151
|
Yamashita A, Park SH, Zeng L, Stiles WR, Ahn S, Bao K, Kim J, Kang H, Choi HS. H-Dot Mediated Nanotherapeutics Mitigate Systemic Toxicity of Platinum-Based Anticancer Drugs. Int J Mol Sci 2023; 24:15466. [PMID: 37895146 PMCID: PMC10607179 DOI: 10.3390/ijms242015466] [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: 09/27/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Platinum-based anticancer agents have revolutionized oncological treatments globally. However, their therapeutic efficacy is often accompanied by systemic toxicity. Carboplatin, recognized for its relatively lower toxicity profile than cisplatin, still presents off-target toxicities, including dose-dependent cardiotoxicity, neurotoxicity, and myelosuppression. In this study, we demonstrate a delivery strategy of carboplatin to mitigate its off-target toxicity by leveraging the potential of zwitterionic nanocarrier, H-dot. The designed carboplatin/H-dot complex (Car/H-dot) exhibits rapid drug release kinetics and notable accumulation in proximity to tumor sites, indicative of amplified tumor targeting precision. Intriguingly, the Car/H-dot shows remarkable efficacy in eliminating tumors across insulinoma animal models. Encouragingly, concerns linked to carboplatin-induced cardiotoxicity are effectively alleviated by adopting the Car/H-dot nanotherapeutic approach. This pioneering investigation not only underscores the viability of H-dot as an organic nanocarrier for platinum drugs but also emphasizes its pivotal role in ameliorating associated toxicities. Thus, this study heralds a promising advancement in refining the therapeutic landscape of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Atsushi Yamashita
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
| | - Seung Hun Park
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
| | - Lingxue Zeng
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
- Department of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Wesley R. Stiles
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
| | - Sung Ahn
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
| | - Kai Bao
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
| | - Jonghan Kim
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
- Department of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.Y.); (S.H.P.); (L.Z.); (W.R.S.); (S.A.); (K.B.); (J.K.); (H.K.)
| |
Collapse
|
152
|
Li C, Zhao X, Yin F, Bi H, Wang Y, Xie P. Structural changes in DNA by binding mitochondrion-targeted monofunctional platinum(II) complexes using molecular dynamics simulation study. J Inorg Biochem 2023; 250:112419. [PMID: 39492371 DOI: 10.1016/j.jinorgbio.2023.112419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/12/2023] [Accepted: 10/21/2023] [Indexed: 11/05/2024]
Abstract
Triphenylphosphonium (Ph3P+, TPP) is a highly effective mitochondrial targeting group, an example of using which on mitochondrion-targeted monofunctional platinum(II) agent as anticancer drug was OPT, with the -CH2Ph3P+ group at ortho position of the pyriplatin pyridine ring. To study how carrier ligands might affect the efficacy of OPT, we constructed two platinum(II) agents with bulky bidentate ligands based on OPT. DNA structural changes caused by these three platinum(II) agents using molecular dynamics simulations were analysed. Data regarding DNA conformational changes including helical parameter, base stacking, average structure, and principal component analyses has been obtained. We found that TPP-based monofunctional platinum(II) complexes with bulky carrier ligands may induce more significant DNA conformational changes. These results are beneficial for developing highly efficient mitochondrion-targeted platinum anticancer drugs with carrier ligands of different steric hindrance.
Collapse
Affiliation(s)
- Chaoqun Li
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemistry, Chemical Engineering and Materials, Handan University, Handan, 056005, Hebei province, China.
| | - Xiaojia Zhao
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemistry, Chemical Engineering and Materials, Handan University, Handan, 056005, Hebei province, China.
| | - Fangqian Yin
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemistry, Chemical Engineering and Materials, Handan University, Handan, 056005, Hebei province, China
| | - Huimin Bi
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemistry, Chemical Engineering and Materials, Handan University, Handan, 056005, Hebei province, China
| | - Yan Wang
- College of Chemistry, Beijing Normal University, 19# Xinjiekouwai Street, Beijing 100875, China
| | - Pengtao Xie
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemistry, Chemical Engineering and Materials, Handan University, Handan, 056005, Hebei province, China
| |
Collapse
|
153
|
Yang L, Liu YN, Gu Y, Guo Q. Deltonin enhances gastric carcinoma cell apoptosis and chemosensitivity to cisplatin via inhibiting PI3K/AKT/mTOR and MAPK signaling. World J Gastrointest Oncol 2023; 15:1739-1755. [PMID: 37969408 PMCID: PMC10631430 DOI: 10.4251/wjgo.v15.i10.1739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/23/2023] [Accepted: 07/19/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND As an active ingredient derived from Dioscorea zingiberensis C.H. Wright, deltonin has been reported to show anti-cancer effects in a variety of malignancies. AIM To investigate the role and mechanism of action of deltonin in promoting gastric carcinoma (GC) cell apoptosis and chemosensitivity to cisplatin. METHODS The GC cell lines AGS, HGC-27, and MKN-45 were treated with deltonin and then subjected to flow cytometry and 3-(4,5-dimethylthiazol-2-yl)-3,5-diphenyltetrazolium bromide assays for cell apoptosis and viability determination. Western blot analysis was conducted to examine alterations in the expression of apoptosis-related proteins (Bax, Bid, Bad, and Fas), DNA repair-associated proteins (Rad51 and MDM2), and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of the rapamycin (PI3K/AKT/mTOR) and p38-mitogen-activated protein kinase (MAPK) axis proteins. Additionally, the influence of deltonin on GC cell chemosensitivity to cisplatin was evaluated both in vitro and in vivo. RESULTS Deltonin treatment weakened viability, enhanced apoptosis, and dampened DNA repair in GC cell lines in a dose-dependent pattern. Furthermore, deltonin mitigated PI3K, AKT, mTOR, and p38-MAPK phosphorylation. HS-173, an inhibitor of PI3K, attenuated GC cell viability and abolished deltonin inhibition of GC cell viability and PI3K/AKT/mTOR and p38-MAPK pathway activation. Deltonin also promoted the chemosensitivity of GC cells to cisplatin via repressing GC cell proliferation and growth and accelerating apoptosis. CONCLUSION Deltonin can boost the chemosensitivity of GC cells to cisplatin via inactivating p38-MAPK and PI3K/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Lin Yang
- Intensive Care Unit, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ya-Nan Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yi Gu
- Nursing Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qi Guo
- Department of Radiotherapy, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
154
|
Chandra J, Hasan N, Nasir N, Wahab S, Thanikachalam PV, Sahebkar A, Ahmad FJ, Kesharwani P. Nanotechnology-empowered strategies in treatment of skin cancer. ENVIRONMENTAL RESEARCH 2023; 235:116649. [PMID: 37451568 DOI: 10.1016/j.envres.2023.116649] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
In current scenario skin cancer is a serious condition that has a significant impact on world health. Skin cancer is divided into two categories: melanoma skin cancer (MSC) and non-melanoma skin cancer (NMSC). Because of its significant psychosocial effects and need for significant investment in new technology and therapies, skin cancer is an illness of global health relevance. From the patient's perspective chemotherapy considered to be the most acceptable form of treatment. However, significant negatives of chemotherapy such as severe toxicities and drug resistance pose serious challenges to the treatment. The field of nanomedicine holds significant promise for enhancing the specificity of targeting neoplastic cells through the facilitation of targeted drug delivery to tumour cells. The integration of multiple therapeutic modalities to selectively address cancer-promoting or cell-maintaining pathways constitutes a fundamental aspect of cancer treatment. The use of mono-therapy remains prevalent in the treatment of various types of cancer, it is widely acknowledged in the academic community that this conventional approach is generally considered to be less efficacious compared to the combination treatment strategy. The employment of combination therapy in cancer treatment has become increasingly widespread due to its ability to produce synergistic anticancer effects, mitigate toxicity associated with drugs, and inhibit multi-drug resistance by means of diverse mechanisms. Nanotechnology based combination therapy represents a promising avenue for the development of efficacious therapies for skin cancer within the context of this endeavour. The objective of this article is to provide a description of distinct challenges for efficient delivery of drugs via skin. This article also provides a summary of the various nanotechnology based combinatorial therapy available for skin cancer with their recent advances. This review also focuses on current status of clinical trials of such therapies.
Collapse
Affiliation(s)
- Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazim Nasir
- Department of Basic Medical Sciences, College of Applied Medical Sciences, Khamis Mushait, Kingdom of Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 61421, Saudi Arabia
| | - Punniyakoti Veeraveedu Thanikachalam
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
155
|
Li Y, Wang B, Ahmad Khan Z, He J, Cheung E, Su W, Wang A, Jiang H, Jiang L, Ding X. Platinum-Chimeric Carrier Cells for Ultratrace Cell Analysis in Mass Cytometry. Anal Chem 2023; 95:14998-15007. [PMID: 37767956 DOI: 10.1021/acs.analchem.3c02706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Mass cytometry by time-of-flight (CyTOF), a high-dimensional single-cell analysis platform, detects up to 50 biomarkers at single-cell resolution. However, CyTOF analysis of biological samples with a minimal number of available cells or rare cell subsets remains a major technical challenge due to the extensive loss of cells during cell recovery, staining, and acquisition. Here, we introduce a platinum-chimeric carrier cell strategy for mass cytometry profiling of ultratrace cell samples. Cisplatin can rapidly enter broken plasma membranes of dead cells and form a chimeric interaction with cellular proteins, peptides, and amino acids. Thus, 198Pt-cisplatin is adopted to tag carrier cells in the pretreatment stage. We investigated 8 cell lines that are commonly accessible in laboratories for their potential as carrier cells to preserve rare target cells for CyTOF analysis. We designed a panel of 35 protein biomarkers to evaluate the comprehensive single-cell subtype classification capability with or without the carrier cell strategy. We further demonstrated the detection and analysis of as few as 1 × 104 immune cells using our method. The proposed method thus allows CyTOF analysis on precious clinical samples with less abundant cells.
Collapse
Affiliation(s)
- Yiyang Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Boqian Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Zara Ahmad Khan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Jie He
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Edwin Cheung
- Cancer Centre, University of Macau, Taipa 999078, Macau SAR
- Centre for Precision Medicine Research and Training, University of Macau, Taipa 999078, Macau SAR
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa 999078, Macau SAR
- Faculty of Health Sciences, University of Macau, Taipa 999078, Macau SAR
| | - Wenqiong Su
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Aiting Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Hui Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| |
Collapse
|
156
|
Kavukcu S, Ensarioğlu HK, Karabıyık H, Vatansever HS, Türkmen H. Cell Death Mechanism of Organometallic Ruthenium(II) and Iridium(III) Arene Complexes on HepG2 and Vero Cells. ACS OMEGA 2023; 8:37549-37563. [PMID: 37841164 PMCID: PMC10569012 DOI: 10.1021/acsomega.3c05898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023]
Abstract
Due to side effects and toxicity associated with platinum-derived metal-based drugs, extensive research has been conducted on ruthenium (Ru) complexes. We aim to synthesize a highly oil soluble Ru(II)-p-cymene complex (Ru1) with an aliphatic chain group, a bimetallic Ru(II)-p-cymene complex (Ru2) with N,S,S triple-coordination and a bimetallic Ir(III)-pentamethylcyclopentadienyl complex (Ir1) with S,S double-coordination. Subsequently, we investigate the effects of these complexes on Vero and HepG2 cell lines, focusing on cell death mechanisms. Characterization of the complexes is performed through nuclear magnetic resonance spectroscopy (1H and 13C NMR) and Fourier-transform infrared spectroscopy. The effective doses are determined using the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) (MTT) assay, applying different doses of the complexes to the two cell lines for 24 and 48 h, respectively. Immunoreactivities of Bax, Bcl2, caspase-3, RIP3, and RIPK1 are analyzed using the indirect immunoperoxidase technique. Notably, all the complexes (Ru1, Ru2, and Ir1) exhibit distinct cell death mechanisms, showing greater effectiveness than cisplatin. This study reveals the diverse mechanisms of action of Ru and Ir complexes based on different ligands. To the best of our knowledge, this study represents the first investigation of a novel RAED-type complex (Ru1) and unexpected bimetallic complexes (Ru2 and Ir1).
