1
|
Chu Y, Nie Q, Zhou X, Yang J, Fang J, Zhang J. Berberrubine as a novel TrxR inhibitor enhances cisplatin sensitivity in the treatment of non-small cell lung cancer. Bioorg Chem 2025; 158:108329. [PMID: 40056602 DOI: 10.1016/j.bioorg.2025.108329] [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: 01/09/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
Thioredoxin reductase (TrxR, TXNRD) is an essential enzyme implicated in the processes of cancer development and progression, positioning it as a promising target for cancer therapeutics. In this study, we employed target-based structural screening to identify berberrubine (BRB), a natural product characterized by an unprecedented isoquinoline scaffold that differs from known TrxR inhibitors. Our findings demonstrate that BRB serves as an effective inhibitor of TrxR, both in the context of the purified enzyme and within cancer cells. Since TrxR is highly expressed in non-small cell lung cancer (NSCLC) and is linked to patient prognosis and drug resistance, our results demonstrate, for the first time, that BRB can enhance the sensitivity of cisplatin to impede the proliferation of A549 cells, which was further confirmed in a xenograft model. The primary reason for cisplatin resistance in NSCLC is the DNA repair mechanism of apoptotic tumor cells. Our subsequent mechanistic investigation discovered that BRB selectively inhibits TrxR and impairs the biologically functional thioredoxin, which ultimately inhibits DNA synthesis and repair in cancer cells. Inhibition of TrxR by BRB led to a significant ROS accumulation in A549 cells, which contributed to oxidative stress-mediated apoptosis when used in combination with cisplatin. Our results conclude that BRB is a novel chemical entity of TrxR inhibitor that can increase the effectiveness of cisplatin in slowing down the growth of NSCLC both in vitro and in vivo. This provides a new perspective on the potential application of the combination of the two in the treatment of NSCLC.
Collapse
Affiliation(s)
- Yajun Chu
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Qiuying Nie
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Xiedong Zhou
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Junwei Yang
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China.; School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing 210094, China..
| | - Junmin Zhang
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China..
| |
Collapse
|
2
|
Batista JAD, de Oliveira RM, Lima CHM, Lana Júnior ML, dos Anjos VC, Bell MJV, Rocha MS. Probing the potential of rare earth elements in the development of new anticancer drugs: single molecule studies. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:187-194. [PMID: 39968169 PMCID: PMC11833172 DOI: 10.3762/bjnano.16.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025]
Abstract
We use optical tweezers and atomic force microscopy to investigate the potential of rare earth elements to be used as anticancer agents in the development of new chemotherapeutic drugs by characterizing the binding of three rare earths (ytterbium, neodymium, and erbium) to double-stranded DNA, which is one of the main targets for these drugs inside cells. The three elements presented a significant interaction with the biopolymer in buffers of physiological relevance, typically binding with very high equilibrium association constants (106 to 107 M-1) at the DNA grooves. Furthermore, neodymium and erbium can also induce a very strong compaction/condensation of the double helix at high concentrations, promoting DNA collapse at the single molecule level in a similar way to what occurs with classical DNA condensing agents such as polycations and depletants.
Collapse
Affiliation(s)
- Josiane A D Batista
- Departamento de Física, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Rayane M de Oliveira
- Departamento de Física, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Carlos H M Lima
- Departamento de Ciências Naturais, Universidade Federal do Acre, Rio Branco, Acre, Brazil
| | - Milton L Lana Júnior
- Departamento de Física, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Virgílio C dos Anjos
- Departamento de Física, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Maria J V Bell
- Departamento de Física, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Márcio S Rocha
- Departamento de Física, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| |
Collapse
|
3
|
Bełdzińska P, Galikowska-Bogut B, Zakrzewski M, Bury K, Jamrógiewicz M, Wyrzykowski D, Gołuński G, Sądej R, Piosik J. Platinum as both a drug and its modulator - Do platinum nanoparticles influence cisplatin activity? Chem Biol Interact 2025; 407:111365. [PMID: 39743036 DOI: 10.1016/j.cbi.2024.111365] [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: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Breast cancer was the most frequent cause of cancer death in females in 2022. Despite the development of personalized therapies, chemotherapy frequently remains the only available treatment method. However, the administration of classic antineoplastic drugs, like cisplatin (CDDP), often causes severe side effects and may lead to drug resistance making the therapy inefficient. Therefore, there is a great need for new, effective treatment regimens development. For this reason, we applied platinum nanoparticles (PtNPs) to verify if they can influence the CDDP activity with particular emphasis on the differences due to nanoparticles' sizes. We employed a broad spectrum of physicochemical methods, including Dynamic Light Scattering, Atomic Force Microscopy, Isothermal Titration Calorimetry, Fourier Transform Infrared Spectroscopy, and Near Infrared Spectroscopy and also Differential Scanning Calorimetry, to characterize the possible interactions between nanoparticles and CDDP. Moreover, the impact of PtNPs on CDDP biological activity was investigated using the Ames mutagenicity test on Salmonella enterica serovar Typhimurium TA102 and MTT assay on two breast cancer cell lines MDA-MB-231 and SKBR3. The obtained results revealed PtNPs direct interactions with CDDP dependent on the nanoparticles' size. Despite the lack of explicit confirmation of PtNPs aggregation by AFM imaging and DLS, further physicochemical methods indicated structural changes between nanoparticles alone and PtNPs-CDDP mixtures. Moreover, the biological assays confirmed that PtNPs decrease CDDP mutagenicity and also slightly increase its cytotoxicity on the chosen cell lines. The latter effects are ambiguous, nevertheless, provide a valuable basis for further research.
Collapse
Affiliation(s)
| | | | | | - Katarzyna Bury
- Laboratory of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | | | - Dariusz Wyrzykowski
- Department of General and Inorganic Chemistry, University of Gdansk, Gdansk, Poland
| | | | - Rafał Sądej
- Laboratory of Molecular Enzymology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Jacek Piosik
- Laboratory of Biophysics, University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
4
|
Oliveira RMD, Rezende AGSD, Campos DF, Ribeiro NDA, Rocha MS. DNA binding to small anionic ligands: the case of Orange G dye. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2025; 54:65-71. [PMID: 39920339 DOI: 10.1007/s00249-025-01733-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/15/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025]
Abstract
Here we advance in the understanding of nucleic acids interactions with small anionic ligands by characterizing the binding of the Orange G (OG) dye to double-stranded DNA via single molecule force spectroscopy. While there is no detectable interaction at low ionic strengths, we found that for [ Na + ] = 150 mM OG was able to interact with the double-helix via groove binding in a non-cooperative way, with a relatively high equilibrium association constant ( ∼ 10 5 M - 1 ) that is compatible to other classic DNA small ligands. Furthermore, experiments performed with a fixed OG concentration at various ionic strengths clearly show that the binding can be turned "on / off" by regulating the concentration of available counterions, a result that can guide the development of new synthetic ligands and shows how to modulate their interactions with nucleic acids. The present work therefore advances in evaluating the fundamental role of the ionic strength on the DNA interactions with small anionic ligands.
Collapse
Affiliation(s)
- Rayane M de Oliveira
- Departamento de Física, Universidade Federal de Viçosa, Av. P. H. Rolfs s/n, Viçosa, 36.570-900, Minas Gerais, Brazil
| | - Arthur G S de Rezende
- Departamento de Física, Universidade Federal de Viçosa, Av. P. H. Rolfs s/n, Viçosa, 36.570-900, Minas Gerais, Brazil
| | - Daniel F Campos
- Departamento de Física, Universidade Federal de Viçosa, Av. P. H. Rolfs s/n, Viçosa, 36.570-900, Minas Gerais, Brazil
| | - Neemias de A Ribeiro
- Departamento de Física, Universidade Federal de Viçosa, Av. P. H. Rolfs s/n, Viçosa, 36.570-900, Minas Gerais, Brazil
| | - Márcio S Rocha
- Departamento de Física, Universidade Federal de Viçosa, Av. P. H. Rolfs s/n, Viçosa, 36.570-900, Minas Gerais, Brazil.
| |
Collapse
|
5
|
Awasthi S, Dobrolecki LE, Sallas C, Zhang X, Li Y, Khazaei S, Ghosh S, Jeter CR, Liu J, Mills GB, Westin SN, Lewis MT, Peng W, Sood AK, Yap TA, Yi SS, McGrail DJ, Sahni N. UBA1 inhibition sensitizes cancer cells to PARP inhibitors. Cell Rep Med 2024; 5:101834. [PMID: 39626673 PMCID: PMC11722100 DOI: 10.1016/j.xcrm.2024.101834] [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: 04/19/2024] [Revised: 07/31/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
Therapeutic strategies targeting the DNA damage response, such as poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi), have revolutionized cancer treatment in tumors deficient in homologous recombination (HR). However, overcoming innate and acquired resistance to PARPi remains a significant challenge. Here, we employ a genome-wide CRISPR knockout screen and discover that the depletion of ubiquitin-activating enzyme E1 (UBA1) enhances sensitivity to PARPi in HR-proficient ovarian cancer cells. We show that silencing or pharmacological inhibition of UBA1 sensitizes multiple cell lines and organoid models to PARPi. Mechanistic studies uncover that UBA1 inhibition not only impedes HR repair to sensitize cells to PARP inhibition but also increases PARylation, which may subsequently be targeted by PARP inhibition. In vivo experiments using patient-derived xenografts demonstrate that combining PARP and UBA1 inhibition provided significant survival benefit compared to individual therapies with no detectable signs of toxicity, establishing this combination approach as a promising strategy to extend PARPi benefit.
Collapse
Affiliation(s)
- Sharad Awasthi
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Lacey E Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Christina Sallas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Xudong Zhang
- Department of Anatomic Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Li
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sima Khazaei
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sumanta Ghosh
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Collene R Jeter
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinsong Liu
- Department of Anatomic Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon B Mills
- Division of Oncological Science, Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR 97201, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S Stephen Yi
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA; Interdisciplinary Life Sciences Graduate Programs (ILSGP), College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA; Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Daniel J McGrail
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
6
|
Dutra JL, Honorato J, Graminha A, Moraes CAF, de Oliveira KT, Cominetti MR, Castellano EE, Batista AA. Pd(II)/diphosphine/curcumin complexes as potential anticancer agents. Dalton Trans 2024; 53:18902-18916. [PMID: 38938129 DOI: 10.1039/d4dt01045k] [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: 06/29/2024]
Abstract
Palladium(II) complexes have stimulated research interest mainly due to their in vitro cytotoxicity against various cancer cell lines and their low cytotoxicity in healthy cells. Thus, in this work, we combined Pd(II)/phosphine systems with the natural product curcumin as a ligand, obtaining a series of complexes, [Pd(cur)(PPh3)2]PF6 (A1), [Pd(cur)(dppe)]PF6 (A2), [Pd(cur)(dppp)]PF6 (A3), [Pd(cur)(dppb)]PF6 (A4) and [Pd(cur)(dppf)]PF6 (A5), where dppe = 1,2-bis(diphenylphosphino)ethane, dppp = 1,3-bis(diphenylphosphino)propane, dppb = 1,4-bis(diphenylphosphino)butane, and dppf = 1,1'-bis(diphenylphosphino)ferrocene (P-P), which were characterized by elemental analysis, molar conductivity analysis, and mass, NMR (1H, 13C, 31P{1H}), UV-vis, and IR spectroscopies, and four of them (A1, A2, A4, and A5) by X-ray crystallography. The in vitro cell viability of the complexes A1-A5, cisplatin, and the free ligand curcumin against MDA-MB-231 (human triple-negative breast tumor cells), SK-BR-3 (human breast tumor cells), A549 (human lung tumor cells), MRC-5 (non-tumor human lung cells), A2780 (human ovarian carcinoma cells), and A2780cis (cisplatin-resistant human ovarian carcinoma cells), was evaluated by the MTT colorimetric assay. For the tumor cell lines tested, the complexes showed good anticancer activities. The results showed that in general the complexes had lower IC50 values than free curcumin and the precursors [PdCl2(P-P)]. IC50 results obtained for the A1-A5 complexes, in the MCF-7 cell line, are similar to those that had already been observed for some Pd/bipy/curcumin complexes. In the MDA-MB-231 cell line, complexes A1 and A5 stood out, with their lowest IC50 values, around 5 μmol L-1, and the complexes appeared to be more active (lower IC50 values) against the ovarian cell lines. Complex A1 was 23 and 22-fold more cytotoxic than cisplatin, against the A2780 and A2780cis cells, respectively. The complex A1 was studied on A2780cis cells and it was found that this complex inhibits colony formation and induces cell cycle arrest in the sub-G1 phase in a concentration-dependent manner and leads to cell death by apoptosis. The DCFDA assay revealed a potent ROS induction for complex A1.
