoptosis induced by aumdubin in lung cancer cells. These results suggest that gold(I)-containing agent aumdubin induces Bax-dependent apoptosis partly through inhibiting the mitochondrial DUB USP30, which could open new avenues for lung cancer therapy.
Collapse
Affiliation(s)
- Ding Yan
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaofen Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qianqian Yang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qingtian Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Leyi Yao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Peiquan Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wenshuang Sun
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shuhui Lin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Q Ping Dou
- Barbara Ann Karmanos Cancer Institute and Departments of Oncology, Pharmacology & Pathology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China. .,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xin Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China. .,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
41
|
Do HA, Baek KH. Cellular functions regulated by deubiquitinating enzymes in neurodegenerative diseases. Ageing Res Rev 2021; 69:101367. [PMID: 34023421 DOI: 10.1016/j.arr.2021.101367] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022]
Abstract
Neurodegenerative diseases are one of the most common diseases in mankind. Although there are reports of several candidates that cause neurodegenerative diseases, the exact mechanism of pathogenesis is poorly understood. The ubiquitin-proteasome system (UPS) is an important posttranslational modification for protein degradation and control of homeostasis. Enzymes such as E1, E2, E3 ligases, and deubiquitinating enzymes (DUBs) participating in UPS, regulate disease-inducing proteins by controlling the degree of ubiquitination. Therefore, the development of treatments targeting enzymes for degenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS), is emerging as an attractive perspective. In particular, as DUBs are able to regulate one or more degenerative disease-related proteins, the potential as a therapeutic target is even more evident. DUBs influence the regulation of toxic proteins that cause neurodegenerative diseases by not only their removal, but also by regulating signals associated with mitophagy, autophagy, and endoplasmic reticulum-associated degradation (ERAD). In this review, we analyze not only the cellular processes of DUBs, which control neurodegenerative disease-inducing proteins, but also their potentials as a therapeutic agent for neurodegenerative diseases.
Collapse
|
42
|
Exploiting the reactive oxygen species imbalance in high-risk paediatric acute lymphoblastic leukaemia through auranofin. Br J Cancer 2021; 125:55-64. [PMID: 33837299 PMCID: PMC8257682 DOI: 10.1038/s41416-021-01332-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 01/31/2021] [Accepted: 02/19/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The prognosis for high-risk childhood acute leukaemias remains dismal and established treatment protocols often cause long-term side effects in survivors. This study aims to identify more effective and safer therapeutics for these patients. METHODS A high-throughput phenotypic screen of a library of 3707 approved drugs and pharmacologically active compounds was performed to identify compounds with selective cytotoxicity against leukaemia cells followed by further preclinical evaluation in patient-derived xenograft models. RESULTS Auranofin, an FDA-approved agent for the treatment of rheumatoid arthritis, was identified as exerting selective anti-cancer activity against leukaemia cells, including patient-derived xenograft cells from children with high-risk ALL, versus solid tumour and non-cancerous cells. It induced apoptosis in leukaemia cells by increasing reactive oxygen species (ROS) and potentiated the activity of the chemotherapeutic cytarabine against highly aggressive models of infant MLL-rearranged ALL by enhancing DNA damage accumulation. The enhanced sensitivity of leukaemia cells towards auranofin was associated with lower basal levels of the antioxidant glutathione and higher baseline ROS levels compared to solid tumour cells. CONCLUSIONS Our study highlights auranofin as a well-tolerated drug candidate for high-risk paediatric leukaemias that warrants further preclinical investigation for application in high-risk paediatric and adult acute leukaemias.
Collapse
|
43
|
Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases. Int J Mol Sci 2021; 22:ijms22126213. [PMID: 34207520 PMCID: PMC8226605 DOI: 10.3390/ijms22126213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 02/06/2023] Open
Abstract
The 26S proteasome is the principal protease for regulated intracellular proteolysis. This multi-subunit complex is also pivotal for clearance of harmful proteins that are produced throughout the lifetime of eukaryotes. Recent structural and kinetic studies have revealed a multitude of conformational states of the proteasome in substrate-free and substrate-engaged forms. These conformational transitions demonstrate that proteasome is a highly dynamic machinery during substrate processing that can be also controlled by a number of proteasome-associated factors. Essentially, three distinct family of deubiquitinases–USP14, RPN11, and UCH37–are associated with the 19S regulatory particle of human proteasome. USP14 and UCH37 are capable of editing ubiquitin conjugates during the process of their dynamic engagement into the proteasome prior to the catalytic commitment. In contrast, RPN11-mediated deubiquitination is directly coupled to substrate degradation by sensing the proteasome’s conformational switch into the commitment steps. Therefore, proteasome-bound deubiquitinases are likely to tailor the degradation events in accordance with substrate processing steps and for dynamic proteolysis outcomes. Recent chemical screening efforts have yielded highly selective small-molecule inhibitors for targeting proteasomal deubiquitinases, such as USP14 and RPN11. USP14 inhibitors, IU1 and its progeny, were found to promote the degradation of a subset of substrates probably by overriding USP14-imposed checkpoint on the proteasome. On the other hand, capzimin, a RPN11 inhibitor, stabilized the proteasome substrates and showed the anti-proliferative effects on cancer cells. It is highly conceivable that these specific inhibitors will aid to dissect the role of each deubiquitinase on the proteasome. Moreover, customized targeting of proteasome-associated deubiquitinases may also provide versatile therapeutic strategies for induced or repressed protein degradation depending on proteolytic demand and cellular context.