Collapse
Affiliation(s)
| | - Hilal Kabadayı Ensarioğlu
- Manisa
Celal Bayar University, Faculty of Medicine,
Department of Histology and Embryology, Manisa 45030, Turkey
| | - Hande Karabıyık
- Dokuz
Eylül University, Faculty of Science,
Department of Physics, Izmir 35390, Turkey
| | - Hafize Seda Vatansever
- Manisa
Celal Bayar University, Faculty of Medicine,
Department of Histology and Embryology, Manisa 45030, Turkey
- Near
East University, DESAM Institute, Mersin 10, Turkey 99138
| | - Hayati Türkmen
- Ege
University, Faculty of Science,
Department of Chemistry, Izmir 35100, Turkey
| |
Collapse
|
157
|
Guerrero AS, O'Dowd PD, Pigg HC, Alley KR, Griffith DM, DeRose VJ. Comparison of click-capable oxaliplatin and cisplatin derivatives to better understand Pt(ii)-induced nucleolar stress. RSC Chem Biol 2023; 4:785-793. [PMID: 37799581 PMCID: PMC10549245 DOI: 10.1039/d3cb00055a] [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: 04/18/2023] [Accepted: 08/16/2023] [Indexed: 10/07/2023] Open
Abstract
Pt(ii) chemotherapeutic complexes have been used as predominant anticancer drugs for nearly fifty years. Currently there are three FDA-approved chemotherapeutic Pt(ii) complexes: cisplatin, carboplatin, and oxaliplatin. Until recently, it was believed that all three complexes induced cellular apoptosis through the DNA damage response pathway. Studies within the last decade, however, suggest that oxaliplatin may instead induce cell death through a unique nucleolar stress pathway. Pt(ii)-induced nucleolar stress is not well understood and further investigation of this pathway may provide both basic knowledge about nucleolar stress as well as insight for more tunable Pt(ii) chemotherapeutics. Through a previous structure-function analysis, it was determined that nucleolar stress induction is highly sensitive to modifications at the 4-position of the 1,2-diaminocyclohexane (DACH) ring of oxaliplatin. Specifically, more flexible and less rigid substituents (methyl, ethyl, propyl) induce nucleolar stress, while more rigid and bulkier substituents (isopropyl, acetamide) do not. These findings suggest that a click-capable functional group can be installed at the 4-position of the DACH ring while still inducing nucleolar stress. Herein, we report novel click-capable azide-modified oxaliplatin mimics that cause nucleolar stress. Through NPM1 relocalization, fibrillarin redistribution, and γH2AX studies, key differences have been identified between previously studied click-capable cisplatin mimics and these novel click-capable oxaliplatin mimics. These complexes provide new tools to identify cellular targets and localization through post-treatment Cu-catalyzed azide-alkyne cycloaddition and may help to better understand Pt(ii)-induced nucleolar stress. To our knowledge, these are the first reported oxaliplatin mimics to include an azide handle, and cis-[(1R,2R,4S) 4-methylazido-1,2-cyclohexanediamine]dichlorido platinum(ii) is the first azide-functionalized oxaliplatin derivative to induce nucleolar stress.
Collapse
Affiliation(s)
- Andres S Guerrero
- Department of Chemistry and Biochemistry, University of Oregon Eugene OR USA
| | - Paul D O'Dowd
- Department of Chemistry, RCSI Dublin Ireland
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals Ireland
| | - Hannah C Pigg
- Department of Chemistry and Biochemistry, University of Oregon Eugene OR USA
| | - Katelyn R Alley
- Department of Chemistry and Biochemistry, University of Oregon Eugene OR USA
| | - Darren M Griffith
- Department of Chemistry, RCSI Dublin Ireland
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals Ireland
| | - Victoria J DeRose
- Department of Chemistry and Biochemistry, University of Oregon Eugene OR USA
| |
Collapse
|
158
|
Zhao P, Guo C, Du H, Xiao Y, Su J, Wang X, Yeung WSB, Li G, Wang T. Chemotherapy-induced ovarian damage and protective strategies. HUM FERTIL 2023; 26:887-900. [PMID: 38054300 DOI: 10.1080/14647273.2023.2275764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/14/2023] [Indexed: 12/07/2023]
Abstract
More than 9.2 million women worldwide suffer from cancer, and about 5% of them are at reproductive age. Chemotherapy-induced impairment of fertility affects the quality of life of these women. Several chemotherapeutic agents have been proven to cause apoptosis and autophagy by inducing DNA damage and cellular stress. Injuries to the ovarian stroma and micro-vessel network are also considered as pivotal factors resulting in ovarian dysfunction induced by chemotherapeutic agents. Primordial follicle pool over-activation may also be the mechanism inducing damage to the ovarian reserve. Although many studies have explored the mechanisms involved in chemotherapy-induced reproductive toxicity, the exact molecular mechanisms have not been elucidated. It is essential to understand the mechanisms involved in ovarian damage, in order to develop potential protective treatments to preserve fertility. In this article, we reviewed the current knowledge on the mechanism of chemotherapy-induced ovarian damage and possible protective strategies that prevent the ovary from such damages.
Collapse
Affiliation(s)
- Peikun Zhao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Chenxi Guo
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Huijia Du
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Yuan Xiao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Jiaping Su
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Xiaohui Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Willian S B Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Guangxin Li
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, PR China
| | - Tianren Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| |
Collapse
|
159
|
Tan H, Wang F, Hu J, Duan X, Bai W, Wang X, Wang B, Su Y, Hu J. Inhibitory interaction of flavonoids with organic cation transporter 2 and their structure-activity relationships for predicting nephroprotective effects. J Appl Toxicol 2023; 43:1421-1435. [PMID: 37057715 DOI: 10.1002/jat.4474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
Organic cation transporter 2 (OCT2) is mainly responsible for the renal secretion of various cationic drugs, closely associated with drug-induced acute kidney injury (AKI). Screening and identifying potent OCT2 inhibitors with little toxicity in natural products in reducing OCT2-mediated AKI is of great value. Flavonoids are enriched in various vegetables, fruits, and herbal products, and some were reported to produce transporter-mediated drug-drug interactions. This study aimed to screen potential inhibitors of OCT2 from 96 flavonoids, assess the nephroprotective effects on cisplatin-induced kidney injury, and clarify the structure-activity relationships of flavonoids with OCT2. Ten flavonoids exhibited significant inhibition (>50%) on OCT2 in OCT2-HEK293 cells. Among them, the six most potent flavonoid inhibitors, including pectolinarigenin, biochanin A, luteolin, chrysin, 6-hydroxyflavone, and 6-methylflavone markedly decreased cisplatin-induced cytotoxicity. Moreover, in cisplatin-induced renal injury models, they also reduced serum blood urea nitrogen (BUN) and creatinine levels to different degrees, the best of which was 6-methylflavone. The pharmacophore model clarified that the aromatic ring, hydrogen bond acceptors, and hydrogen bond donors might play a vital role in the inhibitory effect of flavonoids on OCT2. Thus, our findings would pave the way to predicting the potential risks of flavonoid-containing food/herb-drug interactions in humans and optimizing flavonoid structure to alleviate OCT2-related AKI.
Collapse
Affiliation(s)
- Huixin Tan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Fenghe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiahuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xiaoyan Duan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Wanting Bai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xinbo Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Baolian Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yan Su
- Department of Health Management and Service, Cangzhou Medical College, Hebei, 061001, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
160
|
Balsa LM, Solernó LM, Rodriguez MR, Parajón-Costa BS, Gonzalez-Baró AC, Alonso DF, Garona J, León IE. Cu(II)-acylhydrazone complex, a potent and selective antitumor agent against human osteosarcoma: Mechanism of action studies over in vitro and in vivo models. Chem Biol Interact 2023; 384:110685. [PMID: 37666443 DOI: 10.1016/j.cbi.2023.110685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/05/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023]
Abstract
Osteosarcoma (OS) is a frequent bone cancer, affecting largely children and young adults. Cisplatin (CDDP) has been efficacious in the treatment of different cancer such us OS but the development of chemoresistance and important side effects leading to therapeutic failure. Novel therapies including copper compounds have shown to be potentially effective as anticancer drugs and one alternative to usually employed platinum compounds. The goal of this work is the evaluation of the in vitro and in vivo antitumoral activity and dilucidate the molecular target of a Cu(II) cationic complex containing a tridentate hydrazone ligand, CuHL for short, H2L=N'-'-(2-hydroxy-3-methoxybenzylidene)thiophene-2-carbohydrazide, against human OS MG-63 cells. Anticancer activity on MG-63 cell line was evaluated in OS monolayer and spheroids. CuHL significantly impaired cell viability in both models (IC50 2D: 2.1 ± 0.3 μM; 3D: 9.1 ± 1.0 μM) (p < 0.001). Additional cell studies demonstrated the copper compound inhibits cell proliferation and conveys cells to apoptosis, determined by flow cytometry. CuHL showed a great genotoxicity, evaluated by comet assay. Proteomic analysis by Orbitrap Mass Spectometry identified 27 differentially expressed proteins: 17 proteins were found overexpressed and 10 underexpressed in MG-63 cells after the CuHL treatment. The response to unfolded protein was the most affected biological process. In addition, in vivo antitumor effects of the compound were evaluated on human OS tumors xenografted in nude mice. CuHL treatment, at a dose of 2 mg/kg i.p., given three times/week for one month, significantly inhibited the progression of OS xenografts and was associated to a reduction in mitotic index and to an increment of tumor necrosis (p < 0.01). Administration of standard-of-care cytotoxic agent CDDP, following the same treatment schedule as CuHL, failed to impair OS growth and progression.
Collapse
Affiliation(s)
- Lucia M Balsa
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, 1900, Argentina
| | - Luisina M Solernó
- Centro de Oncología Molecular y Traslacional (COMTra), Universidad Nacional de Quilmes, Argentina; Centro de Medicina Traslacional (Unidad 6), Hospital de Alta Complejidad en Red El Cruce "Dr. Néstor Carlos Kirchner" S.A.M.I.C, Argentina
| | - Maria R Rodriguez
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, 1900, Argentina
| | - Beatriz S Parajón-Costa
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, 1900, Argentina
| | - Ana C Gonzalez-Baró
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, 1900, Argentina
| | - Daniel F Alonso
- Centro de Oncología Molecular y Traslacional (COMTra), Universidad Nacional de Quilmes, Argentina
| | - Juan Garona
- Centro de Oncología Molecular y Traslacional (COMTra), Universidad Nacional de Quilmes, Argentina; Centro de Medicina Traslacional (Unidad 6), Hospital de Alta Complejidad en Red El Cruce "Dr. Néstor Carlos Kirchner" S.A.M.I.C, Argentina
| | - Ignacio E León
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, 1900, Argentina; Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, 1900, Argentina.
| |
Collapse
|
161
|
Rudd SG. Targeting pan-essential pathways in cancer with cytotoxic chemotherapy: challenges and opportunities. Cancer Chemother Pharmacol 2023; 92:241-251. [PMID: 37452860 PMCID: PMC10435635 DOI: 10.1007/s00280-023-04562-3] [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: 02/06/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
Cytotoxic chemotherapy remains a key modality in cancer treatment. These therapies, successfully used for decades, continue to transform the lives of cancer patients daily. With the high attrition rate of current oncology drug development, combined with the knowledge that most new therapies do not displace standard-of-care treatments and that many healthcare systems cannot afford these new therapies; cytotoxic chemotherapies will remain an important component of cancer therapy for many years to come. The clinical value of these therapies is often under-appreciated within the pre-clinical cancer research community, where this diverse class of agents are often grouped together as non-specific cellular poisons killing tumor cells based solely upon proliferation rate; however, this is inaccurate. This review article seeks to reaffirm the importance of focusing research efforts upon improving our basic understanding of how these drugs work, discussing their ability to target pan-essential pathways in cancer cells, the relationship of this to the chemotherapeutic window, and highlighting basic science approaches that can be employed towards refining their use.
Collapse
Affiliation(s)
- Sean G Rudd
- Science For Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
162
|
Jevtovic V, Alhar MSO, Milenković D, Marković Z, Dimitrić Marković J, Dimić D. Synthesis, Structural Characterization, Cytotoxicity, and Protein/DNA Binding Properties of Pyridoxylidene-Aminoguanidine-Metal (Fe, Co, Zn, Cu) Complexes. Int J Mol Sci 2023; 24:14745. [PMID: 37834192 PMCID: PMC10573062 DOI: 10.3390/ijms241914745] [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: 08/31/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Pyridoxylidene-aminoguanidine (PLAG) and its transition metal complexes are biologically active compounds with interesting properties. In this contribution, three new metal-PLAG complexes, Zn(PLAG)(SO4)(H2O)].∙H2O (Zn-PLAG), [Co(PLAG)2]SO4∙2H2O (Co-PLAG), and [Fe(PLAG)2]SO4∙2H2O) (Fe-PLAG), were synthetized and characterized by the X-ray crystallography. The intermolecular interactions governing the stability of crystal structure were compared to those of Cu(PLAG)(NCS)2 (Cu-PLAG) within Hirshfeld surface analysis. The structures were optimized at B3LYP/6-31+G(d,p)(H,C,N,O,S)/LanL2DZ (Fe,Co,Zn,Cu), and stability was assessed through Natural Bond Orbital Theory and Quantum Theory of Atoms in Molecules. Special emphasis was put on investigating the ligand's stability and reactivity. The binding of these compounds to Bovine and Human serum albumin was investigated by spectrofluorometric titration. The importance of complex geometry and various ligands for protein binding was shown. These results were complemented by the molecular docking study to elucidate the most important interactions. The thermodynamic parameters of the binding process were determined. The binding to DNA, as one of the main pathways in the cell death cycle, was analyzed by molecular docking. The cytotoxicity was determined towards HCT116, A375, MCF-7, and A2780 cell lines. The most active compound was Cu-PLAG due to the presence of PLAG and two thiocyanate ligands.