Collapse
Affiliation(s)
- Jocely L Dutra
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, CP 676, CEP 13561-901, São Carlos, SP, Brazil.
- Departamento de Química, Universidade Federal do Amazonas - UFAM, CEP 69077-000, Itacoatiara, AM, Brazil
| | - João Honorato
- Instituto de Química, Universidade de São Paulo - USP, CEP 05508-900, São Paulo, SP, Brazil
| | - Angélica Graminha
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, CP 676, CEP 13561-901, São Carlos, SP, Brazil.
| | - Carlos André F Moraes
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, CP 676, CEP 13561-901, São Carlos, SP, Brazil.
| | - Kleber T de Oliveira
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, CP 676, CEP 13561-901, São Carlos, SP, Brazil.
| | - Marcia R Cominetti
- Departamento de Gerontologia, Universidade Federal de São Carlos - UFSCar, CP 676, 13561-901, São Carlos, SP, Brazil
| | - Eduardo E Castellano
- Instituto de Física de São Carlos, Universidade de São Paulo - USP, CP 369, CEP 13560-970, São Carlos, SP, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, CP 676, CEP 13561-901, São Carlos, SP, Brazil.
| |
Collapse
|
7
|
Kesimoglu ZN, Bozdag S. Fusing multiplex heterogeneous networks using graph attention-aware fusion networks. Sci Rep 2024; 14:29119. [PMID: 39582056 PMCID: PMC11586420 DOI: 10.1038/s41598-024-78555-4] [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: 04/01/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
Graph Neural Networks (GNN) emerged as a deep learning framework to generate node and graph embeddings for downstream machine learning tasks. Popular GNN-based architectures operate on networks of single node and edge type. However, a large number of real-world networks include multiple types of nodes and edges. Enabling these architectures to work on networks with multiple node and edge types brings additional challenges due to the heterogeneity of the networks and the multiplicity of the existing associations. In this study, we present a framework, named GRAF (Graph Attention-aware Fusion Networks), to convert multiplex heterogeneous networks to homogeneous networks to make them more suitable for graph representation learning. Using attention-based neighborhood aggregation, GRAF learns the importance of each neighbor per node (called node-level attention) followed by the importance of each network layer (called network layer-level attention). Then, GRAF processes a network fusion step weighing each edge according to the learned attentions. After an edge elimination step based on edge weights, GRAF utilizes Graph Convolutional Networks (GCN) on the fused network and incorporates node features on graph-structured data for a node classification or a similar downstream task. To demonstrate GRAF's generalizability, we applied it to four datasets from different domains and observed that GRAF outperformed or was on par with the baselines and state-of-the-art (SOTA) methods. We were able to interpret GRAF's findings utilizing the attention weights. Source code for GRAF is publicly available at https://github.com/bozdaglab/GRAF .
Collapse
Affiliation(s)
- Ziynet Nesibe Kesimoglu
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, USA
- BioDiscovery Institute, University of North Texas, Denton, TX, USA
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, USA.
- Department of Mathematics, University of North Texas, Denton, TX, USA.
- BioDiscovery Institute, University of North Texas, Denton, TX, USA.
| |
Collapse
|
8
|
Yu W, Fu J, Jia F, Jin Q, Wang Y, Ji J. Removable Photocatalysis Microneedle Reactor for Carbon Monoxide Delivery to Enhance Chemosensitization. NANO LETTERS 2024; 24:10024-10031. [PMID: 39115188 DOI: 10.1021/acs.nanolett.4c01582] [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: 08/22/2024]
Abstract
Carbon monoxide (CO) has emerged as a promising therapeutic agent, yet ensuring safe and precise CO delivery remains challenging. Here, we report a removable hydrogel-forming microneedle (MN) reactor for CO delivery via photocatalysis, with an emphasis on chemosensitization. Upon application, body fluids absorbed by the MNs dissolve the effervescent agents, leading to the generation of carbon dioxide (CO2) and triggering the release of the chemotherapeutics cisplatin. Meanwhile, the photocatalysts (PCs) trapped within MNs convert CO2 to CO under 660 nm light irradiation. These PCs can be removed by hydrogel-forming MNs, thereby mitigating potential biological risks associated with residual PCs. Both in vitro and in vivo experiments showed that MN-mediated CO delivery significantly improved tumor sensitivity to cisplatin by suppressing DNA repair, using an A375/CDDP melanoma model. This removable photocatalysis MN reactor offers safe and precise local delivery of CO, potentially creating new opportunities for CO or its combination therapies.
Collapse
Affiliation(s)
- Weijiang Yu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
| | - Junzhe Fu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
| | - Fan Jia
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, P. R. China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
| |
Collapse
|
9
|
Kerdkumthong K, Roytrakul S, Songsurin K, Pratummanee K, Runsaeng P, Obchoei S. Proteomics and Bioinformatics Identify Drug-Resistant-Related Genes with Prognostic Potential in Cholangiocarcinoma. Biomolecules 2024; 14:969. [PMID: 39199357 PMCID: PMC11352417 DOI: 10.3390/biom14080969] [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: 06/02/2024] [Revised: 07/21/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Drug resistance is a major challenge in the treatment of advanced cholangiocarcinoma (CCA). Understanding the mechanisms of drug resistance can aid in identifying novel prognostic biomarkers and therapeutic targets to improve treatment efficacy. This study established 5-fluorouracil- (5-FU) and gemcitabine-resistant CCA cell lines, KKU-213FR and KKU-213GR, and utilized comparative proteomics to identify differentially expressed proteins in drug-resistant cells compared to parental cells. Additionally, bioinformatics analyses were conducted to explore the biological and clinical significance of key proteins. The drug-resistant phenotypes of KKU-213FR and KKU-213GR cell lines were confirmed. In addition, these cells demonstrated increased migration and invasion abilities. Proteomics analysis identified 81 differentially expressed proteins in drug-resistant cells, primarily related to binding functions, biological regulation, and metabolic processes. Protein-protein interaction analysis revealed a highly interconnected network involving MET, LAMB1, ITGA3, NOTCH2, CDH2, and NDRG1. siRNA-mediated knockdown of these genes in drug-resistant cell lines attenuated cell migration and cell invasion abilities and increased sensitivity to 5-FU and gemcitabine. The mRNA expression of these genes is upregulated in CCA patient samples and is associated with poor prognosis in gastrointestinal cancers. Furthermore, the functions of these proteins are closely related to the epithelial-mesenchymal transition (EMT) pathway. These findings elucidate the potential molecular mechanisms underlying drug resistance and tumor progression in CCA, providing insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Kankamol Kerdkumthong
- Department of Biochemistry, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai District, Songkhla 90110, Thailand; (K.K.); (K.S.); (K.P.); (P.R.)
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Tani 12120, Thailand;
| | - Kawinnath Songsurin
- Department of Biochemistry, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai District, Songkhla 90110, Thailand; (K.K.); (K.S.); (K.P.); (P.R.)
| | - Kandawasri Pratummanee
- Department of Biochemistry, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai District, Songkhla 90110, Thailand; (K.K.); (K.S.); (K.P.); (P.R.)
| | - Phanthipha Runsaeng
- Department of Biochemistry, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai District, Songkhla 90110, Thailand; (K.K.); (K.S.); (K.P.); (P.R.)
- Center of Excellence for Biochemistry, Faculty of Science, Prince of Songkla University, Hat Yai District, Songkhla 90110, Thailand
| | - Sumalee Obchoei
- Department of Biochemistry, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai District, Songkhla 90110, Thailand; (K.K.); (K.S.); (K.P.); (P.R.)
- Center of Excellence for Biochemistry, Faculty of Science, Prince of Songkla University, Hat Yai District, Songkhla 90110, Thailand
| |
Collapse
|
10
|
Zhou Y, Chen H, Yan J, Yao Q, Kong C, Peng Y, Xiao S, Yang J. FOXA2 Activates RND1 to Regulate Arachidonic Acid Metabolism Pathway and Suppress Cisplatin Resistance in Lung Squamous Cell Carcinoma. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13814. [PMID: 39129202 PMCID: PMC11317498 DOI: 10.1111/crj.13814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/09/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND The primary cause of cancer-related fatalities globally is lung cancer. Although the chemotherapy drug cisplatin (DDP) has brought certain benefits to patients, the rapid development of drug resistance has greatly hindered treatment success. METHODS We used the lung squamous cell carcinoma (LUSC) mRNA data set to explore the differentially expressed gene (RND1) in LUSC and detected RND1 expression in LUSC cells and DDP-resistant cells by qRT-PCR. Meanwhile, we performed abnormal expression treatment on RND1 and conducted CCK8, colony formation, and flow cytometry to evaluate the impact of RND1 expression on cell proliferation, apoptosis, and DDP resistance. In addition, we analyzed metabolism pathways involving RND1 using GSEA. We also used online tools such as hTFtarget and JASPAR to screen for the upstream transcription factor FOXA2 of RND1 and verified their relationship through CHIP and dual luciferase experiments. Finally, we validated the role of FOXA2-RND1 in DDP resistance in LUSC through the above experiments. RESULTS RND1 was downregulated in LUSC, and overexpression of RND1 repressed proliferation and DDP resistance of LUSC cells and facilitated cell apoptosis. RND1 modulated the arachidonic acid (AA) metabolism pathway, and FOXA2 positively manipulated RND1 expression. By activating FOXA2, stabilizing RND1, and regulating AA levels, the sensitivity of LUSC cells to DDP could be enhanced. CONCLUSION Our study suggested that FOXA2 positively modulated the RND1-AA pathway, which repressed the resistance of LUSC cells to DDP.
Collapse
Affiliation(s)
- Yafu Zhou
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Huiguo Chen
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Jianhua Yan
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Qi Yao
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Chunchu Kong
- Department of RespiratoryHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - You Peng
- Department of GeriatricHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Shengying Xiao
- Department of OncologyHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Jinsong Yang
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| |
Collapse
|
11
|
Nasimi Shad A, Akhlaghipour I, Alshakarchi HI, Saburi E, Moghbeli M. Role of microRNA-363 during tumor progression and invasion. J Physiol Biochem 2024; 80:481-499. [PMID: 38691273 DOI: 10.1007/s13105-024-01022-1] [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: 12/21/2023] [Accepted: 04/05/2024] [Indexed: 05/03/2024]
Abstract
Recent progresses in diagnostic and therapeutic methods have significantly improved prognosis in cancer patients. However, cancer is still considered as one of the main causes of human deaths in the world. Late diagnosis in advanced tumor stages can reduce the effectiveness of treatment methods and increase mortality rate of cancer patients. Therefore, investigating the molecular mechanisms of tumor progression can help to introduce the early diagnostic markers in these patients. MicroRNA (miRNAs) has an important role in regulation of pathophysiological cellular processes. Due to their high stability in body fluids, they are always used as the non-invasive markers in cancer patients. Since, miR-363 deregulation has been reported in a wide range of cancers, we discussed the role of miR-363 during tumor progression and metastasis. It has been reported that miR-363 has mainly a tumor suppressor function through the regulation of transcription factors, apoptosis, cell cycle, and structural proteins. MiR-363 also affected the tumor progression via regulation of various signaling pathways such as WNT, MAPK, TGF-β, NOTCH, and PI3K/AKT. Therefore, miR-363 can be introduced as a probable therapeutic target as well as a non-invasive diagnostic marker in cancer patients.