Collapse
|
44
|
Martínez-Noël G, Vieira VC, Szajner P, Lilienthal EM, Kramer RE, Boyland KA, Smith JA, Howley PM. Live cell, image-based high-throughput screen to quantitate p53 stabilization and viability in human papillomavirus positive cancer cells. Virology 2021; 560:96-109. [PMID: 34051479 DOI: 10.1016/j.virol.2021.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/16/2021] [Accepted: 05/16/2021] [Indexed: 11/16/2022]
Abstract
Approximately 5% of cancers are caused by high-risk human papillomaviruses. Although very effective preventive vaccines will reduce this cancer burden significantly over the next several decades, they have no therapeutic effect for those already infected and remaining at risk for malignant progression of hrHPV lesions. HPV-associated cancers are dependent upon the expression of the viral E6 and E7 oncogenes. The oncogenic function of hrHPV E6 relies partially on its ability to induce p53 degradation. Since p53 is generally wildtype in hrHPV-associated cancers, p53 stabilization arrests proliferation, induces apoptosis and/or results in senescence. Here we describe a live cell, image-based high-throughput screen to identify compounds that stabilize p53 and/or affect viability in HPV-positive cancer HeLa cells. We validate the robustness and potential of this screening assay by assessing the activities of approximately 6,500 known bioactive compounds, illustrating its capability to function as a platform to identify novel therapeutics for hrHPV.
Collapse
Affiliation(s)
- Gustavo Martínez-Noël
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Valdimara Corrêa Vieira
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Patricia Szajner
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Erin M Lilienthal
- ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Rebecca E Kramer
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Kathleen A Boyland
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Jennifer A Smith
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA; ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Peter M Howley
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| |
Collapse
|
45
|
Singh VK, Seed TM. Repurposing Pharmaceuticals Previously Approved by Regulatory Agencies to Medically Counter Injuries Arising Either Early or Late Following Radiation Exposure. Front Pharmacol 2021; 12:624844. [PMID: 34040517 PMCID: PMC8141805 DOI: 10.3389/fphar.2021.624844] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing risks of radiological or nuclear attacks or associated accidents have served to renew interest in developing radiation medical countermeasures. The development of prospective countermeasures and the subsequent gain of Food and Drug Administration (FDA) approval are invariably time consuming and expensive processes, especially in terms of generating essential human data. Due to the limited resources for drug development and the need for expedited drug approval, drug developers have turned, in part, to the strategy of repurposing agents for which safety and clinical data are already available. Approval of drugs that are already in clinical use for one indication and are being repurposed for another indication is inherently faster and more cost effective than for new agents that lack regulatory approval of any sort. There are four known growth factors which have been repurposed in the recent past as radiomitigators following the FDA Animal Rule: Neupogen, Neulasta, Leukine, and Nplate. These four drugs were in clinic for several decades for other indications and were repurposed. A large number of additional agents approved by various regulatory authorities for given indications are currently under investigation for dual use for acute radiation syndrome or for delayed pathological effects of acute radiation exposure. The process of drug repurposing, however, is not without its own set of challenges and limitations.
Collapse
Affiliation(s)
- Vijay K. Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | | |
Collapse
|
46
|
Advances in the Development Ubiquitin-Specific Peptidase (USP) Inhibitors. Int J Mol Sci 2021; 22:ijms22094546. [PMID: 33925279 PMCID: PMC8123678 DOI: 10.3390/ijms22094546] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Ubiquitylation and deubiquitylation are reversible protein post-translational modification (PTM) processes involving the regulation of protein degradation under physiological conditions. Loss of balance in this regulatory system can lead to a wide range of diseases, such as cancer and inflammation. As the main members of the deubiquitinases (DUBs) family, ubiquitin-specific peptidases (USPs) are closely related to biological processes through a variety of molecular signaling pathways, including DNA damage repair, p53 and transforming growth factor-β (TGF-β) pathways. Over the past decade, increasing attention has been drawn to USPs as potential targets for the development of therapeutics across diverse therapeutic areas. In this review, we summarize the crucial roles of USPs in different signaling pathways and focus on advances in the development of USP inhibitors, as well as the methods of screening and identifying USP inhibitors.