Collapse
Affiliation(s)
- Violeta Jevtovic
- Department of Chemistry, College of Science, University Ha’il, Ha’il 81451, Saudi Arabia
| | | | - Dejan Milenković
- Department of Science, Institute for Information Technologies, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Zoran Marković
- Department of Science, Institute for Information Technologies, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | | | - Dušan Dimić
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12–16, 11000 Belgrade, Serbia
| |
Collapse
|
163
|
Wang W, Wang P, Liao X, Yang B, Gao C, Yang J. A Series of Planar Phosphorescent Cyclometalated Platinum(II) Complexes as New Anticancer Theranostic Agents That Induce Oncosis. J Med Chem 2023; 66:13103-13115. [PMID: 37724909 DOI: 10.1021/acs.jmedchem.3c01126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Herein, four planar cyclometalated platinum(II) complexes with a main ligand of enlarged aromatic rings have been assessed as effective anticancer theranostic agents for the first time. With an increased number of aromatic rings in the N∧N ligand, 1a-1d exhibit increased lipophilicity and cytotoxicity selectivity. The intensity of the Pt-Pt interaction of each complex can be indicated by an enhanced near-infrared (NIR) emission in phosphate-buffered saline (PBS), their binding activity with biomolecules of bovine serum albumin (BSA) is accompanied by a vivid turn-on green emission, and the intensity gradually decreased from 1a to 1d, which is consistent with the docking of two complexes with BSA. Both "turn-on" NIR and green emission of 1d can be mainly observed in nuclei of living cell within 24 h, while two phosphorescence traces of 1b were recorded in lysosomes by confocal imaging. Moreover, 1d shows the highest efficiency in inducing oncosis of Hela cells, and the relative process was investigated.
Collapse
Affiliation(s)
- Wenting Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Pengchao Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Xiali Liao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Bo Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Chuanzhu Gao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Jing Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China
| |
Collapse
|
164
|
Becchetti A. Interplay of Ca 2+ and K + signals in cell physiology and cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:15-46. [PMID: 38007266 DOI: 10.1016/bs.ctm.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
The cytoplasmic Ca2+ concentration and the activity of K+ channels on the plasma membrane regulate cellular processes ranging from mitosis to oriented migration. The interplay between Ca2+ and K+ signals is intricate, and different cell types rely on peculiar cellular mechanisms. Derangement of these mechanisms accompanies the neoplastic progression. The calcium signals modulated by voltage-gated (KV) and calcium-dependent (KCa) K+ channel activity regulate progression of the cell division cycle, the release of growth factors, apoptosis, cell motility and migration. Moreover, KV channels regulate the cell response to the local microenvironment by assembling with cell adhesion and growth factor receptors. This chapter summarizes the pathophysiological roles of Ca2+ and K+ fluxes in normal and cancer cells, by concentrating on several biological systems in which these functions have been studied in depth, such as early embryos, mammalian cell lines, T lymphocytes, gliomas and colorectal cancer cells. A full understanding of the underlying mechanisms will offer a comprehensive view of the ion channel implication in cancer biology and suggest potential pharmacological targets for novel therapeutic approaches in oncology.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy.
| |
Collapse
|
165
|
Pincigher L, Valenti F, Bergamini C, Prata C, Fato R, Amorati R, Jin Z, Farruggia G, Fiorentini D, Calonghi N, Zalambani C. Myrcene: A Natural Compound Showing Anticancer Activity in HeLa Cells. Molecules 2023; 28:6728. [PMID: 37764505 PMCID: PMC10537210 DOI: 10.3390/molecules28186728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
γ-terpinene, α-terpinene, p-cymene, and myrcene are monoterpenes found in many essential oils extracted from a variety of plants and spices. Myrcene also occurs naturally in plants such as hops, cannabis, lemongrass, and verbena and is used as a flavoring agent in food and beverage manufacturing. In this research, the biological efficacy of γ-terpinene, α-terpinene, p-cymene, and myrcene was studied in human cell lines (HeLa, SH-SY5Y, and HDFa). Cytotoxicity, cell proliferation, cell migration, and morphology assays were performed to obtain detailed information on the anticancer properties. Our results show that myrcene has potential biological activity, especially in HeLa cells. In this cell line, it leads to an arrest of proliferation, a decrease in motility and morphological changes with loss of sphericity and thickness, and DNA damage. In addition, the interaction of γ-terpinene, α-terpinene, p-terpinene, and myrcene with calf thymus DNA (ct-DNA) was studied by UV-visible spectrophotometry. DNA binding experiments show that only myrcene can interact with DNA with an apparent dissociation constant (Kd) of 29 × 10-6 M.
Collapse
Affiliation(s)
- Luca Pincigher
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| | - Francesca Valenti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| | - Romana Fato
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| | - Riccardo Amorati
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Gobetti 83, 40129 Bologna, Italy; (R.A.); (Z.J.)
| | - Zongxin Jin
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Gobetti 83, 40129 Bologna, Italy; (R.A.); (Z.J.)
| | - Giovanna Farruggia
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
- National Institute of Biostructures and Biosystems, Via delle Medaglie d’Oro 305, 00136 Rome, Italy
| | - Diana Fiorentini
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| | - Chiara Zalambani
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (L.P.); (F.V.); (C.B.); (C.P.); (R.F.); (G.F.); (C.Z.)
| |
Collapse
|
166
|
Wang T, Shi S, Shi Y, Jiang P, Hu G, Ye Q, Shi Z, Yu K, Wang C, Fan G, Zhao S, Ma H, Chang ACY, Li Z, Bian Q, Lin CP. Chemical-induced phase transition and global conformational reorganization of chromatin. Nat Commun 2023; 14:5556. [PMID: 37689690 PMCID: PMC10492836 DOI: 10.1038/s41467-023-41340-4] [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: 01/27/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023] Open
Abstract
Chemicals or drugs can accumulate within biomolecular condensates formed through phase separation in cells. Here, we use super-resolution imaging to search for chemicals that induce phase transition within chromatin at the microscale. This microscopic screening approach reveals that adriamycin (doxorubicin) - a widely used anticancer drug that is known to interact with chromatin - specifically induces visible local condensation and global conformational change of chromatin in cancer and primary cells. Hi-C and ATAC-seq experiments systematically and quantitatively demonstrate that adriamycin-induced chromatin condensation is accompanied by weakened chromatin interaction within topologically associated domains, compartment A/B switching, lower chromatin accessibility, and corresponding transcriptomic changes. Mechanistically, adriamycin complexes with histone H1 and induces phase transition of H1, forming fibrous aggregates in vitro. These results reveal a phase separation-driven mechanism for a chemotherapeutic drug.
Collapse
Affiliation(s)
- Tengfei Wang
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Shuxiang Shi
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- Lingang Laboratory, 200031, Shanghai, China
| | - Yuanyuan Shi
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Peipei Jiang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Ganlu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Qinying Ye
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Zhan Shi
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Kexin Yu
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Chenguang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoping Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Suwen Zhao
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Hanhui Ma
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Alex C Y Chang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Li
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| |
Collapse
|
167
|
Borutzki Y, Skos L, Gerner C, Meier‐Menches SM. Exploring the Potential of Metal-Based Candidate Drugs as Modulators of the Cytoskeleton. Chembiochem 2023; 24:e202300178. [PMID: 37345897 PMCID: PMC10946712 DOI: 10.1002/cbic.202300178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/23/2023]
Abstract
During recent years, accumulating evidence suggested that metal-based candidate drugs are promising modulators of cytoskeletal and cytoskeleton-associated proteins. This was substantiated by the identification and validation of actin, vimentin and plectin as targets of distinct ruthenium(II)- and platinum(II)-based modulators. Despite this, structural information about molecular interaction is scarcely available. Here, we compile the scattered reports about metal-based candidate molecules that influence the cytoskeleton, its associated proteins and explore their potential to interfere in cancer-related processes, including proliferation, invasion and the epithelial-to-mesenchymal transition. Advances in this field depend crucially on determining binding sites and on gaining comprehensive insight into molecular drug-target interactions. These are key steps towards establishing yet elusive structure-activity relationships.
Collapse
Affiliation(s)
- Yasmin Borutzki
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Doctoral School of ChemistryUniversity of Vienna1090ViennaAustria
| | - Lukas Skos
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Doctoral School of ChemistryUniversity of Vienna1090ViennaAustria
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Samuel M. Meier‐Menches
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| |
Collapse
|
168
|
Zheng S, Li G, Shi J, Liu X, Li M, He Z, Tian C, Kamei KI. Emerging platinum(IV) prodrug nanotherapeutics: A new epoch for platinum-based cancer therapy. J Control Release 2023; 361:819-846. [PMID: 37597809 DOI: 10.1016/j.jconrel.2023.08.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Owing to the unique DNA damaging cytotoxicity, platinum (Pt)-based chemotherapy has long been the first-line choice for clinical oncology. Unfortunately, Pt drugs are restricted by the severe dose-dependent toxicity and drug resistance. Correspondingly, Pt(IV) prodrugs are developed with the aim to improve the antitumor performance of Pt drugs. However, as "free" molecules, Pt(IV) prodrugs are still subject to unsatisfactory in vivo destiny and antitumor efficacy. Recently, Pt(IV) prodrug nanotherapeutics, inheriting both the merits of Pt(IV) prodrugs and nanotherapeutics, have emerged and demonstrated the promise to address the underexploited dilemma of Pt-based cancer therapy. Herein, we summarize the latest fronts of emerging Pt(IV) prodrug nanotherapeutics. First, the basic outlines of Pt(IV) prodrug nanotherapeutics are overviewed. Afterwards, how versatile Pt(IV) prodrug nanotherapeutics overcome the multiple biological barriers of antitumor drug delivery is introduced in detail. Moreover, advanced combination therapies based on multimodal Pt(IV) prodrug nanotherapeutics are discussed with special emphasis on the synergistic mechanisms. Finally, prospects and challenges of Pt(IV) prodrug nanotherapeutics for future clinical translation are spotlighted.
Collapse
Affiliation(s)
- Shunzhe Zheng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guanting Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianbin Shi
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinying Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Meng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, Hangzhou 310058, China.
| | - Ken-Ichiro Kamei
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
169
|
Jeon J, Lee S, Park JM, Lee TH, Kang TH. Circadian control of cisplatin-DNA adduct repair and apoptosis in culture cells. Int J Biochem Cell Biol 2023; 162:106454. [PMID: 37574041 DOI: 10.1016/j.biocel.2023.106454] [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: 03/24/2023] [Revised: 07/02/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
Cisplatin, a widely prescribed chemotherapeutic agent for treating solid tumors, induces DNA adducts and activates cellular defense mechanisms, including DNA repair, cell cycle checkpoint control, and apoptosis. Considering the circadian rhythmicity displayed by most chemotherapeutic agents and their varying therapeutic efficacy based on treatment timing, our study aimed to investigate whether the circadian clock system influences the DNA damage responses triggered by cisplatin in synchronized cells. We examined the DNA damage responses in circadian-synchronized wild-type mouse embryonic fibroblasts (WT-MEF; clock-proficient cells), cryptochrome1 and 2 double knock-out MEF (CRYDKO; clock-deficient cells), and mouse hepatocarcinoma Hepa1c1c7 cells. Varying the treatment time resulted in a significant difference in the rate of platinum-DNA adduct removal specifically in circadian-synchronized WT-MEF, while CRYDKO did not exhibit such variation. Moreover, diurnal variation in other DNA damage responses, such as cell cycle checkpoint activity indicated by p53 phosphorylation status and apoptosis measured by DNA break frequency, was observed only in circadian-synchronized WT-MEF, not in CRYDKO or mouse hepatocarcinoma Hepa1c1c7 cells. These findings highlight that the DNA damage responses triggered by cisplatin are indeed governed by circadian control exclusively in clock-proficient cells. This outcome bears potential implications for enhancing or devising chronotherapy approaches for cancer patients.
Collapse
Affiliation(s)
- Jeseok Jeon
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
| | - Sanggon Lee
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
| | - Jeong-Min Park
- Department of Stem Cell Transplantation Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tae-Hee Lee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tae-Hong Kang
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea.
| |
Collapse
|
170
|
Yang Y, Wang P, Ji Z, Xu X, Zhang H, Wang Y. Polysaccharide‑platinum complexes for cancer theranostics. Carbohydr Polym 2023; 315:120997. [PMID: 37230639 DOI: 10.1016/j.carbpol.2023.120997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 05/27/2023]
Abstract
Platinum anticancer drugs have been explored and developed in recent years to reduce systematic toxicities and resist drug resistance. Polysaccharides derived from nature have abundant structures as well as pharmacological activities. The review provides insights on the design, synthesis, characterization and associating therapeutic application of platinum complexes with polysaccharides that are classified by electronic charge. The complexes give birth to multifunctional properties with enhanced drug accumulation, improved tumor selectivity and achieved synergistic antitumor effect in cancer therapy. Several techniques developing polysaccharides-based carriers newly are also discussed. Moreover, the lasted immunoregulatory activities of innate immune reactions triggered by polysaccharides are summarized. Finally, we discuss the current shortcomings and outline potential strategies for improving platinum-based personalized cancer treatment. Using platinum-polysaccharides complexes for improving the immunotherapy efficiency represents a promising framework in future.