Collapse
Affiliation(s)
- Arya Nasimi Shad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hawraa Ibrahim Alshakarchi
- Al-Zahra Center for Medical and Pharmaceutical Research Sciences (ZCMRS), Al-Zahraa University for Women, Karbala, Iraq
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
La Force H, Freindorf M, Kraka E. Ligand Characterization and DNA Intercalation of Ru(II) Polypyridyl Complexes: A Local Vibrational Mode Study. J Phys Chem A 2024; 128:5925-5940. [PMID: 38990174 DOI: 10.1021/acs.jpca.4c02954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
We investigated in this work ruthenium-ligand bonding across the RuN framework in 12 Ru(II) polypyridyl complexes in the gas phase and solution for both singlet and triplet states, in addition to their affinity for DNA binding through π-π stacking interactions with DNA nucleobases. As a tool to assess the intrinsic strength of the ruthenium-ligand bonds, we determined local vibrational force constants via our local vibrational mode analysis software. We introduced a novel local force constant that directly accounts for the intrinsic strength of the π-π stacking interaction between DNA and the intercalated Ru(II) complex. According to our findings, [Ru(phen)2(dppz)]2+ and [Ru(phen)2(11-CN-dppz)]2+ provide an intriguing trade-off between photoinduced complex excitation and the strength of the subsequent π-π stacking interaction with DNA. [Ru(phen)2(dppz)]2+ displays a small singlet-triplet splitting and a strong π-π stacking interaction in its singlet state, suggesting a favorable photoexcitation but potentially weaker interaction with DNA in the excited state. Conversely, [Ru(phen)2(11-CN-dppz)]2+ exhibits a larger singlet-triplet splitting and a stronger π-π stacking interaction with DNA in its triplet state, indicating a less favorable photoinduced transition but a stronger interaction with DNA postexcitation. We hope our study will inspire future experimental and computational work aimed at the design of novel Ru-polypyridyl drug candidates and that our new quantitative measure of π-π stacking interactions in DNA will find a general application in the field.
Collapse
Affiliation(s)
- Hunter La Force
- Computational and Theoretical Chemistry Group (CATCO), Department of Chemistry, Southern Methodist University, 3215 Daniel Avenue, Dallas, Texas 75275-0314, United States
| | - Marek Freindorf
- Computational and Theoretical Chemistry Group (CATCO), Department of Chemistry, Southern Methodist University, 3215 Daniel Avenue, Dallas, Texas 75275-0314, United States
| | - Elfi Kraka
- Computational and Theoretical Chemistry Group (CATCO), Department of Chemistry, Southern Methodist University, 3215 Daniel Avenue, Dallas, Texas 75275-0314, United States
| |
Collapse
|
13
|
Hu Y, Mostert D, Orgler C, Andler O, Zischka H, Kazmaier U, Vollmar AM, Braig S, Sieber SA, Zahler S. Thermal Proteome Profiling Reveals Insight to Antiproliferative and Pro-Apoptotic Effects of Lagunamide A in the Modulation of DNA Damage Repair. Chembiochem 2024; 25:e202400024. [PMID: 38716781 DOI: 10.1002/cbic.202400024] [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: 01/09/2024] [Revised: 05/06/2024] [Indexed: 06/18/2024]
Abstract
Lagunamide A is a biologically active natural product with a yet unidentified molecular mode of action. Cellular studies revealed that lagunamide A is a potent inhibitor of cancer cell proliferation, promotes apoptosis and causes mitochondrial dysfunction. To decipher the cellular mechanism responsible for these effects, we utilized thermal protein profiling (TPP) and identified EYA3 as a stabilized protein in cells upon lagunamide A treatment. EYA3, involved in the DNA damage repair process, was functionally investigated via siRNA based knockdown studies and corresponding effects of lagunamide A on DNA repair were confirmed. Furthermore, we showed that lagunamide A sensitized tumor cells to treatment with the drug doxorubicin highlighting a putative therapeutic strategy.
Collapse
Affiliation(s)
- Yudong Hu
- Department of Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5, 81377, Munich, Germany
| | - Dietrich Mostert
- Center for Functional Protein Assemblies (CPA), Department of Chemistry, Chair of Organic Chemistry II, Technical University of Munich, 85748, Garching, Germany
| | - Christina Orgler
- Department of Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5, 81377, Munich, Germany
| | - Oliver Andler
- Organic Chemistry, Saarland University, 66123, Saarbrücken, Germany
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health Neuherberg, Germany
- Institute of Toxicology and Environmental Hygiene, Technical University of Munich, Munich, Germany
| | - Uli Kazmaier
- Organic Chemistry, Saarland University, 66123, Saarbrücken, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5, 81377, Munich, Germany
| | - Simone Braig
- Department of Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5, 81377, Munich, Germany
| | - Stephan A Sieber
- Center for Functional Protein Assemblies (CPA), Department of Chemistry, Chair of Organic Chemistry II, Technical University of Munich, 85748, Garching, Germany
| | - Stefan Zahler
- Department of Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5, 81377, Munich, Germany
| |
Collapse
|
14
|
Tang H, Pang X, Li S, Tang L. The Double-Edged Effects of MLN4924: Rethinking Anti-Cancer Drugs Targeting the Neddylation Pathway. Biomolecules 2024; 14:738. [PMID: 39062453 PMCID: PMC11274557 DOI: 10.3390/biom14070738] [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: 04/23/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: The neddylation pathway assumes a pivotal role in the initiation and progression of cancer. MLN4924, a potent small-molecule inhibitor of the NEDD8-activating enzyme (NAE), effectively intervenes in the early stages of the neddylation pathway. By instigating diverse cellular responses, such as senescence and apoptosis in cancer cells, MLN4924 also exerts regulatory effects on non-malignant cells within the tumor microenvironment (TME) and tumor virus-infected cells, thereby impeding the onset of tumors. Consequently, MLN4924 has been widely acknowledged as a potent anti-cancer drug. (2) Recent findings: Nevertheless, recent findings have illuminated additional facets of the neddylation pathway, revealing its active involvement in various biological processes detrimental to the survival of cancer cells. This newfound understanding underscores the dual role of MLN4924 in tumor therapy, characterized by both anti-cancer and pro-cancer effects. This dichotomy is herein referred to as the "double-edged effects" of MLN4924. This paper delves into the intricate relationship between the neddylation pathway and cancer, offering a mechanistic exploration and analysis of the causes underlying the double-edged effects of MLN4924-specifically, the accumulation of pro-cancer neddylation substrates. (3) Perspectives: Here, the objective is to furnish theoretical support and novel insights that can guide the development of next-generation anti-cancer drugs targeting the neddylation pathway.
Collapse
Affiliation(s)
- Haoming Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (H.T.); (X.P.)
| | - Xin Pang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (H.T.); (X.P.)
| | - Shun Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
- Department of Spine Surgery, People’s Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen 518109, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (H.T.); (X.P.)
| |
Collapse
|
15
|
Han D, Villanueva-Tagle ME, Peña-Icart M, López-Mesas M, Valiente M. Trace cisplatin adsorption by thiol-functionalized sponge (TFS) and Sn/SnO 2-coated TFS: Adsorption study and mechanism investigation. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134442. [PMID: 38688222 DOI: 10.1016/j.jhazmat.2024.134442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/22/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
To remove trace cisplatin from aqueous solution, commercial sponges were functionalized by esterification with 3-mercaptopropionic acid, followed by reduction with Na2S·9H2O or SnCl2·2H2O. The resulting thiol-functionalized sponges (TFSs), TFS_1 and TFS_2, were tested for the removal of cisplatin (235 μg L-1) achieving maximum removal of 95.5 ± 0.8% and 99.5 ± 0.1% respectively, which were significantly higher than the non-functionalized counterpart. The successful grafting of thiol groups, verified through FTIR, elemental analysis, SEM-EDS, and XPS characterization, facilitated Pt-S complexation during adsorption. The aqua-derivatives of cisplatin, formed through hydration, complexed with thiol sites through ligand displacement. Additionally, the presence of Sn/SnO2 coating on TFS_2 further enhanced the adsorption process. The rapid adsorption process conformed to pseudo-second-order kinetic model, involving both diffusion and chemisorption. While the Langmuir isotherm model generally described the monolayer adsorption behavior of cisplatin, the aggregation of Sn/SnO2 onto TFS_2 at 343 K introduced surface heterogeneity, rendering the Freundlich model a better fit for the adsorption isotherm. Differential pH dependence and the evaluation of mean free energy, derived from the Dubinin-Radushkevich isotherm model, indicated that cisplatin adsorption onto TFS_1 involved physisorption, including electrostatic attraction, while chemisorption predominated for TFS_2. Increasing the temperature notably promoted adsorption by facilitating the thermal-favored formation of Pt-S bonds.
Collapse
Affiliation(s)
- Dong Han
- GTS-UAB Research Group, Department of Chemistry, Faculty of Science, Universitat Autònoma de Barcelona, Bellaterra, (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | | | - Mirella Peña-Icart
- Institute of Materials Science and Technology, University of Havana, Havana 10400, Cuba
| | - Montserrat López-Mesas
- GTS-UAB Research Group, Department of Chemistry, Faculty of Science, Universitat Autònoma de Barcelona, Bellaterra, (Cerdanyola del Vallès), 08193 Barcelona, Spain.