Collapse
|
47
|
Chen X, Dou QP, Liu J, Tang D. Targeting Ubiquitin-Proteasome System With Copper Complexes for Cancer Therapy. Front Mol Biosci 2021; 8:649151. [PMID: 33928122 PMCID: PMC8076789 DOI: 10.3389/fmolb.2021.649151] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Characterizing mechanisms of protein homeostasis, a process of balancing between protein synthesis and protein degradation, is important for understanding the potential causes of human diseases. The ubiquitin–proteasome system (UPS) is a well-studied mechanism of protein catabolism, which is responsible for eliminating misfolded, damaged, or aging proteins, thereby maintaining quality and quantity of cellular proteins. The UPS is composed of multiple components, including a series of enzymes (E1, E2, E3, and deubiquitinase [DUB]) and 26S proteasome (19S regulatory particles + 20S core particle). An impaired UPS pathway is involved in multiple diseases, including cancer. Several proteasome inhibitors, such as bortezomib, carfilzomib, and ixazomib, are approved to treat patients with certain cancers. However, their applications are limited by side effects, drug resistance, and drug–drug interactions observed in their clinical processes. To overcome these shortcomings, alternative UPS inhibitors have been searched for in many fields. Copper complexes (e.g., CuET, CuHQ, CuCQ, CuPDTC, CuPT, and CuHK) are found to be able to inhibit a core component of the UPS machinery, such as 20S proteasome, 19S DUBs, and NPLOC4/NPL4 complex, and are proposed to be one class of metal-based anticancer drugs. In this review, we will summarize functions and applications of copper complexes in a concise perspective, with a focus on connections between the UPS and cancer.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Q Ping Dou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Oncology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States.,Departments of Pharmacology & Pathology, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
48
|
|
49
|
Ghini V, Senzacqua T, Massai L, Gamberi T, Messori L, Turano P. NMR reveals the metabolic changes induced by auranofin in A2780 cancer cells: evidence for glutathione dysregulation. Dalton Trans 2021; 50:6349-6355. [PMID: 33885689 DOI: 10.1039/d1dt00750e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
NMR metabolomics represents a powerful tool to characterize the cellular effects of drugs and gain detailed insight into their mode of action. Here, we have exploited NMR metabolomics to illustrate the changes in the metabolic profile of A2780 ovarian cancer cells elicited by auranofin (AF), a clinically approved gold drug now repurposed as an anticancer agent. An early and large increase in intracellular glutathione is highlighted as the main effect of the treatment accompanied by small but significant changes in the levels of a few additional metabolites; the general implications of these findings are discussed in the frame of the current mechanistic knowledge of AF.
Collapse
Affiliation(s)
- Veronica Ghini
- Center of Magnetic Resonance, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy. and Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy
| | - Tommaso Senzacqua
- Center of Magnetic Resonance, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Lara Massai
- Department of Chemistry, University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, viale Morgagni 50, Firenze, Italy
| | - Luigi Messori
- Department of Chemistry, University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Paola Turano
- Center of Magnetic Resonance, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy. and Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy and Department of Chemistry, University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Firenze, Italy.
| |
Collapse
|
50
|
Targeted inhibition of cooperative mutation- and therapy-induced AKT activation in AML effectively enhances response to chemotherapy. Leukemia 2020; 35:2030-2042. [PMID: 33299144 DOI: 10.1038/s41375-020-01094-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/30/2020] [Accepted: 11/09/2020] [Indexed: 11/09/2022]
Abstract
Most AML patients exhibit mutational activation of the PI3K/AKT signaling pathway, which promotes downstream effects including growth, survival, DNA repair, and resistance to chemotherapy. Herein we demonstrate that the inv(16)/KITD816Y AML mouse model exhibits constitutive activation of PI3K/AKT signaling, which was enhanced by chemotherapy-induced DNA damage through DNA-PK-dependent AKT phosphorylation. Strikingly, inhibitors of either PI3K or DNA-PK markedly reduced chemotherapy-induced AKT phosphorylation and signaling leading to increased DNA damage and apoptosis of inv(16)/KITD816Y AML cells in response to chemotherapy. Consistently, combinations of chemotherapy and PI3K or DNA-PK inhibitors synergistically inhibited growth and survival of clonogenic AML cells without substantially inhibiting normal clonogenic bone marrow cells. Moreover, treatment of inv(16)/KITD816Y AML mice with combinations of chemotherapy and PI3K or DNA-PK inhibitors significantly prolonged survival compared to untreated/single-treated mice. Mechanistically, our findings implicate that constitutive activation of PI3K/AKT signaling driven by mutant KIT, and potentially other mutational activators such as FLT3 and RAS, cooperates with chemotherapy-induced DNA-PK-dependent activation of AKT to promote survival, DNA repair, and chemotherapy resistance in AML. Hence, our study provides a rationale to select AML patients exhibiting constitutive PI3K/AKT activation for simultaneous treatment with chemotherapy and inhibitors of DNA-PK and PI3K to improve chemotherapy response and clinical outcome.
Collapse
|