Collapse
Affiliation(s)
- Yunxia Yang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Province Engineering Research Center of Agricultural Breeding Pollution Control and Resource, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng 224007, China.
| | - Pengge Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Zengrui Ji
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China.
| | - Hongmei Zhang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Yanqing Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China.
| |
Collapse
|
171
|
Ojha R, Junk PC, Bond AM, Deacon GB. Oxidation of the Platinum(II) Anticancer Agent [Pt{( p-BrC 6F 4)NCH 2CH 2NEt 2}Cl(py)] to Platinum(IV) Complexes by Hydrogen Peroxide. Molecules 2023; 28:6402. [PMID: 37687231 PMCID: PMC10490441 DOI: 10.3390/molecules28176402] [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: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
PtIV coordination complexes are of interest as prodrugs of PtII anticancer agents, as they can avoid deactivation pathways owing to their inert nature. Here, we report the oxidation of the antitumor agent [PtII(p-BrC6F4)NCH2CH2NEt2}Cl(py)], 1 (py = pyridine) to dihydroxidoplatinum(IV) solvate complexes [PtIV{(p-BrC6F4)NCH2CH2NEt2}Cl(OH)2(py)].H2O, 2·H2O with hydrogen peroxide (H2O2) at room temperature. To optimize the yield, 1 was oxidized in the presence of added lithium chloride with H2O2 in a 1:2 ratio of Pt: H2O2, in CH2Cl2 producing complex 2·H2O in higher yields in both gold and red forms. Despite the color difference, red and yellow 2·H2O have the same structure as determined by single-crystal and X-ray powder diffraction, namely, an octahedral ligand array with a chelating organoamide, pyridine and chloride ligands in the equatorial plane, and axial hydroxido ligands. When tetrabutylammonium chloride was used as a chloride source, in CH2Cl2, another solvate, [PtIV{(p-BrC6F4)NCH2CH2NEt2}Cl(OH)2(py)].0.5CH2Cl2,3·0.5CH2Cl2, was obtained. These PtIV compounds show reductive dehydration into PtII [Pt{(p-BrC6F4)NCH=CHNEt2}Cl(py)], 1H over time in the solid state, as determined by X-ray powder diffraction, and in solution, as determined by 1H and 19F NMR spectroscopy and mass spectrometry. 1H contains an oxidized coordinating ligand and was previously obtained by oxidation of 1 under more vigorous conditions. Experimental data suggest that oxidation of the ligand is favored in the presence of excess H2O2 and elevated temperatures. In contrast, a smaller amount (1Pt:2H2O2) of H2O2 at room temperature favors the oxidation of the metal and yields platinum(IV) complexes.
Collapse
Affiliation(s)
- Ruchika Ojha
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia; (R.O.); (A.M.B.); (G.B.D.)
| | - Peter C. Junk
- College of Science, Technology & Engineering, James Cook University, Townsville, QLD 4811, Australia
| | - Alan M. Bond
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia; (R.O.); (A.M.B.); (G.B.D.)
| | - Glen B. Deacon
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia; (R.O.); (A.M.B.); (G.B.D.)
| |
Collapse
|
172
|
Smerdon MJ, Wyrick JJ, Delaney S. A half century of exploring DNA excision repair in chromatin. J Biol Chem 2023; 299:105118. [PMID: 37527775 PMCID: PMC10498010 DOI: 10.1016/j.jbc.2023.105118] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023] Open
Abstract
DNA in eukaryotic cells is packaged into the compact and dynamic structure of chromatin. This packaging is a double-edged sword for DNA repair and genomic stability. Chromatin restricts the access of repair proteins to DNA lesions embedded in nucleosomes and higher order chromatin structures. However, chromatin also serves as a signaling platform in which post-translational modifications of histones and other chromatin-bound proteins promote lesion recognition and repair. Similarly, chromatin modulates the formation of DNA damage, promoting or suppressing lesion formation depending on the chromatin context. Therefore, the modulation of DNA damage and its repair in chromatin is crucial to our understanding of the fate of potentially mutagenic and carcinogenic lesions in DNA. Here, we survey many of the landmark findings on DNA damage and repair in chromatin over the last 50 years (i.e., since the beginning of this field), focusing on excision repair, the first repair mechanism studied in the chromatin landscape. For example, we highlight how the impact of chromatin on these processes explains the distinct patterns of somatic mutations observed in cancer genomes.
Collapse
Affiliation(s)
- Michael J Smerdon
- Biochemistry and Biophysics, School of Molecular Biosciences, Washington State University, Pullman, Washington, USA.
| | - John J Wyrick
- Genetics and Cell Biology, School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Sarah Delaney
- Department of Chemistry, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
173
|
Sun Y, Ma H. Application of three-dimensional cell culture technology in screening anticancer drugs. Biotechnol Lett 2023; 45:1073-1092. [PMID: 37421554 DOI: 10.1007/s10529-023-03410-x] [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: 03/03/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/10/2023]
Abstract
The drug development process involves a variety of drug activity evaluations, which can determine drug efficacy, strictly analyze the biological indicators after the drug action, and use these indicators as the preclinical drug evaluation criteria. At present, most of the screening of preclinical anticancer drugs mainly relies on traditional 2D cell culture. However, this traditional technology cannot simulate the tumor microenvironment in vivo, let alone reflect the characteristics of solid tumors in vivo, and has a relatively poor ability to predict drug activity. 3D cell culture is a technology between 2D cell culture and animal experiments, which can better reflect the biological state in vivo and reduce the consumption of animal experiments. 3D cell culture can link the individual study of cells with the study of the whole organism, reproduce in vitro the biological phenotype of cells in vivo more greatly, and thus predict the activity and resistance of anti-tumor drugs more accurately. In this paper, the common techniques of 3D cell culture are discussed, with emphasis on its main advantages and application in the evaluation of anti-tumor resistance, which can provide strategies for the screening of anti-tumor drugs.
Collapse
Affiliation(s)
- Yaqian Sun
- Oncology laboratory, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China.
| | - Haiyang Ma
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, People's Republic of China
| |
Collapse
|
174
|
Gillespie KP, Pirnie R, Mesaros C, Blair IA. Cisplatin Dependent Secretion of Immunomodulatory High Mobility Group Box 1 (HMGB1) Protein from Lung Cancer Cells. Biomolecules 2023; 13:1335. [PMID: 37759736 PMCID: PMC10526420 DOI: 10.3390/biom13091335] [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: 07/12/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
High mobility group box 1 (HMGB1) is secreted from activated immune cells, necrotic cells, and certain cancers. Previous studies have reported that different patterns of post-translational modification, particularly acetylation and oxidation, mediate HMGB1 release and confer distinct extracellular HMGB1 signaling activity. Here we report that cisplatin but not carboplatin induces secretion of HMGB1 from human A549 non-small cell lung cancer (NSCLC) cells. Cisplatin-mediated HMGB1 secretion was dose-dependent and was regulated by nuclear exportin 1 (XPO1) also known as chromosomal maintenance 1 (CRM1) rather than adenosine diphosphate (ADP)-ribosylation, acetylation, or oxidation. HMGB1, as well as lysine acetylation and cysteine disulfide oxidation of secreted HMGB1, were monitored by sensitive and specific assays using immunoprecipitation, stable isotope dilution, differential alkylation, and nano liquid chromatography parallel reaction monitoring/high-resolution mass spectrometry (nano-LC-PRM/HRMS). A major fraction of the HMGB1 secreted by low-dose cisplatin treatment of A549 NSCLC cells was found to be in the fully reduced form. In contrast, mainly oxidized forms of HMGB1 were secreted by dimethyl sulfoxide (DMSO)-mediated apoptosis. These findings suggest that inhibition of XPO1 could potentiate the anti-tumor activity of cisplatin by increasing the nuclear accumulation of HMGB1 protein, an inhibitor of cisplatin DNA-adduct repair. Furthermore, low-dose cisplatin therapy could modulate the immune response in NSCLC through the established chemokine activity of extracellular reduced HMGB1. This could potentially enhance the efficacy of subsequent immunotherapy treatment.
Collapse
Affiliation(s)
| | | | | | - Ian A. Blair
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
175
|
Kshetri M, Jogadi W, Alqarni S, Datta P, Cheline M, Sharma A, Betters T, Broyles D, Zheng YR. Exploring the Impact of Head Group Modifications on the Anticancer Activities of Fatty-Acid-like Platinum(IV) Prodrugs: A Structure-Activity Relationship Study. Int J Mol Sci 2023; 24:13301. [PMID: 37686109 PMCID: PMC10487970 DOI: 10.3390/ijms241713301] [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: 07/30/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
We conducted the first comprehensive investigation on the impact of head group modifications on the anticancer activities of fatty-acid-like Pt(IV) prodrugs (FALPs), which are a class of platinum-based metallodrugs that target mitochondria. We created a small library of FALPs (1-9) with diverse head group modifications. The outcomes of our study demonstrate that hydrophilic modifications exclusively enhance the potency of these metallodrugs, whereas hydrophobic modifications significantly decrease their cytotoxicity. To further understand this interesting structure-activity relationship, we chose two representative FALPs (compounds 2 and 7) as model compounds: one (2) with a hydrophilic polyethylene glycol (PEG) head group, and the other (7) with a hydrophobic hydrocarbon modification of the same molecular weight. Using these FALPs, we conducted a targeted investigation on the mechanism of action. Our study revealed that compound 2, with hydrophilic modifications, exhibited remarkable penetration into cancer cells and mitochondria, leading to subsequent mitochondrial and DNA damage, and effectively eradicating cancer cells. In contrast, compound 7, with hydrophobic modifications, displayed a significantly lower uptake and weaker cellular responses. The collective results present a different perspective, indicating that increased hydrophobicity may not necessarily enhance cellular uptake as is conventionally believed. These findings provide valuable new insights into the fundamental principles of developing metallodrugs.
Collapse
Affiliation(s)
- Man Kshetri
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Wjdan Jogadi
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Suha Alqarni
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
- Department of Chemistry, University of Bisha, Bisha 67714, Saudi Arabia
| | - Payel Datta
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - May Cheline
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Arpit Sharma
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Tyler Betters
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Deonya Broyles
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Yao-Rong Zheng
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| |
Collapse
|
176
|
Gorle AK, Malde AK, Chang CW, Rajaratnam P, von Itzstein M, Berners-Price SJ, Farrell NP. Probing Disaccharide Binding to Triplatin as Models for Tumor Cell Heparan Sulfate (GAG) Interactions. Inorg Chem 2023; 62:13212-13220. [PMID: 37552525 PMCID: PMC10445638 DOI: 10.1021/acs.inorgchem.3c01391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Indexed: 08/09/2023]
Abstract
In this study, we have used [1H, 15N] NMR spectroscopy to investigate the interactions of the trinuclear platinum anticancer drug triplatin (1) (1,0,1/t,t,t or BBR3464) with site-specific sulfated and carboxylated disaccharides. Specifically, the disaccharides GlcNS(6S)-GlcA (I) and GlcNS(6S)-IdoA(2S) (II) are useful models of longer-chain glycosaminoglycans (GAGs) such as heparan sulfate (HS). For both the reactions of 15N-1 with I and II, equilibrium conditions were achieved more slowly (65 h) compared to the reaction with the monosaccharide GlcNS(6S) (9 h). The data suggest both carboxylate and sulfate binding of disaccharide I to the Pt with the sulfato species accounting for <1% of the total species at equilibrium. The rate constant for sulfate displacement of the aqua ligand (kL2) is 4 times higher than the analogous rate constant for carboxylate displacement (kL1). There are marked differences in the equilibrium concentrations of the chlorido, aqua, and carboxy-bound species for reactions with the two disaccharides, notably a significantly higher concentration of carboxylate-bound species for II, where sulfate-bound species were barely detectable. The trend mirrors that reported for the corresponding dinuclear platinum complex 1,1/t,t, where the rate constant for sulfate displacement of the aqua ligand was 3 times higher than that for acetate. Also similar to what we observed for the reactions of 1,1/t,t with the simple anions, aquation of the sulfato group is rapid, and the rate constant k-L2 is 3 orders of magnitude higher than that for displacement of the carboxylate (k-L1). Molecular dynamics calculations suggest that extra hydrogen-bonding interactions with the more sulfated disaccharide II may prevent or diminish sulfate binding of the triplatin moiety. The overall results suggest that Pt-O donor interactions should be considered in any full description of platinum complex cellular chemistry.