| | - Manuel Valiente
- GTS-UAB Research Group, Department of Chemistry, Faculty of Science, Universitat Autònoma de Barcelona, Bellaterra, (Cerdanyola del Vallès), 08193 Barcelona, Spain
| |
Collapse
|
16
|
Zhou Q, Meng Y, Li D, Yao L, Le J, Liu Y, Sun Y, Zeng F, Chen X, Deng G. Ferroptosis in cancer: From molecular mechanisms to therapeutic strategies. Signal Transduct Target Ther 2024; 9:55. [PMID: 38453898 PMCID: PMC10920854 DOI: 10.1038/s41392-024-01769-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 159.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/21/2024] [Accepted: 02/03/2024] [Indexed: 03/09/2024] Open
Abstract
Ferroptosis is a non-apoptotic form of regulated cell death characterized by the lethal accumulation of iron-dependent membrane-localized lipid peroxides. It acts as an innate tumor suppressor mechanism and participates in the biological processes of tumors. Intriguingly, mesenchymal and dedifferentiated cancer cells, which are usually resistant to apoptosis and traditional therapies, are exquisitely vulnerable to ferroptosis, further underscoring its potential as a treatment approach for cancers, especially for refractory cancers. However, the impact of ferroptosis on cancer extends beyond its direct cytotoxic effect on tumor cells. Ferroptosis induction not only inhibits cancer but also promotes cancer development due to its potential negative impact on anticancer immunity. Thus, a comprehensive understanding of the role of ferroptosis in cancer is crucial for the successful translation of ferroptosis therapy from the laboratory to clinical applications. In this review, we provide an overview of the recent advancements in understanding ferroptosis in cancer, covering molecular mechanisms, biological functions, regulatory pathways, and interactions with the tumor microenvironment. We also summarize the potential applications of ferroptosis induction in immunotherapy, radiotherapy, and systemic therapy, as well as ferroptosis inhibition for cancer treatment in various conditions. We finally discuss ferroptosis markers, the current challenges and future directions of ferroptosis in the treatment of cancer.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Lei Yao
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yihuang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
17
|
Meng D, Ren M, Li M, Wang M, Geng W, Shang Q. Molecular mechanism of α-Hederin in tumor progression. Biomed Pharmacother 2024; 170:116097. [PMID: 38160624 DOI: 10.1016/j.biopha.2023.116097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
α-Hederin is a monosaccharide pentacyclic triterpene saponin compound derived from the Chinese herb, Pulsatilla. It has garnered considerable attention for its anti-tumor, anti-inflammatory, and spasmolytic pharmacological activities. Given the rising incidence of cancer and the pronounced adverse reactions associated with chemotherapy drugs-which profoundly impact the quality of life for cancer patients-there is an immediate need for safe and effective antitumor agents. Traditional drugs and their anticancer effects have become a focal point of research in recent years. Studies indicate that α-Hederin can hinder tumor cell proliferation and impede the advancement of various cancers, including breast, lung, colorectal, and liver cancers. The principal mechanism behind its anti-tumor activity involves inhibiting tumor cell proliferation, facilitating tumor cell apoptosis, and arresting the cell cycle process. Current evidence suggests that α-Hederin can exert its anti-tumor properties through diverse mechanisms, positioning it as a promising agent in anti-tumor therapy. However, a comprehensive literature search revealed a gap in the comprehensive understanding of α-Hederin. This paper aims to review the available literature on the anti-tumor mechanisms of α-Hederin, hoping to provide valuable insights for the clinical treatment of malignant tumors and the innovation of novel anti-tumor medications.
Collapse
Affiliation(s)
- Dandan Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Meng Ren
- Department of Physical Education, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Maofeng Li
- College of Foreign Chinese, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Min Wang
- Experimental Center of Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Wei Geng
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238, Jingshi East Road, Lixia District, Jinan 250014, China
| | - Qingxin Shang
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China.
| |
Collapse
|
18
|
Marima R, Mosoane B, Mtshali N, Basera A, Kgatle M, Grech G, Dlamini Z. Mechanisms of chemotherapy resistance in cervical cancer. STRATEGIES FOR OVERCOMING CHEMOTHERAPY RESISTANCE IN CERVICAL CANCER 2024:53-70. [DOI: 10.1016/b978-0-443-28985-9.00012-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Fallet M, Wilson R, Sarkies P. Cisplatin exposure alters tRNA-derived small RNAs but does not affect epimutations in C. elegans. BMC Biol 2023; 21:276. [PMID: 38031056 PMCID: PMC10688063 DOI: 10.1186/s12915-023-01767-z] [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: 07/20/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND The individual lifestyle and environment of an organism can influence its phenotype and potentially the phenotype of its offspring. The different genetic and non-genetic components of the inheritance system and their mutual interactions are key mechanisms to generate inherited phenotypic changes. Epigenetic changes can be transmitted between generations independently from changes in DNA sequence. In Caenorhabditis elegans, epigenetic differences, i.e. epimutations, mediated by small non-coding RNAs, particularly 22G-RNAs, as well as chromatin have been identified, and their average persistence is three to five generations. In addition, previous research showed that some epimutations had a longer duration and concerned genes that were enriched for multiple components of xenobiotic response pathways. These results raise the possibility that environmental stresses might change the rate at which epimutations occur, with potential significance for adaptation. RESULTS In this work, we explore this question by propagating C. elegans lines either in control conditions or in moderate or high doses of cisplatin, which introduces genotoxic stress by damaging DNA. Our results show that cisplatin has a limited effect on global small non-coding RNA epimutations and epimutations in gene expression levels. However, cisplatin exposure leads to increased fluctuations in the levels of small non-coding RNAs derived from tRNA cleavage. We show that changes in tRNA-derived small RNAs may be associated with gene expression changes. CONCLUSIONS Our work shows that epimutations are not substantially altered by cisplatin exposure but identifies transient changes in tRNA-derived small RNAs as a potential source of variation induced by genotoxic stress.
Collapse
Affiliation(s)
- Manon Fallet
- Department of Biochemistry, Evolutionary Epigenetics Group, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Rd., Oxford, OX1 3QU, UK.
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, 70182, Örebro, Sweden.
| | - Rachel Wilson
- Department of Biochemistry, Evolutionary Epigenetics Group, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Rd., Oxford, OX1 3QU, UK
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Peter Sarkies
- Department of Biochemistry, Evolutionary Epigenetics Group, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Rd., Oxford, OX1 3QU, UK.
| |
Collapse
|
20
|
α-Hederin Saponin Augments the Chemopreventive Effect of Cisplatin against Ehrlich Tumors and Bioinformatic Approach Identifying the Role of SDF1/CXCR4/p-AKT-1/NFκB Signaling. Pharmaceuticals (Basel) 2023; 16:ph16030405. [PMID: 36986504 PMCID: PMC10056433 DOI: 10.3390/ph16030405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/10/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF1) and its C-X-C chemokine receptor type 4 receptor (CXCR4) are significant mediators for cancer cells’ proliferation, and we studied their expression in Ehrlich solid tumors (ESTs) grown in mice. α-Hederin is a pentacyclic triterpenoid saponin found in Hedera or Nigella species with biological activity that involves suppression of growth of breast cancer cell lines. The aim of this study was to explore the chemopreventive activity of α-hederin with/without cisplatin; this was achieved by measuring the reduction in tumor masses and the downregulation in SDF1/CXCR4/pAKT signaling proteins and nuclear factor kappa B (NFκB). Ehrlich carcinoma cells were injected in four groups of Swiss albino female mice (Group1: EST control group, Group2: EST + α-hederin group, Group3: EST + cisplatin group, and Group4: EST+α-hederin/cisplatin treated group). Tumors were dissected and weighed, one EST was processed for histopathological staining with hematoxylin and eosin (HE), and the second MC was frozen and processed for estimation of signaling proteins. Computational analysis for these target proteins interactions showed direct-ordered interactions. The dissected solid tumors revealed decreases in tumor masses (~21%) and diminished viable tumor regions with significant necrotic surrounds, particularly with the combination regimens. Immunohistochemistry showed reductions (~50%) in intratumoral NFκβ in the mouse group that received the combination therapy. The combination treatment lowered the SDF1/CXCR4/p-AKT proteins in ESTs compared to the control. In conclusion, α-hederin augmented the chemotherapeutic potential of cisplatin against ESTs; this effect was at least partly mediated through suppressing the chemokine SDF1/CXCR4/p-AKT/NFκB signaling. Further studies are recommended to verify the chemotherapeutic potential of α-hederin in other breast cancer models.
Collapse
|
21
|
Ramírez-Palma LG, Castro-Ramírez R, Lozano-Ramos L, Galindo-Murillo R, Barba-Behrens N, Cortés-Guzmán F. DNA recognition site of anticancer tinidazole copper(II) complexes. Dalton Trans 2023; 52:2087-2097. [PMID: 36692493 DOI: 10.1039/d2dt02854a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
This paper describes the recognition process of tetrahedral [CuII(tnz)2X2] (X = Cl, Br) complexes by a DNA chain, analyzing the specific interaction between the DNA bases and backbone with the metal and the tinidazole (tnz) ligand. We identified the coordination of the copper metal center with one or two phosphates as the first recognition site for the tinidazole copper(II) complexes, while the ligands present partial intercalation into the minor groove. Also, we discuss a novel trigonal copper(I) tnz bromide complex, obtained by reducing the previously reported [Cu(tnz)2Br2]. This complex sheds light on the mechanism of action of tnz metal complexes as one of the most stable DNA-complex adducts depicts a trigonal geometry around the copper ion.
Collapse
Affiliation(s)
- Lillian G Ramírez-Palma
- Instituto de Química, Universidad Nacional Autónoma de México, Av. Universidad 3000, C. U., México City, 04510, Mexico.
| | - Rodrigo Castro-Ramírez
- Facultad de Química, Universidad Nacional Autónoma de México, Av. Universidad 3000, C. U., México City, 04510, Mexico.
| | - León Lozano-Ramos
- Facultad de Química, Universidad Nacional Autónoma de México, Av. Universidad 3000, C. U., México City, 04510, Mexico.
| | - Rodrigo Galindo-Murillo
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, 2000 East 30 South Skaggs 201, Salt Lake City, UT 84112, USA
| | - Norah Barba-Behrens
- Facultad de Química, Universidad Nacional Autónoma de México, Av. Universidad 3000, C. U., México City, 04510, Mexico.
| | - Fernando Cortés-Guzmán
- Instituto de Química, Universidad Nacional Autónoma de México, Av. Universidad 3000, C. U., México City, 04510, Mexico.
| |
Collapse
|
22
|
Tao X, Li Y, Fan S, Wu L, Xin J, Su Y, Xian X, Huang Y, Huang R, Fang W, Liu Z. Downregulation of Linc00173 increases BCL2 mRNA stability via the miR-1275/PROCA1/ZFP36L2 axis and induces acquired cisplatin resistance of lung adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:12. [PMID: 36627670 PMCID: PMC9830831 DOI: 10.1186/s13046-022-02560-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 12/04/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND LINC00173 had been reported as a cisplatin (cis-diamminedichloroplatinum, DDP) chemotherapy-resistant inducer in small-cell lung cancer (SCLC) and lung squamous cell carcinoma (LUSC). This study aimed to display reverse data for LINC00173 as a DDP chemosensitivity-inducing factor in lung adenocarcinoma (LUAD). METHODS LINC00173 was screened from the Gene Expression Omnibus database (GSE43493). The expression level of LINC00173 in LUAD tissues and cell lines was detected using in situ hybridization and quantitative reverse transcription-polymerase chain reaction. Colony formation, cell viability, half-maximal inhibitory concentration, flow cytometry, and xenograft mouse model were used to evaluate the role of LINC00173 in the chemosensitivity of LUAD to DDP. The mechanism of LINC00173 in DDP resistance by mediating miR-1275/PROCA1/ZFP36L2 axis to impair BCL2 mRNA stability was applied, and co-immunoprecipitation, chromatin immunoprecipitation, RNA antisense purification, RNA immunoprecipitation, and luciferase reporter assays were performed. RESULTS LINC00173 downregulation in patients with DDP-resistant LUAD was correlated with poor prognosis. Further, LINC00173 expression was significantly reduced in DDP-resistant LUAD cells and DDP-treated human LUAD tissues. Suppressed LINC00173 expression in LUAD cells enhanced DDP chemoresistance in vivo and in vitro, while restored LINC00173 expression in DDP-resistant LUAD cells markedly regained chemosensitivity to DDP. Mechanistically, DDP-resistant LUAD cells activated PI3K/AKT signal and further elevated the c-Myc expression. The c-Myc, as an oncogenic transcriptional factor, bound to the promoter of LINC00173 and suppressed its expression. The reduced LINC00173 expression attenuated the adsorption of oncogenic miR-1275, downregulating the expression of miR-1275 target gene PROCA1. PROCA1 played a potential tumor-suppressive role inducing cell apoptosis and DDP chemosensitivity via recruiting ZFP36L2 to bind to the 3' untranslated region of BCL2, reducing the stability of BCL2 mRNA and thus activating the apoptotic signal. CONCLUSIONS This study demonstrated a novel and critical role of LINC00173. It was transcriptionally repressed by DDP-activated PI3K/AKT/c-Myc signal in LUAD, promoting DDP-acquired chemotherapeutic resistance by regulating miR-1275 to suppress PROCA1/ZFP36L2-induced BCL2 degradation, which led to apoptotic signal reduction. These data were not consistent with the previously described role of LINC00173 in SCLC or LUSC, which suggested that LINC00173 could play fine-tuned DDP resistance roles in different pathological subtypes of lung cancer. This study demonstrated that the diminished expression of LINC00173 might serve as an indicator of DDP-acquired resistance in LUAD.