Collapse
Affiliation(s)
- Anil K. Gorle
- Institute
for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Alpeshkumar K. Malde
- Institute
for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Chih-Wei Chang
- Institute
for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Premraj Rajaratnam
- Institute
for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Mark von Itzstein
- Institute
for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Susan J. Berners-Price
- Institute
for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Nicholas P. Farrell
- Institute
for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| |
Collapse
|
177
|
Nechay M, Wang D, Kleiner RE. Inhibition of nucleolar transcription by oxaliplatin involves ATM/ATR kinase signaling. Cell Chem Biol 2023; 30:906-919.e4. [PMID: 37433295 PMCID: PMC10529435 DOI: 10.1016/j.chembiol.2023.06.010] [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: 05/06/2022] [Revised: 03/25/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023]
Abstract
Platinum (Pt) compounds are an important class of anti-cancer therapeutics, but outstanding questions remain regarding their mechanism of action. Here, we demonstrate that oxaliplatin, a Pt drug used to treat colorectal cancer, inhibits rRNA transcription through ATM and ATR signaling, and induces DNA damage and nucleolar disruption. We show that oxaliplatin causes nucleolar accumulation of the nucleolar DNA damage response proteins (n-DDR) NBS1 and TOPBP1; however transcriptional inhibition does not depend upon NBS1 or TOPBP1, nor does oxaliplatin induce substantial amounts of nucleolar DNA damage, distinguishing the nucleolar response from previously characterized n-DDR pathways. Taken together, our work indicates that oxaliplatin induces a distinct ATM and ATR signaling pathway that functions to inhibit Pol I transcription in the absence of direct nucleolar DNA damage, demonstrating how nucleolar stress and transcriptional silencing can be linked to DNA damage signaling and highlighting an important mechanism of Pt drug cytotoxicity.
Collapse
Affiliation(s)
- Misha Nechay
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Danyang Wang
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Ralph E Kleiner
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
178
|
Xu Z, Zhang M, Wang W, Zhou S, Yu M, Qiu X, Jiang S, Wang X, Tang C, Li S, Wang CH, Zhu R, Peng WX, Zhao L, Fu X, Patzak A, Persson PB, Zhao L, Mao J, Shu Q, Lai EY, Zhang G. Dihydromyricetin attenuates cisplatin-induced acute kidney injury by reducing oxidative stress, inflammation and ferroptosis. Toxicol Appl Pharmacol 2023; 473:116595. [PMID: 37328118 DOI: 10.1016/j.taap.2023.116595] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Cisplatin is effective against various types of cancers. However, its clinical application is limited owing to its adverse effects, especially acute kidney injury (AKI). Dihydromyricetin (DHM), a flavonoid derived from Ampelopsis grossedentata, has varied pharmacological activities. This research aimed to determine the molecular mechanism for cisplatin-induced AKI. METHODS A murine model of cisplatin-induced AKI (22 mg/kg, I.P.) and a HK-2 cell model of cisplatin-induced damage (30 μM) were established to evaluate the protective function of DHM. Renal dysfunction markers, renal morphology and potential signaling pathways were investigated. RESULTS DHM decreased the levels of renal function biomarkers (blood urea nitrogen and serum creatinine), mitigated renal morphological damage, and downregulated the protein levels of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin. It upregulated the expression levels of antioxidant enzymes (superoxide dismutase and catalase expression), nuclear factor-erythroid-2-related factor 2 (Nrf2) and its downstream proteins, including heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic (GCLC) and modulatory (GCLM) subunits, thus eventually reducing cisplatin-induced reactive oxygen species (ROS) production. Moreover, DHM partially inhibited the phosphorylation of the active fragments of caspase-8 and -3 and mitogen-activated protein kinase and restored glutathione peroxidase 4 expression, which attenuated renal apoptosis and ferroptosis in cisplatin-treated animals. DHM also mitigated the activation of NLRP3 inflammasome and nuclear factor (NF)-κB, attenuating the inflammatory response. In addition, it reduced cisplatin-induced HK-2 cell apoptosis and ROS production, both of which were blocked by the Nrf2 inhibitor ML385. CONCLUSIONS DHM suppressed cisplatin-induced oxidative stress, inflammation and ferroptosis probably through regulating of Nrf2/HO-1, MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Zheming Xu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China
| | - Minjing Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China
| | - Wenwen Wang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310051, China
| | - Suhan Zhou
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Minghua Yu
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xingyu Qiu
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shan Jiang
- Department of Nephrology, Center of Kidney and Urology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Xiaohua Wang
- Department of Nephrology, Center of Kidney and Urology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Chun Tang
- Department of Nephrology, Center of Kidney and Urology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Shuijie Li
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chih-Hong Wang
- Tulane Hypertension and Renal Center of Excellence, Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Runzhi Zhu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China
| | - Wan Xin Peng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China
| | - Lin Zhao
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China
| | - Xiaodong Fu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 510260, China
| | - Andreas Patzak
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institute of Translational Physiology, Berlin, Germany
| | - Pontus B Persson
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institute of Translational Physiology, Berlin, Germany
| | - Liang Zhao
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China; Children's Hospital, Zhejiang University School of Medicine, Pediatric Nephrology & Urology Medical Research Center, Hangzhou 310052, China
| | - Jianhua Mao
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China; Children's Hospital, Zhejiang University School of Medicine, Pediatric Nephrology & Urology Medical Research Center, Hangzhou 310052, China
| | - Qiang Shu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China.
| | - En Yin Lai
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institute of Translational Physiology, Berlin, Germany.
| | - Gensheng Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China; Children's Hospital, Zhejiang University School of Medicine, Pediatric Nephrology & Urology Medical Research Center, Hangzhou 310052, China.
| |
Collapse
|
179
|
Jin S, Yin E, Feng C, Sun Y, Yang T, Yuan H, Guo Z, Wang X. Regulating tumor glycometabolism and the immune microenvironment by inhibiting lactate dehydrogenase with platinum(iv) complexes. Chem Sci 2023; 14:8327-8337. [PMID: 37564403 PMCID: PMC10411615 DOI: 10.1039/d3sc01874a] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Lactate dehydrogenase (LDH) is a key enzyme involved in the process of glycolysis, assisting cancer cells to take in glucose and generate lactate, as well as to suppress and evade the immune system by altering the tumor microenvironment (TME). Platinum(iv) complexes MDP and DDP were prepared by modifying cisplatin with diclofenac at the axial position(s). These complexes exhibited potent antiproliferative activity against a panel of human cancer cell lines. In particular, DDP downregulated the expression of LDHA, LDHB, and MCTs to inhibit the production and influx/efflux of lactate in cancer cells, impeding both glycolysis and glucose oxidation. MDP and DDP also reduced the expression of HIF-1α, ARG1 and VEGF, thereby disrupting the formation of tumor vasculature. Furthermore, they promoted the repolarization of macrophages from the tumor-supportive M2 phenotype to the tumor-suppressive M1 phenotype in the TME, thus enhancing the antitumor immune response. The antitumor mechanism involves reprogramming the energy metabolism of tumor cells and relieving the immunosuppressive TME.
Collapse
Affiliation(s)
- Suxing Jin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 P. R. China +86 25 89684549 +86 25 89684549
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University Nanjing 210023 P. R. China
| | - Enmao Yin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 P. R. China +86 25 89684549 +86 25 89684549
| | - Chenyao Feng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 P. R. China +86 25 89684549 +86 25 89684549
| | - Yuewen Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 P. R. China +86 25 89684549 +86 25 89684549
| | - Tao Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University Nanjing 210023 P. R. China
| | - Hao Yuan
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University Nanjing 210023 P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 P. R. China +86 25 89684549 +86 25 89684549
| |
Collapse
|
180
|
Tarai SK, Pan A, Biswas P, Bhaduri R, Mandal S, Paul A, Baitalik S, Bhattacharjee A, Moi SC. Anticancer Behavior of Pyrrolidine-Based Palladium(II) Complexes and Biophysical Approach on Their DNA, BSA Binding Activity, Molecular Docking, and DFT Study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:10947-10964. [PMID: 37501125 DOI: 10.1021/acs.langmuir.3c01186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
A series of pyrrolidine-based Pd(II) complexes, [Pd(AEP)Cl2] (C-1), [Pd(AEP)(OH2)2]2+(C-2), [Pd(AEP)(L-cys)]+ (C-3), [Pd(AEP)(N-ac-L-cys)] (C-4), [Pd(AEP)(GSH)] (C-5), and [Pd(AEP)(DL-meth)]2+ (C-6) (where, AEP = 1-(2-aminoethyl)pyrrolidine, L-cys = l-cysteine, N-ac-L-cys = N-acetyl-l-cysteine, GSH = glutathione, and DL-meth = dl-methionine), as anticancer drug candidates have been synthesized and characterized. The DNA binding property of the complexes was executed by gel electrophoresis and spectrophotometric and viscometric methods, and their interaction with BSA was also investigated by various spectroscopic methodologies. The binding activity of the Pd(II) complexes with DNA and BSA were assessed to evaluate their binding mode and binding constants. Molecular docking was performed to correlate with the experimental results on the interaction of the complexes with DNA and BSA. The changes in the microenvironmental and structural properties of BSA are monitored by a synchronous and 3D fluorescence study. The structural properties were evaluated by DFT and TD-DFT studies. The anticarcinogenic activity of the Pd(II) complexes was assessed by PASS prediction software to corroborate with the experimental results of the anticancer activity of the complexes. The ROS generation in cancer cell lines has been investigated, and the cell death mechanism through apoptosis was confirmed by measuring the protein expression. All these complexes have excellent anticancer activity compared to ancillary ligands. The cancer cell line (HCT116) shows almost similar or better cell inhibition activity when treated with the Pd(II) complexes compared to cisplatin, whereas the adverse effect is minimum on a normal cell (NKE). Both the Pd(II) and Pt(II) complexes carrying the same ligands reveal almost similar antiproliferative activity.
Collapse
Affiliation(s)
- Swarup Kumar Tarai
- Department of Chemistry, National Institute of Technology Durgapur, Durgapur, West Bengal 713209, India
| | - Angana Pan
- Department of Chemistry, National Institute of Technology Durgapur, Durgapur, West Bengal 713209, India
| | - Pritam Biswas
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal 713209, India
| | - Rituparna Bhaduri
- Department of Chemistry, National Institute of Technology Durgapur, Durgapur, West Bengal 713209, India
| | - Saikat Mandal
- Department of Chemistry, National Institute of Technology Durgapur, Durgapur, West Bengal 713209, India
| | - Animesh Paul
- Inorganic Chemistry Section, Department of Chemistry, Jadavpur University, Kolkata 700032, India
| | - Sujoy Baitalik
- Inorganic Chemistry Section, Department of Chemistry, Jadavpur University, Kolkata 700032, India
| | - Ashish Bhattacharjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal 713209, India
| | - Sankar Ch Moi
- Department of Chemistry, National Institute of Technology Durgapur, Durgapur, West Bengal 713209, India
| |
Collapse
|
181
|
Jayawardhana AMDS, Bhandari S, Kaspi-Kaneti AW, Kshetri M, Qiu Z, Cheline M, Shen H, Dunietz BD, Zheng YR. Visible light-activatable platinum(IV) prodrugs harnessing CD36 for ovarian cancer therapy. Dalton Trans 2023; 52:10942-10950. [PMID: 37490033 PMCID: PMC11298056 DOI: 10.1039/d3dt01292a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
We hereby engineered photoactivatable Pt(IV) metallodrugs that harness CD36 to target ovarian cancer cells. Pt(IV) compounds mimic the structure of fatty acids and take advantage of CD36 as a "Trojan horse" to gain entry into the cells. We confirmed that CD36-dependent entry occurs using graphite furnace atomic absorption spectroscopy with ovarian cancer cells expressing different levels of CD36 and a CD36 inhibitor, SSO. Once the Pt(IV) metallodrugs enter the cancer cells, they can be activated to form Pt(II) with characteristics of cisplatin under visible light (490 nm) irradiation, promoting photoinduced electron transfer from the attached fluorophore to the metal center. This light-induced activation can increase the cytotoxicity of the Pt(IV) metallodrugs by up to 20 times toward ovarian cancer cells, inducing DNA damage and enabling efficient elimination of drug-resistant cancer cells.