Collapse
Affiliation(s)
- Xingyu Tao
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Yang Li
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Songqing Fan
- grid.452708.c0000 0004 1803 0208The Second Xiangya Hospital of Central South University, Changsha, 410008 China
| | - Liyang Wu
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Jianyang Xin
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Yun Su
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Xiaoyang Xian
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Yingying Huang
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Rongquan Huang
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Weiyi Fang
- grid.284723.80000 0000 8877 7471Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China ,grid.284723.80000 0000 8877 7471Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315 China
| | - Zhen Liu
- grid.284723.80000 0000 8877 7471Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315 China ,grid.410737.60000 0000 8653 1072Department of Pathology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| |
Collapse
|
23
|
STRESS granule-associated RNA-binding protein CAPRIN1 drives cancer progression and regulates treatment response in nasopharyngeal carcinoma. Med Oncol 2023; 40:47. [PMID: 36515758 PMCID: PMC9750908 DOI: 10.1007/s12032-022-01910-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignancy of the head and neck that is mainly diagnosed in southern China and Southeast Asia, with a strong etiological link to Epstein‒Barr virus infection. Those with advanced-stage disease have a significantly worse prognosis. There is an urgent need to identify novel therapeutic targets for the recurrent or metastatic nasopharyngeal carcinoma. With a particular focus on Cell Cycle Associated Protein 1 (CAPRIN1), one of the important RNA-binding proteints associated with stress granule formation, we used RT‒qPCR and immunohistochemistry to validate CAPRIN1 expression in NPC tissues and cell lines. Further, CAPRIN1 expression was knocked down using siRNA, and the effect on cell proliferation and migration was systematically assessed by in vitro assays. As a result, we demonstrated that CAPRIN1 was elevated in NPC compared to adjacent normal tissues. Knockdown of CAPRIN1 in NPC cells inhibited proliferation and migration, involving the regulation of cell cycle protein CCND2 and EMT signaling, respectively. Notably, we found that CAPRIN1 knockdown promoted cell apoptosis by regulation of the expression of apoptosis-related proteins cleaved-PARP and cleaved-Caspase3. Knockdown of CAPRIN1 increased NPC cell sensitivity to rapamycin, and increased NPC cell sensitivity to cisplatin and to X-rays. In conclusion, CAPRIN1 might drive NPC proliferation, regulate cell cycle and apoptosis, and affect tumor cell response to anti-cancer agents and X-ray irradiation. CAPRIN1 might serve as a potential target for NPC.
Collapse
|
24
|
Ahmedy OA, El-Tanbouly DM, Al-Mokaddem AK, El-Said YA. Insights into the role of P2X7R/DUSP6/ERK1/2 and SIRT2/MDM2 signaling in the nephroprotective effect of berberine against cisplatin-induced renal fibrosis in rats. Life Sci 2022; 309:121040. [DOI: 10.1016/j.lfs.2022.121040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022]
|
25
|
Bulathge AW, Villones RLE, Herbert FC, Gassensmith JJ, Meloni G. Comparative cisplatin reactivity towards human Zn7-metallothionein-2 and MTF-1 zinc fingers: potential implications in anticancer drug resistance. Metallomics 2022; 14:mfac061. [PMID: 36026541 PMCID: PMC9477119 DOI: 10.1093/mtomcs/mfac061] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/03/2022] [Indexed: 11/12/2022]
Abstract
Cis-diamminedichloroplatinum(II) (cisplatin) is a widely used metal-based chemotherapeutic drug for the treatment of cancers. However, intrinsic and acquired drug resistance limit the efficacy of cisplatin-based treatments. Increased production of intracellular thiol-rich molecules, in particular metallothioneins (MTs), which form stable coordination complexes with the electrophilic cisplatin, results in cisplatin sequestration leading to pre-target resistance. MT-1/-2 are overexpressed in cancer cells, and their expression is controlled by the metal response element (MRE)-binding transcription factor-1 (MTF-1), featuring six Cys2His2-type zinc fingers which, upon zinc metalation, recognize specific MRE sequences in the promoter region of MT genes triggering their expression. Cisplatin can efficiently react with protein metal binding sites featuring nucleophilic cysteine and/or histidine residues, including MTs and zinc fingers proteins, but the preferential reactivity towards specific targets with competing binding sites cannot be easily predicted. In this work, by in vitro competition reactions, we investigated the thermodynamic and kinetic preferential reactivity of cisplatin towards human Zn7MT-2, each of the six MTF-1 zinc fingers, and the entire human MTF-1 zinc finger domain. By spectroscopic, spectrometric, and electrophoretic mobility shift assays (EMSA), we demonstrated that cisplatin preferentially reacts with Zn7MT-2 to form Cys4-Pt(II) complexes, resulting in zinc release from MT-2. Zinc transfer from MT-2 to the MTF-1 triggers MTF-1 metalation, activation, and binding to target MRE sequences, as demonstrated by EMSA with DNA oligonucleotides. The cisplatin-dependent MT-mediated MTF-1 activation leading to apo-MT overexpression potentially establishes one of the molecular mechanisms underlying the development and potentiation of MT-mediated pre-target resistance.
Collapse
Affiliation(s)
- Anjala W Bulathge
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX-75080, USA
| | - Rhiza Lyne E Villones
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX-75080, USA
| | - Fabian C Herbert
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX-75080, USA
| | - Jeremiah J Gassensmith
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX-75080, USA
| | - Gabriele Meloni
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX-75080, USA
| |
Collapse
|
26
|
Bhattacharjee R, Dey T, Kumar L, Kar S, Sarkar R, Ghorai M, Malik S, Jha NK, Vellingiri B, Kesari KK, Pérez de la Lastra JM, Dey A. Cellular landscaping of cisplatin resistance in cervical cancer. Biomed Pharmacother 2022; 153:113345. [PMID: 35810692 DOI: 10.1016/j.biopha.2022.113345] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022] Open
Abstract
Cervical cancer (CC) caused by human papillomavirus (HPV) is one of the largest causes of malignancies in women worldwide. Cisplatin is one of the widely used drugs for the treatment of CC is rendered ineffective owing to drug resistance. This review highlights the cause of resistance and the mechanism of cisplatin resistance cells in CC to develop therapeutic ventures and strategies that could be utilized to overcome the aforementioned issue. These strategies would include the application of nanocarries, miRNA, CRIPSR/Cas system, and chemotherapeutics in synergy with cisplatin to not only overcome the issues of drug resistance but also enhance its anti-cancer efficiency. Moreover, we have also discussed the signaling network of cisplatin resistance cells in CC that would provide insights to develop therapeutic target sites and inhibitors. Furthermore, we have discussed the role of CC metabolism on cisplatin resistance cells and the physical and biological factors affecting the tumor microenvironments.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Tanima Dey
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, Kerala, India
| | - Sulagna Kar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Ritayan Sarkar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Mimosa Ghorai
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand 834001, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India.
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641-046, India
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo 00076, Finland; Department of Bio-products and Bio-systems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - José M Pérez de la Lastra
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), Avda. Astrofísico Francisco Sánchez, 3, 38206 San Cristóbal de la Laguna (Santa Cruz de Tenerife), Spain.
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|
27
|
Ahmed S, Jayathuna MA, Mahendiran D, Bharathi S, Kalilur Rahiman A. Heteroleptic silver(I), nickel(II), and copper(II) complexes of N
4
‐substituted thiosemicarbazones and ciprofloxacin: Theoretical, in vitro anti‐proliferative, and in silico molecular modeling and pharmacokinetics studies. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sumeer Ahmed
- Post‐Graduate and Research Department of Chemistry, The New College (Autonomous) University of Madras Chennai India
| | - Mugamathu Ali Jayathuna
- Post‐Graduate and Research Department of Chemistry, The New College (Autonomous) University of Madras Chennai India
| | - Dharmasivam Mahendiran
- Post‐Graduate and Research Department of Chemistry, The New College (Autonomous) University of Madras Chennai India
- Centre for Cancer Cell Biology and Drug Discovery Griffith Institute for Drug Discovery, Griffith University, Nathan Brisbane Queensland Australia
| | - Sundaram Bharathi
- Post‐Graduate and Research Department of Chemistry, The New College (Autonomous) University of Madras Chennai India
- Department of Chemistry, School of Basic Sciences Vels Institute of Science, Technology and Advanced Studies Chennai India
| | - Aziz Kalilur Rahiman
- Post‐Graduate and Research Department of Chemistry, The New College (Autonomous) University of Madras Chennai India
| |
Collapse
|
28
|
Namwan N, Senawong G, Phaosiri C, Kumboonma P, Somsakeesit LO, Samankul A, Leerat C, Senawong T. HDAC Inhibitory and Anti-Cancer Activities of Curcumin and Curcumin Derivative CU17 against Human Lung Cancer A549 Cells. Molecules 2022; 27:4014. [PMID: 35807258 PMCID: PMC9268269 DOI: 10.3390/molecules27134014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Previous research reported that the curcumin derivative (CU17) inhibited several cancer cell growths in vitro. However, its anticancer potential against human lung cancer cells (A549 cell lines) has not yet been evaluated. The purpose of this research was to examine the HDAC inhibitory and anti-cancer activities of CU17 compared to curcumin (CU) in A549 cells. An in vitro study showed that CU17 had greater HDAC inhibitory activity than CU. CU17 inhibited HDAC activity in a dose dependent manner with the half-maximal inhibitory concentration (IC50) value of 0.30 ± 0.086 µg/mL against HDAC enzymes from HeLa nuclear extract. In addition, CU17 could bind at the active pockets of both human class I HDACs (HDAC1, 2, 3, and 8) and class II HDACs (HDAC4, 6, and 7) demonstrated by molecular docking studies, and caused hyperacetylation of histone H3 (Ac-H3) in A549 cells shown by Western blot analysis. MTT assay indicated that both CU and CU17 suppressed A549 cell growth in a dose- and time-dependent manner. Besides, CU and CU17 induced G2/M phase cell cycle arrest and p53-independent apoptosis in A549 cells. Both CU and CU17 down-regulated the expression of p53, p21, Bcl-2, and pERK1/2, but up-regulated Bax expression in this cell line. Although CU17 inhibited the growth of lung cancer cells less effectively than CU, it showed less toxicity than CU for non-cancer cells. Accordingly, CU17 is a promising agent for lung cancer treatment. Additionally, CU17 synergized the antiproliferative activity of Gem in A549 cells, indicating the possibility of employing CU17 as an adjuvant treatment to enhance the chemotherapeutic effect of Gem in lung cancer.
Collapse
Affiliation(s)
- Narissara Namwan
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; (N.N.); (G.S.); (A.S.); (C.L.)
| | - Gulsiri Senawong
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; (N.N.); (G.S.); (A.S.); (C.L.)
| | - Chanokbhorn Phaosiri
- Department of Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Pakit Kumboonma
- Department of Applied Chemistry, Faculty of Science and Liberal Arts, Rajamangala University of Technology Isan, Nakhon Ratchasima 30000, Thailand;
| | - La-or Somsakeesit
- Department of Chemistry, Faculty of Engineering, Rajamangala University of Technology Isan, Khon Kaen 40000, Thailand;
| | - Arunta Samankul
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; (N.N.); (G.S.); (A.S.); (C.L.)
| | - Chadaporn Leerat
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; (N.N.); (G.S.); (A.S.); (C.L.)
| | - Thanaset Senawong
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; (N.N.); (G.S.); (A.S.); (C.L.)
| |
Collapse
|
29
|
Kaku Y, Chiba K, Sato K, Onishi A, Ishida T, Okada K, Fujisawa M. Protective effects of tadalafil against cisplatin-induced spermatogenic dysfunction. Biochem Biophys Res Commun 2022; 603:123-129. [DOI: 10.1016/j.bbrc.2022.02.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 12/14/2022]
|
30
|
de Oliveira RM, Moura TA, Rocha MS. Caffeine Enhances the Toxicity of Platinum-Based Drugs at the Molecular Level Even Outside of the Intracellular Environment: A Single-Molecule Force Spectroscopy Study. J Phys Chem B 2022; 126:3291-3299. [PMID: 35442688 DOI: 10.1021/acs.jpcb.2c01553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
It is well reported in the literature that caffeine, the most consumed alkaloid around the world, enhances the anticancer effects of the drug cisplatin by inhibiting DNA repair by the cellular machinery. Here, we perform single-molecule force spectroscopy assays with optical tweezers to show that caffeine enhances the toxicity not only of cisplatin but also of various different platinum-based drugs already at the molecular level, using samples containing only double-stranded (ds)DNA, platinum drugs, and the alkaloid in a simple phosphate buffer, that is, completely out of the complex environment found inside real living cells. In fact, our results show that caffeine acts as an allosteric catalyst which increases the effective equilibrium binding constant between DNA and the platinum drugs, also interfering in the cooperativity of the binding reactions. To the best of our knowledge, this is the first time that such a property of caffeine was demonstrated and characterized from a pure physicochemical perspective, outside the cellular environment. Thus, the present work provides new insights into the use of this alkaloid for current chemotherapeutic applications.
Collapse
Affiliation(s)
- Rayane M de Oliveira
- Departamento de Física, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Tiago A Moura
- Departamento de Física, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Márcio S Rocha
- Departamento de Física, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| |
Collapse
|
31
|
Jones TM, Espitia CM, Ooi A, Bauman JE, Carew JS, Nawrocki ST. Targeted CUL4A inhibition synergizes with cisplatin to yield long-term survival in models of head and neck squamous cell carcinoma through a DDB2-mediated mechanism. Cell Death Dis 2022; 13:350. [PMID: 35428778 PMCID: PMC9012827 DOI: 10.1038/s41419-022-04798-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/24/2022]
Abstract
Patients with late-stage and human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC) continue to have a very poor prognosis. The development of more effective novel therapies that improve overall survival and overcome drug resistance is an urgent priority. Here we report that HNSCC tumors significantly overexpress NEDD8 and exhibit high sensitivity to the first-in-class NEDD8-activating enzyme (NAE) inhibitor pevonedistat. Additional studies established that disruption of NEDD8-mediated protein turnover with pevonedistat dramatically augmented cisplatin-induced DNA damage and apoptosis in HNSCC models. Further analysis revealed that the specific pevonedistat target CUL4A played an essential role in driving the synergy of the pevonedistat and cisplatin combination. Targeted inhibition of CUL4A resulted in significant downregulation in Damage Specific DNA binding protein 2 (DDB2), a DNA-damage recognition protein that promotes nucleotide excision repair and resistance to cisplatin. Silencing of CUL4A or DDB2 enhanced cisplatin-induced DNA damage and apoptosis in a manner similar to that of pevonedistat demonstrating that targeted inhibition of CUL4A may be a novel approach to augment cisplatin therapy. Administration of pevonedistat to mice bearing HNSCC tumors significantly decreased DDB2 expression in tumor cells, increased DNA damage and potently enhanced the activity of cisplatin to yield tumor regression and long-term survival of all animals. Our findings provide strong rationale for clinical investigation of CUL4A inhibition with pevonedistat as a novel strategy to augment the efficacy of cisplatin therapy for patients with HNSCC and identify loss of DDB2 as a key pharmacodynamic mediator controlling sensitivity to this regimen.
Collapse
Affiliation(s)
- Trace M Jones
- University of Arizona Cancer Center, Tucson, AZ, USA
| | | | - Aikseng Ooi
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | | | | | | |
Collapse
|
32
|
Jiang M, Yang T, Chu Y, Zhang Z, Sun H, Liang H, Yang F. Design of a novel Pt(II) complex to reverse cisplatin-induced resistance in lung cancer via a multi-mechanism. Dalton Trans 2022; 51:5257-5270. [PMID: 35285843 DOI: 10.1039/d1dt03964d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In order to develop a novel platinum (Pt) complex aiming to overcome cisplatin resistance, we synthesised a series of novel Pt complexes (C1-C6). These Pt complexes displayed potent cytotoxicity activity against resistant lung cancer cells (A549cisR) in vitro and efficiently inhibited tumour growth in vivo. The Pt complexes can target DNA, lead to DNA platination and cause cell cycle arrest in the S phase, thus impeding precise DNA synthesis. C6, in particular, induced not only apoptosis but also lethal autophagy in A549cisR cells. In addition, these novel Pt complexes reversed cisplatin-induced resistance via inhibiting the expression of P-glycoprotein and decreasing the level of glutathione in A549cisR cells. Moreover, the ERK pathway was involved in C6-induced overcoming cisplatin resistance.
Collapse
Affiliation(s)
- Ming Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China. .,School of food and biochemical engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Tongfu Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Yong Chu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| |
Collapse
|
33
|
Chandra A, Datta A. A Peptide-Based Fluorescent Sensor for Anionic Phospholipids. ACS OMEGA 2022; 7:10347-10354. [PMID: 35382295 PMCID: PMC8973094 DOI: 10.1021/acsomega.1c06981] [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: 12/10/2021] [Accepted: 02/25/2022] [Indexed: 06/14/2023]
Abstract
Anionic phospholipids are key cell signal mediators. The distribution of these lipids on the cell membrane and intracellular organelle membranes guides the recruitment of signaling proteins leading to the regulation of cellular processes. Hence, fluorescent sensors that can detect anionic phospholipids within living cells can provide a handle into revealing molecular mechanisms underlying lipid-mediated signal regulation. A major challenge in the detection of anionic phospholipids is related to the presence of these phospholipids mostly in the inner leaflet of the plasma membrane and in the membranes of intracellular organelles. Hence, cell-permeable sensors would provide an advantage by enabling the rapid detection and tracking of intracellular pools of anionic phospholipids. We have developed a peptide-based, cell-permeable, water-soluble, and ratiometric fluorescent sensor that entered cells within 15 min of incubation via the endosomal machinery and showed punctate labeling in the cytoplasm. The probe could also be introduced into living cells via lipofection, which allows bypassing of endosomal uptake, to image anionic phospholipids in the cell membrane. We validated the ability of the sensor toward detection of intracellular anionic phospholipids by colocalization studies with a fluorescently tagged lipid and a protein-based anionic phospholipid sensor. Further, the sensor could image the externalization of anionic phospholipids during programmed cell death, indicating the ability of the probe toward detection of both intra- and extracellular anionic phospholipids based on the biological context.
Collapse
|
34
|
Feng T, Karges J, Liao X, Ji L, Chao H. Engineered exosomes as a natural nanoplatform for cancer targeted delivery of metal-based drugs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Sharma A, Sinha S, Shrivastava N. Apigenin and kaempferol as novel renoprotective agent against cisplatin-induced toxicity: an in vitro study. Nat Prod Res 2022; 36:6085-6090. [PMID: 35227143 DOI: 10.1080/14786419.2022.2045603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cisplatin is one of the highly consumed and potent antineoplastic drugs. However, its side effects in normal tissues, notably nephrotoxicity, is a major stumbling block and dose-limiting factor. Renoprotective approaches are being developed, however, the protective benefits are usually only partial implying the need for combinatorial strategies. Therefore, in this study, we investigated the nephroprotective efficacy of apigenin and kaempferol as dietary supplements against cisplatin-induced renal injury using human embryonic kidney (HEK-293) cells as our in vitro model. Our findings from MTT data, morphology studies, comet and ROS analysis suggest that CIS 11.36 µM + API 12.5 µg/mL and CIS 11.36 µM + KMP 25 µg/mL protects against cisplatin-induced nephrotoxicity. Results of western blot analysis further suggest the involvement of NGAL in the API and KMP mediated nephroprotection. Collectively, our studies suggest that API and KMP are promising candidates to be further developed as renoprotective agents against cisplatin-induced toxicity.
Collapse
Affiliation(s)
- Abhilasha Sharma
- B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India.,Department of Life science, Gujarat University, Ahmedabad, Gujarat, India
| | - Sonam Sinha
- B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India.,Kashiv Biosciences, Ahmedabad, Gujarat, India
| | - Neeta Shrivastava
- B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India.,Shri B.V. Patel Education Trust, Ahmedabad, Gujarat, India
| |
Collapse
|
36
|
Wan L, Zhang W, Liu Z, Yang Z, Tu C, Li Z. A Novel Glutamine Metabolism-Related Gene Signature in Prognostic Prediction of Osteosarcoma. Int J Gen Med 2022; 15:997-1011. [PMID: 35136353 PMCID: PMC8817953 DOI: 10.2147/ijgm.s352859] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/21/2022] [Indexed: 12/18/2022] Open
Affiliation(s)
- Lu Wan
- Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Wenchao Zhang
- Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhongyue Liu
- Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhimin Yang
- Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Chao Tu
- Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Correspondence: Chao Tu; Zhihong Li, Tel +86 15974290117; +86 13975112458, Email ;
| | - Zhihong Li
- Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
37
|
Su J, Chen S, Dou Y, Zhao Z, Jia X, Ding X, Song S. Smartphone-Based Electrochemical Biosensors for Directly Detecting Serum-Derived Exosomes and Monitoring Their Secretion. Anal Chem 2022; 94:3235-3244. [PMID: 35084842 DOI: 10.1021/acs.analchem.1c04910] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exosomes are potential biomarkers, which play an important role in early diagnosis and prognosis prediction of cancer-related diseases. Nevertheless, direct quantification of exosomes in biological fluid, especially in point-of-care tests (POCTs), remains extremely challenging. Herein, we developed a sensitive and portable electrochemical biosensor in combination with smartphones for quantitative analysis of exosomes. The improved double-antibody sandwich method-based poly-enzyme signal amplification was adopted to detect exosomes. We could detect as low as 7.23 ng of CD63-positive exosomes in 5 μL of serum within 2 h. Importantly, we demonstrated that the biosensor worked well with microliter-level serum and cell culture supernatant. The biosensor holds great potential for the detection of CD-63-expressing exosomes in early diagnosis of prostate disease because CD63-positive exosomes were less detected from the prostate patient serum. Also, the biosensor was used to monitor the secretion of exosomes with the drug therapy, showing a close relationship between the secretion of exosomes and the concentration of cisplatin. The biosensing platform provides a novel way toward POCT for the diagnosis and prognosis prediction of prostate disease and other diseases via biomarker expression levels of exosomes.