Collapse
Affiliation(s)
| | - Srijana Bhandari
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| | - Ariela W Kaspi-Kaneti
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| | - Man Kshetri
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| | - Zihan Qiu
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| | - May Cheline
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| | - Hao Shen
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| | - Barry D Dunietz
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| | - Yao-Rong Zheng
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA.
| |
Collapse
|
182
|
Ahmad MG, Balamurali MM, Chanda K. Click-derived multifunctional metal complexes for diverse applications. Chem Soc Rev 2023; 52:5051-5087. [PMID: 37431583 DOI: 10.1039/d3cs00343d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
The Click reaction that involves Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) serves as the most potent and highly dependable tool for the development of many complex architectures. It has paved the way for the synthesis of numerous drug molecules with enhanced synthetic flexibility, reliability, specificity and modularity. It is all about bringing two different molecular entities together to achieve the required molecular properties. The utilization of Click chemistry has been well demonstrated in organic synthesis, particularly in reactions that involve biocompatible precursors. In pharmaceutical research, Click chemistry is extensively utilized for drug delivery applications. The exhibited bio-compatibility and dormancy towards other biological components under cellular environments makes Click chemistry an identified boon in bio-medical research. In this review, various click-derived transition metal complexes are discussed in terms of their applications and uniqueness. The scope of this chemistry towards other streams of applied sciences is also discussed.
Collapse
Affiliation(s)
- Md Gulzar Ahmad
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India.
| | - M M Balamurali
- Chemistry Division, School of Advanced Sciences, Vellore Institute of Technology, Chennai campus, Chennai 600127, Tamilnadu, India.
| | - Kaushik Chanda
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India.
| |
Collapse
|
183
|
Yang Y, Lin M, Sun M, Zhang GQ, Guo J, Li J. Nanotechnology boosts the efficiency of tumor diagnosis and therapy. Front Bioeng Biotechnol 2023; 11:1249875. [PMID: 37576984 PMCID: PMC10419217 DOI: 10.3389/fbioe.2023.1249875] [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: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023] Open
Abstract
The incidence and mortality of cancer are gradually increasing. The highly invasive and metastasis of tumor cells increase the difficulty of diagnosis and treatment, so people pay more and more attention to the diagnosis and treatment of cancer. Conventional treatment methods, including surgery, radiotherapy and chemotherapy, are difficult to eliminate tumor cells completely. And the emergence of nanotechnology has boosted the efficiency of tumor diagnosis and therapy. Herein, the research progress of nanotechnology used for tumor diagnosis and treatment is reviewed, and the emerging detection technology and the application of nanodrugs in clinic are summarized and prospected. The first part refers to the application of different nanomaterials for imaging in vivo and detection in vitro, which includes magnetic resonance imaging, fluorescence imaging, photoacoustic imaging and biomarker detection. The distinctive physical and chemical advantages of nanomaterials can improve the detection sensitivity and accuracy to achieve tumor detection in early stage. The second part is about the nanodrug used in clinic for tumor treatment. Nanomaterials have been widely used as drug carriers, including the albumin paclitaxel, liposome drugs, mRNA-LNP, protein nanocages, micelles, membrane nanocomplexes, microspheres et al., which could improve the drug accumulate in tumor tissue through enhanced permeability and retention effect to kill tumor cells with high efficiency. But there are still some challenges to revolutionize traditional tumor diagnosis and anti-drug resistance based on nanotechnology.
Collapse
Affiliation(s)
| | | | | | | | - Jianshuang Guo
- Pharmacology and Toxicology Research Laboratory, College of Pharmaceutical Science, Hebei University, Baoding, Hebei, China
| | - Jianheng Li
- Pharmacology and Toxicology Research Laboratory, College of Pharmaceutical Science, Hebei University, Baoding, Hebei, China
| |
Collapse
|
184
|
Du K, Liao P, Yang S, von Trentini D, Sharma K, Shi X, Murray CB, Li D, Dmochowski IJ. Chelate-functionalized magnetic micelles for sequestration of cisplatin. NANOSCALE ADVANCES 2023; 5:3955-3963. [PMID: 37496616 PMCID: PMC10367963 DOI: 10.1039/d3na00290j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/02/2023] [Indexed: 07/28/2023]
Abstract
Many cancer patients suffer permanent hearing loss due to accumulation of ototoxic cisplatin in the inner ear. In this study, two types of 100 nm magnetic micelles were developed to sequester cisplatin from aqueous solutions, with the goal of eliminating cochlear ototoxins via magnetic microsurgery. The micellar surface was quantitatively functionalized with anionic S-rich ligands and the micelle core encapsulated superparamagnetic iron oxide nanoparticles. Exceptionally effective sequestration is demonstrated, with removal of greater than 95 and 50% of solution Pt, by means of centrifugal filtration and magnetic extraction. Attraction between negatively charged micellar surfaces and cationic Pt-species played a critical role and was only partially screened by physiologic salt solution. Importantly, magnetic micelles introduce negligible impact on the integrity of inner ear hair cells, demonstrating excellent biocompatibility. This study showcases successful magnetic sequestration of Pt-based ototoxins using highly applicable nano-micellar materials. More generally, these examples highlight features of the micelle-water interfacial environment that are important in developing nanomaterials for metallo-medicinal applications.
Collapse
Affiliation(s)
- Kang Du
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| | - Pan Liao
- Department of Otorhinolaryngology, University of Pennsylvania Philadelphia PA 19104 USA
| | - Shengsong Yang
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| | - Dora von Trentini
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| | - Kushal Sharma
- Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University Portland OR 97239 USA
| | - Xiaorui Shi
- Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University Portland OR 97239 USA
| | - Christopher B Murray
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
- Department of Materials Science and Engineering, University of Pennsylvania Philadelphia PA 19104 USA
| | - Daqing Li
- Department of Otorhinolaryngology, University of Pennsylvania Philadelphia PA 19104 USA
| | - Ivan J Dmochowski
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| |
Collapse
|
185
|
Jevtovic V, Alshamari AK, Milenković D, Dimitrić Marković J, Marković Z, Dimić D. The Effect of Metal Ions (Fe, Co, Ni, and Cu) on the Molecular-Structural, Protein Binding, and Cytotoxic Properties of Metal Pyridoxal-Thiosemicarbazone Complexes. Int J Mol Sci 2023; 24:11910. [PMID: 37569285 PMCID: PMC10419307 DOI: 10.3390/ijms241511910] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Thiosemicarbazones and their transition metal complexes are biologically active compounds and anticancer agents with versatile structural properties. In this contribution, the structural features and stability of four pyridoxal-thiosemicarbazone (PLTSC) complexes with Fe, Co, Ni, and Cu were investigated using the density functional theory and natural bond orbital approach. Special emphasis was placed on the analysis of the donor atom-metal interactions. The geometry of compounds and crystallographic structures were further examined by Hirshfeld surface analysis, and the main intermolecular interactions were outlined. It has been shown that the geometry and the number of PLTSC units in the structure determine the type and contribution of the specific interactions. The binding of all four complexes to bovine and human serum albumin was investigated through spectrofluorometric titration. The dependency of the thermodynamic parameters on the present metal ion and geometry was explained by the possible interactions through molecular docking simulations. The binding of complexes to DNA, as one of the possible ways the compounds could induce cell death, was examined by molecular docking. The cytotoxicity was measured towards HCT116, A375, MCF-7, A2780, and MCF5 cell lines, with Cu-PLTSC being the most active, as it had the highest affinity towards DNA and proteins.
Collapse
Affiliation(s)
- Violeta Jevtovic
- Department of Chemistry, College of Science, University Ha’il, Ha’il 81451, Saudi Arabia
| | - Asma K. Alshamari
- Department of Chemistry, College of Science, University Ha’il, Ha’il 81451, Saudi Arabia
| | - Dejan Milenković
- Department of Science, Institute for Information Technologies, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | | | - Zoran Marković
- Department of Science, Institute for Information Technologies, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Dušan Dimić
- Faculty of Physical Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia
| |
Collapse
|
186
|
Swaminathan S, Karvembu R. Dichloro Ru(II)- p-cymene-1,3,5-triaza-7-phosphaadamantane (RAPTA-C): A Case Study. ACS Pharmacol Transl Sci 2023; 6:982-996. [PMID: 37470017 PMCID: PMC10353064 DOI: 10.1021/acsptsci.3c00085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 07/21/2023]
Abstract
The use of organometallic compounds to treat various phenotypes of cancer has attracted increased interest in recent decades. Organometallic compounds, which are transitional between conventional inorganic and organic materials, have outstanding and one-of-a-kind features that offer fresh insight into the development of inorganic medicinal chemistry. The therapeutic potential of ruthenium(II)-arene RAPTA-type compounds is being thoroughly investigated, specifically owing to the excellent antimetastatic property of the initial candidate RAPTA-C. This review gives a thorough analysis of this complex and its evolution as a potential anticancer drug candidate. The numerous mechanistic investigations of RAPTA-C are discussed, and they are connected to the macroscopic biological characteristics that have been found. The "multitargeted" complex described here target enzymes, peptides, and intracellular proteins in addition to DNA that allow it to specifically target cancer cells. Understanding these may allow researchers to find specific targets and tune a new-generation organometallic complex accordingly.
Collapse
Affiliation(s)
- Srividya Swaminathan
- Department
of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
- Center
for Computational Modeling, Chennai Institute
of Technology (CIT), Chennai 600069, India
| | - Ramasamy Karvembu
- Department
of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
| |
Collapse
|
187
|
Barresi E, Robello M, Baglini E, Poggetti V, Viviano M, Salerno S, Da Settimo F, Taliani S. Indol-3-ylglyoxylamide as Privileged Scaffold in Medicinal Chemistry. Pharmaceuticals (Basel) 2023; 16:997. [PMID: 37513909 PMCID: PMC10386336 DOI: 10.3390/ph16070997] [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: 06/12/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, indolylglyoxylamide-based derivatives have received much attention due to their application in drug design and discovery, leading to the development of a wide array of compounds that have shown a variety of pharmacological activities. Combining the indole nucleus, already validated as a "privileged structure," with the glyoxylamide function allowed for an excellent template to be obtained that is suitable to a great number of structural modifications aimed at permitting interaction with specific molecular targets and producing desirable therapeutic effects. The present review provides insight into how medicinal chemists have elegantly exploited the indolylglyoxylamide moiety to obtain potentially useful drugs, with a particular focus on compounds exhibiting activity in in vivo models or reaching clinical trials. All in all, this information provides exciting new perspectives on existing data that can be useful in further design of indolylglyoxylamide-based molecules with interesting pharmacological profiles. The aim of this report is to present an update of collection data dealing with the employment of this moiety in the rational design of compounds that are able to interact with a specific target, referring to the last 20 years.
Collapse
Affiliation(s)
- Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Marco Robello
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
188
|
Compadre AJ, van Biljon LN, Valentine MC, Llop-Guevara A, Graham E, Fashemi B, Herencia-Ropero A, Kotnik EN, Cooper I, Harrington SP, Kuroki LM, McCourt CK, Hagemann AR, Thaker PH, Mutch DG, Powell MA, Sun L, Mosammaparast N, Serra V, Zhao P, Lomonosova E, Khabele D, Mullen MM. RAD51 Foci as a Biomarker Predictive of Platinum Chemotherapy Response in Ovarian Cancer. Clin Cancer Res 2023; 29:2466-2479. [PMID: 37097615 PMCID: PMC10320470 DOI: 10.1158/1078-0432.ccr-22-3335] [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: 10/31/2022] [Revised: 01/31/2023] [Accepted: 04/20/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE To determine the ability of RAD51 foci to predict platinum chemotherapy response in high-grade serous ovarian cancer (HGSOC) patient-derived samples. EXPERIMENTAL DESIGN RAD51 and γH2AX nuclear foci were evaluated by immunofluorescence in HGSOC patient-derived cell lines (n = 5), organoids (n = 11), and formalin-fixed, paraffin-embedded tumor samples (discovery n = 31, validation n = 148). Samples were defined as RAD51-High if >10% of geminin-positive cells had ≥5 RAD51 foci. Associations between RAD51 scores, platinum chemotherapy response, and survival were evaluated. RESULTS RAD51 scores correlated with in vitro response to platinum chemotherapy in established and primary ovarian cancer cell lines (Pearson r = 0.96, P = 0.01). Organoids from platinum-nonresponsive tumors had significantly higher RAD51 scores than those from platinum-responsive tumors (P < 0.001). In a discovery cohort, RAD51-Low tumors were more likely to have a pathologic complete response (RR, 5.28; P < 0.001) and to be platinum-sensitive (RR, ∞; P = 0.05). The RAD51 score was predictive of chemotherapy response score [AUC, 0.90; 95% confidence interval (CI), 0.78-1.0; P < 0.001). A novel automatic quantification system accurately reflected the manual assay (92%). In a validation cohort, RAD51-Low tumors were more likely to be platinum-sensitive (RR, ∞; P < 0.001) than RAD51-High tumors. Moreover, RAD51-Low status predicted platinum sensitivity with 100% positive predictive value and was associated with better progression-free (HR, 0.53; 95% CI, 0.33-0.85; P < 0.001) and overall survival (HR, 0.43; 95% CI, 0.25-0.75; P = 0.003) than RAD51-High status. CONCLUSIONS RAD51 foci are a robust marker of platinum chemotherapy response and survival in ovarian cancer. The utility of RAD51 foci as a predictive biomarker for HGSOC should be tested in clinical trials.
Collapse
Affiliation(s)
- Amanda J. Compadre
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Lillian N. van Biljon
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Mark C. Valentine
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Alba Llop-Guevara
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Emily Graham
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Bisiayo Fashemi
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Andrea Herencia-Ropero
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
| | - Emilee N. Kotnik
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Isaac Cooper
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | | | - Lindsay M. Kuroki
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Carolyn K. McCourt
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Andrea R. Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Premal H. Thaker
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - David G. Mutch
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Matthew A. Powell
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Lulu Sun
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Nima Mosammaparast
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Peinan Zhao
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University, St Louis, Missouri
| | - Elena Lomonosova
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Dineo Khabele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| | - Mary M. Mullen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, Missouri
| |
Collapse
|
189
|
Liu K, Qin Z, Ge Y, Bian A, Xu X, Wu B, Xing C, Mao H. Acute kidney injury in advanced lung cancer patients treated with PD-1 inhibitors: a single center observational study. J Cancer Res Clin Oncol 2023; 149:5061-5070. [PMID: 36326913 DOI: 10.1007/s00432-022-04437-9] [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: 08/05/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Immune checkpoint inhibitor (ICI) therapy is now the stand of care for lung cancer. Due to the low incidence, the study of acute kidney injury (AKI) in lung cancer patients treated with ICIs was hardly reported. We focused on the incidence, characteristics, risk factors, and mortality of AKI in advanced lung cancer patients receiving PD-1 inhibitors. METHODS We reviewed advanced lung cancer patients receiving PD-1 inhibitors between January 2018 to August 2020 at Jiangsu Province Hospital. Patients were followed up for 6 months. We used the logistic regression model to evaluate risk factors for AKI, and Kaplan-Meier method to assess the association between AKI and mortality. RESULTS A total of 305 advanced lung cancer patients treated with PD-1 inhibitors. The median age was 64 years and 80.6% of patients were male. The incidence of AKI was 10.2%, and the incidence of ICI-AKI was 4.6%. Multivariate analysis showed that concomitant use of nonsteroidal anti-inflammatory drugs (NSAIDs) (OR 2.509; 95% CI 1.053-5.974) and renin-angiotensin-aldosterone system (RAAS) inhibitors (OR 2.656; 95% CI 1.091-6.466) were risk factors for AKI. In addition, concomitant use of NSAIDs (OR 5.170; 95% CI 1.087-24.595) and RAAS inhibitors (OR 5.921; 95% CI 1.871-18.737), and the occurrence of extra-renal immune-related adverse events (OR 4.726; 95% CI 1.462-15.280) were significantly associated with ICI-AKI. ICI-AKI was not associated with mortality while severe AKI was associated with higher risk of mortality. CONCLUSION AKI is common in advanced lung cancer patients treated with PD-1 inhibitors. The characteristics and risk factors of ICI-AKI were similar to those previously reported in other solid organ malignancies treated with ICIs. Severe AKI may indicate higher mortality.
Collapse
Affiliation(s)
- Kang Liu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Zhongke Qin
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
- Department of Nephrology, Jiangyin People's Hospital, Wuxi, 214400, China
| | - Yifei Ge
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Ao Bian
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Xueqiang Xu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Buyun Wu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Changying Xing
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Huijuan Mao
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China.
| |
Collapse
|
190
|
Zhao BW, Yang L, Long CY, Li HL, He YT, Wang XQ. Ni-Catalyzed Protecting Group Free Diphenic Acid Analog Synthesis. Org Lett 2023; 25:4700-4704. [PMID: 37314939 DOI: 10.1021/acs.orglett.3c01625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Severe side effects and drug resistance are major drawbacks of Pt-based chemotherapy in clinical practice, leading to the search for new Pt-based drugs through the tuning of coordination ligands. Therefore, seeking appropriate ligands has attracted significant interest in this area. In this study, we report a Ni-catalyzed coupling strategy for the divergent synthesis of diphenic acid derivatives and the application of these newly prepared acids in Pt(II) agent synthesis.
Collapse
Affiliation(s)
- Bo-Wei Zhao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), the Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Liu Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Cheng-Yu Long
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Han-Lu Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), the Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yu-Ting He
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), the Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xue-Qiang Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), the Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, Hangzhou Normal University, Hangzhou 311121, People's Republic of China
| |
Collapse
|
191
|
Liu G, Zhang Y, Yao H, Deng Z, Chen S, Wang Y, Peng W, Sun G, Tse MK, Chen X, Yue J, Peng YK, Wang L, Zhu G. An ultrasound-activatable platinum prodrug for sono-sensitized chemotherapy. SCIENCE ADVANCES 2023; 9:eadg5964. [PMID: 37343091 DOI: 10.1126/sciadv.adg5964] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/12/2023] [Indexed: 06/23/2023]
Abstract
Despite the great success achieved by photoactivated chemotherapy, eradicating deep tumors using external sources with high tissue penetration depth remains a challenge. Here, we present cyaninplatin, a paradigm of Pt(IV) anticancer prodrug that can be activated by ultrasound in a precise and spatiotemporally controllable manner. Upon sono-activation, mitochondria-accumulated cyaninplatin exhibits strengthened mitochondrial DNA damage and cell killing efficiency, and the prodrug overcomes drug resistance as a consequence of combined effects from released Pt(II) chemotherapeutics, the depletion of intracellular reductants, and the burst of reactive oxygen species, which gives rise to a therapeutic approach, namely sono-sensitized chemotherapy (SSCT). Guided by high-resolution ultrasound, optical, and photoacoustic imaging modalities, cyaninplatin realizes the overall theranostics of tumors in vivo with superior efficacy and biosafety. This work highlights the practical utility of ultrasound to precisely activate Pt(IV) anticancer prodrugs for the eradication of deep tumor lesions and broadens the biomedical uses of Pt coordination complexes.
Collapse
Affiliation(s)
- Gongyuan Liu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, P.R. China
| | - Yachao Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Houzong Yao
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, P.R. China
| | - Zhiqin Deng
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, P.R. China
| | - Shu Chen
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, P.R. China
| | - Yue Wang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Wang Peng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Guohan Sun
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Man-Kit Tse
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Xianfeng Chen
- School of Engineering, Institute for Bioengineering, University of Edinburgh, UK
| | - Jianbo Yue
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, P.R. China
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan 215316, P.R. China
| | - Yung-Kang Peng
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Lidai Wang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P.R. China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, P.R. China
| |
Collapse
|
192
|
Wang G, Guo H, Ren Y, Chen W, Wang Y, Li J, Liu H, Xing J, Zhang Y, Li N. Triptolide enhances carboplatin-induced apoptosis by inhibiting nucleotide excision repair (NER) activity in melanoma. Front Pharmacol 2023; 14:1157433. [PMID: 37324464 PMCID: PMC10267402 DOI: 10.3389/fphar.2023.1157433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Carboplatin (CBP) is a DNA damaging drug used to treat various cancers, including advanced melanoma. Yet we still face low response rates and short survival due to resistance. Triptolide (TPL) is considered to have multifunctional antitumor effects and has been confirmed to enhance the cytotoxic effects of chemotherapeutic drugs. Herein, we aimed to investigate the knowledge about the effects and mechanisms for the combined application of TPL and CBP against melanoma. Methods: Melanoma cell lines and xenograft mouse model were used to uncover the antitumor effects and the underlying molecular mechanisms of the alone or combined treatment of TPL and CBP in melanoma. Cell viability, migration, invasion, apoptosis, and DNA damage were detected by conventional methods. The rate-limiting proteins of the NER pathway were quantitated using PCR and Western blot. Fluorescent reporter plasmids were used to test the NER repair capacity. Results: Our results showed that the presence of TPL in CBP treatment could selectively inhibit NER pathway activity, and TPL exerts a synergistic effect with CBP to inhibit viability, migration, invasion, and induce apoptosis of A375 and B16 cells. Moreover, combined treatment with TPL and CBP significantly inhibited tumor progression in nude mice by suppressing cell proliferation and inducing apoptosis. Discussion: This study reveals the NER inhibitor TPL which has great potential in treating melanoma, either alone or in combination with CBP.
Collapse
Affiliation(s)
- Geng Wang
- Health Science Center, Ningbo University, Ningbo, China
| | - Hongmin Guo
- People’s Hospital of Changshou Chongqing, Chongqing, China
| | - Yan Ren
- Health Science Center, Ningbo University, Ningbo, China
| | - Weiyi Chen
- Health Science Center, Ningbo University, Ningbo, China
| | - Yixuan Wang
- Health Science Center, Ningbo University, Ningbo, China
| | - Jianing Li
- Health Science Center, Ningbo University, Ningbo, China
| | - Hua Liu
- Health Science Center, Ningbo University, Ningbo, China
| | - Jingjun Xing
- Health Science Center, Ningbo University, Ningbo, China
| | - Yanru Zhang
- Health Science Center, Ningbo University, Ningbo, China
| | - Na Li
- Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
193
|
Arceri L, Nguyen TK, Gibson S, Baker S, Wingert RA. Cannabinoid Signaling in Kidney Disease. Cells 2023; 12:1419. [PMID: 37408253 DOI: 10.3390/cells12101419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 07/07/2023] Open
Abstract
Endocannabinoid signaling plays crucial roles in human physiology in the function of multiple systems. The two cannabinoid receptors, CB1 and CB2, are cell membrane proteins that interact with both exogenous and endogenous bioactive lipid ligands, or endocannabinoids. Recent evidence has established that endocannabinoid signaling operates within the human kidney, as well as suggests the important role it plays in multiple renal pathologies. CB1, specifically, has been identified as the more prominent ECS receptor within the kidney, allowing us to place emphasis on this receptor. The activity of CB1 has been repeatedly shown to contribute to both diabetic and non-diabetic chronic kidney disease (CKD). Interestingly, recent reports of acute kidney injury (AKI) have been attributed to synthetic cannabinoid use. Therefore, the exploration of the ECS, its receptors, and its ligands can help provide better insight into new methods of treatment for a range of renal diseases. This review explores the endocannabinoid system, with a focus on its impacts within the healthy and diseased kidney.
Collapse
Affiliation(s)
- Liana Arceri
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shannon Gibson
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sophia Baker
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
194
|
Nasef SM, Khozemy EE, Mahmoud GA. pH-responsive chitosan/acrylamide/gold/nanocomposite supported with silver nanoparticles for controlled release of anticancer drug. Sci Rep 2023; 13:7818. [PMID: 37188828 DOI: 10.1038/s41598-023-34870-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023] Open
Abstract
In this study, we prepared a pH-responsive nanocomposite hydrogel based on chitosan grafted with acrylamide monomer and gold nanoparticles using gamma irradiation method (Cs-g-PAAm/AuNPs). The nanocomposite was enhanced with a layer coating of silver nanoparticles to improve the controlled release of the anticancer drug fluorouracil while increasing antimicrobial activity and decreasing the cytotoxicity of silver nanoparticles in nanocomposite hydrogel by combining with gold nanoparticles to enhance the ability to kill a high number of liver cancer cells. The structure of the nanocomposite materials was studied using FTIR spectroscopy and XRD patterns, which demonstrated the entrapment of gold and silver nanoparticles within the prepared polymer matrix. Dynamic light scattering data revealed the presence of gold and silver in the nanoscale with the polydispersity indexes in the mid-range values, indicating that distribution systems work best. Swelling experiments at various pH levels revealed that the prepared Cs-g-PAAm/Au-Ag-NPs nanocomposite hydrogels were highly responsive to pH changes. Bimetallic pH-responsive Cs-g-PAAm/Au-Ag-NPs nanocomposites exhibit strong antimicrobial activity. The presence of AuNPs reduced the cytotoxicity of AgNPs while increasing their ability to kill a high number of liver cancer cells.Cs-g-PAAm/Au-Ag-NPs has a high amount of fluorouracil drug loaded at pH 7.4 reaching 95 mg/g with a maximum drug release of 97% within 300 min. Cs-g-PAAm/Au-Ag-NPs have been recommended to use as oral delivery of anticancer drugs because they secure the encapsulated drug in the acidic medium of the stomach and release it in the intestinal pH.