Collapse
Affiliation(s)
- Jing Su
- Center for Research and Interdisciplinary, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shixing Chen
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Yanzhi Dou
- University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Zhihan Zhao
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Xiaolong Jia
- Department of Urology, Ningbo First Hospital Ningbo, Hospital of Zhejiang University, 17 Ningbo, Ningbo 315010, Zhejiang Province, China
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shiping Song
- Center for Research and Interdisciplinary, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| |
Collapse
|
38
|
Wang W, Yang C, Deng H. Overexpression of 15-Hydroxyprostaglandin Dehydrogenase Inhibits A549 Lung Adenocarcinoma Cell Growth via Inducing Cell Cycle Arrest and Inhibiting Epithelial-Mesenchymal Transition. Cancer Manag Res 2021; 13:8887-8900. [PMID: 34876851 PMCID: PMC8643138 DOI: 10.2147/cmar.s331222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/20/2021] [Indexed: 01/22/2023] Open
Abstract
Purpose Lung cancer is one of the most commonly diagnosed cancer as well as the leading cause of cancer-related mortality worldwide, among which lung adenocarcinoma (LUAD) is the most frequent form of lung cancer. Previous studies have shown that 15-hydroxyprostaglandin dehydrogenase (15-PGDH) catalyzes the oxidation of prostaglandins to reduce their biological activities and behaves as a tumor suppressor in various cancers. Thus, we aimed to systematically examine the effects of 15-PGDH overexpression on cellular processes in lung adenocarcinoma cells. Methods The stable 15-PGDH-overexpressing A549 cell line was constructed using lentivirus particles. CCK-8 assay was used to determine the cell proliferation rate and sensitivity to cisplatin. Tandem mass tag (TMT)-based quantitative proteomic analysis was used to identify differentially expressed proteins between control and 15-PGDH-overexpression cells. The cell cycle was determined by a flow cytometer. The expression levels of mesenchymal and epithelial markers were measured using Western blotting. Wound healing and transwell assays were used to detect the cell migration and cell invasion ability, respectively. Results Analysis of datasets in The Cancer Genome Atlas revealed that the PGDH gene expression level in the lung adenocarcinoma tissues was significantly lower than that in the pericarcinous tissues. 15-PGDH overexpression in A549 cells reduced cell proliferation rate. Quantitative proteomics revealed that 15-PGDH overexpression inhibited PI3K/AKT/mTOR signaling pathway, which is a signaling pathway driving tumor cell growth and epithelial-mesenchymal transition (EMT) process. In addition, both cell cycle and DNA repair-related proteins were down-regulated in 15-PGDH overexpressed cells. 15-PGDH overexpression induced G1/S cell cycle arrest and increased susceptibility to DNA damaging reagent cisplatin. Importantly, overexpression of 15-PGDH inhibited EMT process with the downregulation of β-catenin and Snail-1 as well as upregulation of E-cadherin and ZO-1. Conclusion 15-PGDH is a tumor suppressor in lung cancer and may serve as a potential therapeutic target to prevent lung adenocarcinoma.
Collapse
Affiliation(s)
- Weixuan Wang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Changmei Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| |
Collapse
|
39
|
The complex build algorithm to set up starting structures of lanthanoid complexes with stereochemical control for molecular modeling. Sci Rep 2021; 11:21493. [PMID: 34728757 PMCID: PMC8564551 DOI: 10.1038/s41598-021-99525-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/27/2021] [Indexed: 01/20/2023] Open
Abstract
When handling metallic centers of higher coordination numbers, one is commonly deluded with the presumption that any assembled metal complex geometry (including a crystallographic one) is good enough as a starting structure for computational chemistry calculations; all oblivious to the fact that such a structure is nothing short of just one out of several, sometimes dozens, or even thousands of other stereoisomers. Moreover, coordination chirality, so frequently present in complexes of higher coordination numbers, is another often overlooked property, rarely recognized as such. The Complex Build algorithm advanced in this article has been designed with the purpose of generating starting structures for molecular modeling calculations with full stereochemical control, including stereoisomer complete identification and coordination chirality recognition. Besides being in the chosen correct stereochemistry, the ligands are positioned by the Complex Build algorithm in a very unobstructed and unclogged manner, so that their degrees of freedom do not hinder or even choke one another, something that would otherwise tend to lead to negative force constants after further geometry optimizations by more advanced computational model chemistries. The Complex Build algorithm has been conceived for any metallic center, but at present is targeting primarily lanthanoids whose coordination numbers range mostly from 5 to 12 and often lead to a combinatorial explosion of stereoisomers.
Collapse
|
40
|
Chen G, Zeng H, Li X, Liu J, Li Z, Xu R, Ma Y, Liu C, Xue B. Activation of G protein coupled estrogen receptor prevents chemotherapy-induced intestinal mucositis by inhibiting the DNA damage in crypt cell in an extracellular signal-regulated kinase 1- and 2- dependent manner. Cell Death Dis 2021; 12:1034. [PMID: 34718327 PMCID: PMC8557214 DOI: 10.1038/s41419-021-04325-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a common adverse reaction to antineoplastic treatment with few appropriate, specific interventions. We aimed to identify the role of the G protein coupled estrogen receptor (GPER) in CIM and its mechanism. Adult male C57BL/6 mice were intraperitoneally injected with 5-fluorouracil to establish the CIM model. The selective GPER agonist G-1 significantly inhibited weight loss and histological damage in CIM mice and restored mucosal barrier dysfunction, including improving the expression of ZO-1, increasing the number of goblet cells, and decreasing mucosal permeability. Moreover, G-1 treatment did not alter the antitumor effect of 5-fluorouracil. In the CIM model, G-1 therapy reduced the expression of proapoptotic protein and cyclin D1 and cyclin B1, reversed the changes in the number of TUNEL+ cells, Ki67+ and bromodeoxyuridine+ cells in crypts. The selective GPER antagonist G15 eliminated all of the above effects caused by G-1 on CIM, and application of G15 alone increased the severity of CIM. GPER was predominantly expressed in ileal crypts, and G-1 inhibited the DNA damage induced by 5-fluorouracil in vivo and vitro, as confirmed by the decrease in the number of γH2AX+ cells in the crypts and the comet assay results. Referring to the data from GEO dataset we verified GPER activation restored ERK1/2 activity in CIM and 5-fluorouracil-treated IEC-6 cells. Once the effects of G-1 on ERK1/2 activity were abolished with the ERK1/2 inhibitor PD0325901, the effects of G-1 on DNA damage both in vivo and in vitro were eliminated. Correspondingly, all of the manifestations of G-1 protection against CIM were inhibited by PD0325901, such as body weight and histological changes, the mucosal barrier, the apoptosis and proliferation of crypt cells. In conclusion, GPER activation prevents CIM by inhibiting crypt cell DNA damage in an ERK1/2-dependent manner, suggesting GPER might be a target preventing CIM.
Collapse
Affiliation(s)
- Guanyu Chen
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Honghui Zeng
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinyun Li
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Jianbo Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhao Li
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Runze Xu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuntao Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chuanyong Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Xue
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
41
|
Lee SH, Choi SJ, Choi W, Cho S, Cho M, Kim DS, Kang BW, Kim JG, Lee YM, Cho H, Kang H. Cisplatin Resistance in Epstein-Barr-Virus-Associated Gastric Carcinoma Acquired through ATM Methylation. Cancers (Basel) 2021; 13:cancers13174252. [PMID: 34503060 PMCID: PMC8428228 DOI: 10.3390/cancers13174252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/20/2021] [Accepted: 08/22/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Gastric cancer (GC) is the fifth-leading type of cancer and the third –leading cause of death from cancer. Epstein-Barr virus-associated gastric carcinoma (EBVaGC) is recently accountable for 10% of all the GC worldwide. Platinum drugs such as cisplatin and oxaliplatin are the first-line choice in GC chemotherapy. The widespread use of cisplatin leads to make tumor cells develop single or multiple drug resistance via various mechanisms. DNA hypermethylation on tumor suppressor genes is one of causes leading to drug resistances. 5-Azacytidine (5-AZA) is a chemical analogue of cytidine and inhibits DNA methyltransferase, resulting in DNA hypomethylation. Our main objective was to identify synergistic effect of two important GC drugs whose mechanisms may be in complementary cooperation. We found that cisplatin enhances its anticancer activity with 5-AZA through DNA demethylation in EBVaGC. Identifying this synergistic effect of two important GC drugs can be useful to treat EBVaGC which shows resistance to platinum-based chemotherapy. Abstract Epstein–Barr-virus-associated gastric carcinoma (EBVaGC), first reported in 1992, currently accounts for 10% of all gastric carcinoma worldwide. EBVaGC has unique DNA hypermethylation phenotypes that allow for higher proportions of DNA methylation than any other gastric cancer. CpG islands in the gene promoter region are one of the major regions in which DNA methylation controls gene transcription. Despite cisplatin-based chemotherapy being one of the standard treatment regimens for advanced gastric cancer, including EBVaGC, cisplatin alone or in combination with 5-fluorouracil has been limited by its less potent anticancer activity and the occurrence of cisplatin resistance. Accordingly, the current study evaluated the anticancer activities of a combination of cisplatin and 5-Azacytidine (5-AZA) against EBVaGC. Our findings showed that cisplatin upregulated the DNMT3A gene, whereas shRNA-targeted removal of DNMT3A mRNA contributed to cisplatin-mediated EBV lytic reactivation. Moreover, the removal of DNMT3A mRNA upregulated the ATM gene through DNA demethylation on the ATM promoter. Furthermore, CRISPR/Cas9-targeted removal of the ATM gene resulted in significantly reduced cell susceptibility and EBV lytic reactivation by a combination of cisplatin and DNMT3A inhibitor 5-AZA. Finally, 5-AZA exhibited a synergistic effect with cisplatin in anti-EBV and anti-EBVaGC activities by increasing drug susceptibility and EBV lytic reactivation. The aforementioned results suggest that cisplatin combined with DNA methylation inhibitors could be a novel therapeutic approach for EBVaGC.
Collapse
Affiliation(s)
- Sun Hee Lee
- Vessel-Organ Interaction Research Center, VOICE (MRC), Cancer Research Institute, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.H.L.); (S.J.C.); (S.C.); (M.C.)
| | - Su Jin Choi
- Vessel-Organ Interaction Research Center, VOICE (MRC), Cancer Research Institute, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.H.L.); (S.J.C.); (S.C.); (M.C.)
| | - Wonhyeok Choi
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University, Seoul 01369, Korea;
| | - Subin Cho
- Vessel-Organ Interaction Research Center, VOICE (MRC), Cancer Research Institute, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.H.L.); (S.J.C.); (S.C.); (M.C.)
| | - Miyeon Cho
- Vessel-Organ Interaction Research Center, VOICE (MRC), Cancer Research Institute, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.H.L.); (S.J.C.); (S.C.); (M.C.)
| | - Dong Sun Kim
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Byung Woog Kang
- Department of Oncology/Hematology, Cancer Research Institute, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41405, Korea; (B.W.K.); (J.G.K.)
| | - Jong Gwang Kim
- Department of Oncology/Hematology, Cancer Research Institute, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41405, Korea; (B.W.K.); (J.G.K.)
| | - You Mie Lee
- Vessel-Organ Interaction Research Center, VOICE (MRC), Department of Molecular Pathophysiology, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea;
| | - Hyosun Cho
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University, Seoul 01369, Korea;
- Correspondence: (H.C.); (H.K.); Tel.: +82-02-901-8678 (H.C.); +82-053-950-8569 (H.K.); Fax: +82-02-901-8386 (H.C.); +82-053-950-8557 (H.K.)
| | - Hyojeung Kang
- Vessel-Organ Interaction Research Center, VOICE (MRC), Cancer Research Institute, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.H.L.); (S.J.C.); (S.C.); (M.C.)