Collapse
Affiliation(s)
- Shaimaa M Nasef
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Ehab E Khozemy
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ghada A Mahmoud
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
195
|
Zhu L, Liu H, Dou Y, Luo Q, Gu L, Liu X, Zhou Q, Han J, Wang F. A Photoactivated Ru (II) Polypyridine Complex Induced Oncotic Necrosis of A549 Cells by Activating Oxidative Phosphorylation and Inhibiting DNA Synthesis as Revealed by Quantitative Proteomics. Int J Mol Sci 2023; 24:ijms24097756. [PMID: 37175463 PMCID: PMC10178167 DOI: 10.3390/ijms24097756] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The ruthenium polypyridine complex [Ru(dppa)2(pytp)] (PF6)2 (termed as ZQX-1), where dppa = 4,7-diphenyl-1,10-phenanthroline and pytp = 4'-pyrene-2,2':6',2''-terpyridine, has been shown a high and selective cytotoxicity to hypoxic and cisplatin-resistant cancer cells either under irradiation with blue light or upon two-photon excitation. The IC50 values of ZQX-1 towards A549 cancer cells and HEK293 health cells are 0.16 ± 0.09 µM and >100 µM under irradiation at 420 nm, respectively. However, the mechanism of action of ZQX-1 remains unclear. In this work, using the quantitative proteomics method we identified 84 differentially expressed proteins (DEPs) with |fold-change| ≥ 1.2 in A549 cancer cells exposed to ZQX-1 under irradiation at 420 nm. Bioinformatics analysis of the DEPs revealed that photoactivated ZQX-1 generated reactive oxygen species (ROS) to activate oxidative phosphorylation signaling to overproduce ATP; it also released ROS and pyrene derivative to damage DNA and arrest A549 cells at S-phase, which synergistically led to oncotic necrosis and apoptosis of A549 cells to deplete excess ATP, evidenced by the elevated level of PRAP1 and cleaved capase-3. Moreover, the DNA damage inhibited the expression of DNA repair-related proteins, such as RBX1 and GPS1, enhancing photocytotoxicity of ZQX-1, which was reflected in the inhibition of integrin signaling and disruption of ribosome assembly. Importantly, the photoactivated ZQX-1 was shown to activate hypoxia-inducible factor 1A (HIF1A) survival signaling, implying that combining use of ZQX-1 with HIF1A signaling inhibitors may further promote the photocytotoxicity of the prodrug.
Collapse
Affiliation(s)
- Li Zhu
- College of Applied Science and Technology, Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100101, China
| | - Hui Liu
- College of Applied Science and Technology, Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100101, China
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Centre for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yang Dou
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Centre for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Centre for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liangzhen Gu
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Centre for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xingkai Liu
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Centre for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Qianxiong Zhou
- Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Juanjuan Han
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Centre for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Centre for Excellence in Molecular Sciences, National Centre for Mass Spectrometry in Beijing, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
196
|
Medvedíková M, Ranc V, Vančo J, Trávníček Z, Anzenbacher P. Highly Cytotoxic Copper(II) Mixed-Ligand Quinolinonato Complexes: Pharmacokinetic Properties and Interactions with Drug Metabolizing Cytochromes P450. Pharmaceutics 2023; 15:pharmaceutics15041314. [PMID: 37111801 PMCID: PMC10146558 DOI: 10.3390/pharmaceutics15041314] [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: 02/28/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
The effects of two anticancer active copper(II) mixed-ligand complexes of the type [Cu(qui)(mphen)]Y·H2O, where Hqui = 2-phenyl-3-hydroxy- 1H-quinolin-4-one, mphen = bathophenanthroline, and Y = NO3 (complex 1) or BF4 (complex 2) on the activities of different isoenzymes of cytochrome P450 (CYP) have been evaluated. The screening revealed significant inhibitory effects of the complexes on CYP3A4/5 (IC50 values were 2.46 and 4.88 μM), CYP2C9 (IC50 values were 16.34 and 37.25 μM), and CYP2C19 (IC50 values were 61.21 and 77.07 μM). Further, the analysis of mechanisms of action uncovered a non-competitive type of inhibition for both the studied compounds. Consequent studies of pharmacokinetic properties proved good stability of both the complexes in phosphate buffer saline (>96% stability) and human plasma (>91% stability) after 2 h of incubation. Both compounds are moderately metabolised by human liver microsomes (<30% after 1 h of incubation), and over 90% of the complexes bind to plasma proteins. The obtained results showed the potential of complexes 1 and 2 to interact with major metabolic pathways of drugs and, as a consequence of this finding, their apparent incompatibility in combination therapy with most chemotherapeutic agents.
Collapse
Affiliation(s)
- Martina Medvedíková
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacký University in Olomouc, Hněvotínská 3, 779 00 Olomouc, Czech Republic
- Institute of Molecular and Translation Medicine, Faculty of Medicine and Dentistry, Palacký University in Olomouc, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| | - Václav Ranc
- Institute of Molecular and Translation Medicine, Faculty of Medicine and Dentistry, Palacký University in Olomouc, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| | - Ján Vančo
- Regional Centre of Advanced Technologies and Materials (RCPTM), Czech Advanced Technology and Research Institute (CATRIN), Palacký University in Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Zdeněk Trávníček
- Regional Centre of Advanced Technologies and Materials (RCPTM), Czech Advanced Technology and Research Institute (CATRIN), Palacký University in Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Pavel Anzenbacher
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacký University in Olomouc, Hněvotínská 3, 779 00 Olomouc, Czech Republic
- Institute of Molecular and Translation Medicine, Faculty of Medicine and Dentistry, Palacký University in Olomouc, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| |
Collapse
|
197
|
Wang Z, Zhou L, Chen B, Li X, Zou Q, Xu W, Fang L, Wu A, Li Z, Chen Y. microRNA- 660 Enhances Cisplatin Sensitivity via Decreasing SATB2 Expression in Lung Adenocarcinoma. Genes (Basel) 2023; 14:genes14040911. [PMID: 37107669 PMCID: PMC10137726 DOI: 10.3390/genes14040911] [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: 02/02/2023] [Revised: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Increasing evidence suggests that microRNAs' (miRNAs) abnormal expression is one of the main factors of chemotherapy resistance in various cancers. However, the role of miRNAs in lung adenocarcinoma (LUAD) resistance to cisplatin is still unclear. In this study, we analyzed a microarray dataset to investigate miRNAs related to cisplatin resistance in LUAD. The expression of miRNAs in LUAD tissues and cell lines was detected using real-time quantitative polymerase chain reaction (RT-qPCR). Special AT-Rich Sequence-Binding Protein 2 (SATB2) in LUAD cell lines was detected using RT-qPCR and Western blot. Cell proliferation was measured by CCK8 and colony formation assays, while cell cycle and apoptosis were measured by flow cytometry. A dual-luciferase reporter assay was performed to confirm that SATB2 is a target gene of microRNA-660 (miR-660). We showed that the expression of miR-660 was not only decreased in LUAD cells and tissues but also further decreased in the cisplatin-resistant A549 cell line. The overexpression of miR-660 increased cisplatin sensitivity in LUAD cells. In addition, we identified SATB2 as a direct target gene of miR-660. We also revealed that miR-660 increased cisplatin sensitivity in LUAD cells via targeting SATB2. In conclusion, miR-660/SATB2 axis is a key regulator of cisplatin resistance in LUAD.
Collapse
Affiliation(s)
- Ziyao Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Lingxuan Zhou
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Bisong Chen
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Xu Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Qiuyi Zou
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Wei Xu
- NHC Key Laboratory of Carcinogenesis, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Li Fang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Anbang Wu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Zheng Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
- NHC Key Laboratory of Carcinogenesis, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Yuejun Chen
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| |
Collapse
|
198
|
Yang G, Lang Y. Extract identification and evaluation of the cytotoxic activity of Polygala fallax Hemsl in Heilongjiang ethnic medicine against tumors. Technol Health Care 2023; 31:565-575. [PMID: 37066951 DOI: 10.3233/thc-236050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND Heilongjiang Province is a frontier province with distinctive characteristics, fertile land and rich products. OBJECTIVE This study provides a new method for qualitatively studying flavonoids in traditional Chinese medicine and a new auxiliary means for identifying flavonoid isomers. METHODS The flavonoids in Polygala fallax Hemsl were identified by ultra-performance liquid chromatography-photo-diode array (PDA)-quadrupole-electro- static field orbitrap mass spectrometry tandem by UV Spectrum, primary and secondary high-resolution mass spectrometry (MS1/MS2) cleavage of fragments combined with databases, mass spectrometry cleavage patterns and literature. RESULTS The established QSRR model was used to verify the flavonoids identified from the Polygala fallax Hemsl. CONCLUSION The structure of multiple Polygala fallax Hemsl has been identified using various spectral methods. The tumor cytotoxic activity of the isolated compounds was evaluated. This paper is of great significance for further elucidating the pharmacodynamic substance basis and further developing and utilizing Polygala fallax Hemsl.
Collapse
Affiliation(s)
- Guang Yang
- Business Economics Research Institute, Harbin University of Commerce, Harbin, Heilongjiang, China
| | - Yan Lang
- Department of Rehabilitation Therapy, Wuyi University, Nanping, Fujian, China
| |
Collapse
|
199
|
El-Mahdy HA, Mohamadin AM, Abulsoud AI, Khidr EG, El-Husseiny AA, Ismail A, Elsakka EGE, Mokhlis HA, El-Husseiny HM, Doghish AS. miRNAs as potential game-changers in head and neck cancer: Future clinical and medicinal uses. Pathol Res Pract 2023; 245:154457. [PMID: 37058745 DOI: 10.1016/j.prp.2023.154457] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/16/2023]
Abstract
Head and neck cancers (HNCs) are a group of heterogeneous tumors formed most frequently from epithelial cells of the larynx, lips, oropharynx, nasopharynx, and mouth. Numerous epigenetic components, including miRNAs, have been demonstrated to have an impact on HNCs characteristics like progression, angiogenesis, initiation, and resistance to therapeutic interventions. The miRNAs may control the production of numerous genes linked to HNCs pathogenesis. The roles that miRNAs play in angiogenesis, invasion, metastasis, cell cycle, proliferation, and apoptosis are responsible for this impact. The miRNAs also have an impact on crucial HNCs-related mechanistic networks like the WNT/β-catenin signaling, PTEN/Akt/mTOR pathway, TGFβ, and KRAS mutations. miRNAs may affect how the HNCs respond to treatments like radiation and chemotherapy in addition to pathophysiology. This review aims to demonstrate the relationship between miRNAs and HNCs with a particular emphasis on how miRNAs impact HNCs signaling networks.
Collapse
Affiliation(s)
- Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt.
| | - Ahmed M Mohamadin
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt; Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr, Cairo 11829, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt
| | - Hamada Ahmed Mokhlis
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Nasr, Cairo 11231, Egypt
| | - Hussein M El-Husseiny
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Al Qalyubia 13736, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr, Cairo 11231, Egypt.
| |
Collapse
|
200
|
Saif-Elnasr M, El-Ghlban S, Bayomi AI, El-Sayyad GS, Maghraby MS. Gallic acid and/or cerium oxide nanoparticles synthesized by gamma-irradiation protect cisplatin-induced nephrotoxicity via modulating oxidative stress, inflammation and apoptosis. Arch Biochem Biophys 2023; 740:109594. [PMID: 37023935 DOI: 10.1016/j.abb.2023.109594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Cisplatin is one of the most significant anticancer. However, its use is associated with numerous toxicities especially nephrotoxicity. The main aim of this work was to examine the protective effect of Gallic acid (GA) and/or cerium oxide nanoparticles (CONPs) synthesized by gamma-irradiation on cisplatin-induced nephrotoxicity in rats. To do that, 48 adult male albino rats were separated into eight groups and received GA (100 mg/kg orally) and/or CONPs (15 mg/kg i. p.) for 10 days before injection with a single dose of cisplatin (7.5 mg/kg i. p.). The findings showed that cisplatin treatment impaired kidney functioning as shown by elevated serum levels of urea and creatinine. Additionally, the oxidative stress indicators (MDA and NO), levels of NF-kB, pro-inflammatory cytokines (IL1-and TNF-) and pro-apoptotic proteins (BAX and caspase-3) were raised after cisplatin injection, while levels of intrinsic anti-oxidants (CAT, SOD, and GSH) and anti-apoptotic protein (Bcl-2) were reduced. Moreover, renal toxicity was confirmed by alteration in normal histological architecture of the kidneys. On the other hand, pretreatment with CONPs and/or GA ameliorated cisplatin-induced nephrotoxicity as evidenced by improvement of renal function parameters and levels of oxidative stress, inflammatory and apoptotic markers in renal tissue along with the renal histopathological changes. This study clarifies how GA and CONPs protect against cisplatin-induced nephrotoxicity and demonstrates any potential synergism between them. Therefore, they can be considered as promising nephroprotective agents during chemotherapy.
Collapse
Affiliation(s)
- Mostafa Saif-Elnasr
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Samah El-Ghlban
- Biochemistry Division, Department of Chemistry, Faculty of Science, El Menoufia University, Shebin El-kom, Egypt
| | - Asmaa I Bayomi
- Zoology Department, Faculty of Science, Menoufia University, Menoufia, Egypt
| | - Gharieb S El-Sayyad
- Microbiology and Immunology Department, Faculty of Pharmacy, Ahram Canadian University (ACU), Giza, Egypt; Drug Microbiology Lab., Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| | - Mohamed Said Maghraby
- Biochemistry Division, Department of Chemistry, Faculty of Science, El Menoufia University, Shebin El-kom, Egypt.
| |
Collapse
|