- Correspondence: (H.C.); (H.K.); Tel.: +82-02-901-8678 (H.C.); +82-053-950-8569 (H.K.); Fax: +82-02-901-8386 (H.C.); +82-053-950-8557 (H.K.)
| |
Collapse
|
42
|
McPherson KS, Korzhnev DM. Targeting protein-protein interactions in the DNA damage response pathways for cancer chemotherapy. RSC Chem Biol 2021; 2:1167-1195. [PMID: 34458830 PMCID: PMC8342002 DOI: 10.1039/d1cb00101a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/20/2021] [Indexed: 12/11/2022] Open
Abstract
Cellular DNA damage response (DDR) is an extensive signaling network that orchestrates DNA damage recognition, repair and avoidance, cell cycle progression and cell death. DDR alteration is a hallmark of cancer, with the deficiency in one DDR capability often compensated by a dependency on alternative pathways endowing cancer cells with survival and growth advantage. Targeting these DDR pathways has provided multiple opportunities for the development of cancer therapies. Traditional drug discovery has mainly focused on catalytic inhibitors that block enzyme active sites, which limits the number of potential drug targets within the DDR pathways. This review article describes the emerging approach to the development of cancer therapeutics targeting essential protein-protein interactions (PPIs) in the DDR network. The overall strategy for the structure-based design of small molecule PPI inhibitors is discussed, followed by an overview of the major DNA damage sensing, DNA repair, and DNA damage tolerance pathways with a specific focus on PPI targets for anti-cancer drug design. The existing small molecule inhibitors of DDR PPIs are summarized that selectively kill cancer cells and/or sensitize cancers to front-line genotoxic therapies, and a range of new PPI targets are proposed that may lead to the development of novel chemotherapeutics.
Collapse
Affiliation(s)
- Kerry Silva McPherson
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center Farmington CT 06030 USA +1 860 679 3408 +1 860 679 2849
| | - Dmitry M Korzhnev
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center Farmington CT 06030 USA +1 860 679 3408 +1 860 679 2849
| |
Collapse
|
43
|
Muruzabal D, Sanz-Serrano J, Sauvaigo S, Treillard B, Olsen AK, López de Cerain A, Vettorazzi A, Azqueta A. Validation of the in vitro comet assay for DNA cross-links and altered bases detection. Arch Toxicol 2021; 95:2825-2838. [PMID: 34196753 PMCID: PMC8298235 DOI: 10.1007/s00204-021-03102-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/15/2021] [Indexed: 11/27/2022]
Abstract
Mechanistic toxicology is gaining weight for human health risk assessment. Different mechanistic assays are available, such as the comet assay, which detects DNA damage at the level of individual cells. However, the conventional alkaline version only detects strand breaks and alkali-labile sites. We have validated two modifications of the in vitro assay to generate mechanistic information: (1) use of DNA-repair enzymes (i.e., formamidopyrimidine DNA glycosylase, endonuclease III, human 8-oxoguanine DNA glycosylase I and human alkyladenine DNA glycosylase) for detection of oxidized and alkylated bases as well as (2) a modification for detecting cross-links. Seven genotoxicants with different mechanisms of action (potassium bromate, methyl methanesulfonate, ethyl methanesulfonate, hydrogen peroxide, cisplatin, mitomycin C, and benzo[a]pyrene diol epoxide), as well as a non-genotoxic compound (dimethyl sulfoxide) and a cytotoxic compound (Triton X-100) were tested on TK-6 cells. We were able to detect with high sensitivity and clearly differentiate oxidizing, alkylating and cross-linking agents. These modifications of the comet assay significantly increase its sensitivity and its specificity towards DNA lesions, providing mechanistic information regarding the type of damage.
Collapse
Affiliation(s)
- Damián Muruzabal
- Department of Pharmacology and Toxicology, University of Navarra, C/Irunlarrea 1, 31009, Pamplona, Spain
| | - Julen Sanz-Serrano
- Department of Pharmacology and Toxicology, University of Navarra, C/Irunlarrea 1, 31009, Pamplona, Spain
| | - Sylvie Sauvaigo
- LXRepair, Biopolis, 5 Avenue du Grand Sablon, 38700, La Tronche, France
| | | | - Ann-Karin Olsen
- Section of Molecular Toxicology, Department of Environmental Health, Norwegian Institute of Public Health, Skøyen, PO Box 222, 0213, Oslo, Norway
| | - Adela López de Cerain
- Department of Pharmacology and Toxicology, University of Navarra, C/Irunlarrea 1, 31009, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Ariane Vettorazzi
- Department of Pharmacology and Toxicology, University of Navarra, C/Irunlarrea 1, 31009, Pamplona, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, University of Navarra, C/Irunlarrea 1, 31009, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
44
|
Lu P, Zhang C, Fu L, Wei Y, Huang Y, Wang X, Lv C, Chen L. Near-Infrared Fluorescent Probe for Imaging and Evaluating the Role of Vanin-1 in Chemotherapy. Anal Chem 2021; 93:10378-10387. [PMID: 34275284 DOI: 10.1021/acs.analchem.1c02386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Pantetheinase (also known as Vanin-1) is highly expressed in the liver, kidneys, and intestine and is closely associated with a number of diseases. Vanin-1 can hydrolyze pantetheine to pantothenic acid (vitamin B5) and cysteamine and participate in the synthesis of glutathione (GSH). GSH is highly expressed in tumor cells and plays a major role in the resistance of tumor cells to cisplatin. Therefore, we urgently need a method to monitor the activity level of Vanin-1 in tumor cells and tissues and elucidate the relationship between the role of Vanin-1 in GSH synthesis and tumor resistance. Herein, we report a Cy-Pa fluorescent probe for imaging Vanin-1 in cells and in vivo that can qualitatively and quantitatively detect the fluctuation of Vanin-1 concentrations in HepG2 and HepG2/DDP cells or tumor tissues of tumor-bearing mice. This probe shows excellent potential in in situ real-time monitoring of endogenous Vanin-1. Moreover, we proved that Vanin-1 can inhibit GSH synthesis using the probe. When the Vanin-1 inhibitor RR6 was used in combination with cisplatin, HepG2 and HepG2/DDP cells showed increased resistance to cisplatin, while the therapeutic efficiency of cisplatin was reduced in HepG2 and HepG2/DDP xenografts. In this study, Vanin-1 was shown to play an important role in the treatment of cancer, and the study of Vanin-1 may provide an idea for the treatment of cancer in the future.
Collapse
Affiliation(s)
- Pengpeng Lu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.,CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Caiyun Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Lili Fu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.,CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Yinghui Wei
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.,Department of Respiratory Medicine, Binzhou Medical University Hospital, Binzhou 256603, China
| | - Yan Huang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.,CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Xiaoyan Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.,CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Changjun Lv
- Department of Respiratory Medicine, Binzhou Medical University Hospital, Binzhou 256603, China
| | - Lingxin Chen
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.,CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.,College of Chemistry and Chemical Engineering, Qufu Normal University, University, Qufu 273165, China
| |
Collapse
|
45
|
Sun Y, Liu W, Zhao Q, Zhang R, Wang J, Pan P, Shang H, Liu C, Wang C. Down-Regulating the Expression of miRNA-21 Inhibits the Glucose Metabolism of A549/DDP Cells and Promotes Cell Death Through the PI3K/AKT/mTOR/HIF-1α Pathway. Front Oncol 2021; 11:653596. [PMID: 34046349 PMCID: PMC8144645 DOI: 10.3389/fonc.2021.653596] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/12/2021] [Indexed: 12/28/2022] Open
Abstract
miRNA-21 is a single-stranded non-coding RNA that is highly expressed in a variety of tumor cells. It participates in tumor cell proliferation, metabolism, metastasis, and drug resistance. Here, we tested the potential mechanism of miRNA-21 in cisplatin-resistant non-small cell lung cancer A549/DDP (human lung adenocarcinoma drug-resistant cell line) cells. A549 and A549/DDP RNAs were sequenced to show that miRNA-21 was highly expressed in the latter, and this was verified by qRT-PCR. In addition, we found that miRNA-21 combined with cisplatin can significantly inhibit glycolysis and glycolysis rate-limiting enzyme protein expression in A549/DDP cells. We also found that miRNA-21 combined with cisplatin can promote A549/DDP cell death. Further investigations showed that miRNA-21 combined with cisplatin caused excessive inactivation of the pI3K/AKT/mTOR/HIF-1α signaling pathway in cisplatin-resistant A549/DDP cells. Hence, reduction of the expression of miRNA-21 in combination with cisplatin chemotherapy may effectively improve the therapeutic effect on patients with non-small cell lung cancer, and this may provide a theoretical basis for the treatment of this disease.
Collapse
Affiliation(s)
- Ye Sun
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Shenyang, China
| | - Wenjun Liu
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Qiuyu Zhao
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine (TCM) Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | | | - Jianbo Wang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine (TCM) Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Pengyu Pan
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Hai Shang
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Chunying Liu
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Chun Wang
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
46
|
HIF in Nephrotoxicity during Cisplatin Chemotherapy: Regulation, Function and Therapeutic Potential. Cancers (Basel) 2021; 13:cancers13020180. [PMID: 33430279 PMCID: PMC7825709 DOI: 10.3390/cancers13020180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/27/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Cisplatin is a widely used chemotherapy drug, but its use and efficacy are limited by its nephrotoxicity. HIF has protective effects against kidney injury during cisplatin chemotherapy, but it may attenuate the anti-cancer effect of cisplatin. In this review, we describe the role and regulation of HIF in cisplatin-induced nephrotoxicity and highlight the therapeutic potential of targeting HIF in chemotherapy. Abstract Cisplatin is a highly effective, broad-spectrum chemotherapeutic drug, yet its clinical use and efficacy are limited by its side effects. Particularly, cancer patients receiving cisplatin chemotherapy have high incidence of kidney problems. Hypoxia-inducible factor (HIF) is the “master” transcription factor that is induced under hypoxia to trans-activate various genes for adaptation to the low oxygen condition. Numerous studies have reported that HIF activation protects against AKI and promotes kidney recovery in experimental models of cisplatin-induced acute kidney injury (AKI). In contrast, little is known about the effects of HIF on chronic kidney problems following cisplatin chemotherapy. Prolyl hydroxylase (PHD) inhibitors are potent HIF inducers that recently entered clinical use. By inducing HIF, PHD inhibitors may protect kidneys during cisplatin chemotherapy. However, HIF activation by PHD inhibitors may reduce the anti-cancer effect of cisplatin in tumors. Future studies should test PHD inhibitors in tumor-bearing animal models to verify their effects in kidneys and tumors.
Collapse
|
47
|
Deng Z, Wang N, Ai F, Wang Z, Zhu G. Nanomaterial‐mediated platinum drug‐based combinatorial cancer therapy. VIEW 2020. [DOI: 10.1002/viw.20200030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Zhiqin Deng
- Department of Chemistry City University of Hong Kong Hong Kong SAR P. R. China
- Shenzhen Research Institute City University of Hong Kong Shenzhen P. R. China
| | - Na Wang
- Department of Chemistry City University of Hong Kong Hong Kong SAR P. R. China
- Shenzhen Research Institute City University of Hong Kong Shenzhen P. R. China
| | - Fujin Ai
- College of Health Science and Environment Engineering Shenzhen Technology University Shenzhen P. R. China
| | - Zhigang Wang
- School of Pharmaceutical Sciences Health Science Center Shenzhen University Shenzhen P. R. China
| | - Guangyu Zhu
- Department of Chemistry City University of Hong Kong Hong Kong SAR P. R. China
- Shenzhen Research Institute City University of Hong Kong Shenzhen P. R. China
| |
Collapse
|
48
|
Yin H, Gao J, Chen X, Ma B, Yang Z, Tang J, Wang B, Chen T, Wang C, Gao S, Zhang J. A Gallium(III) Complex that Engages Protein Disulfide Isomerase A3 (PDIA3) as an Anticancer Target. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Hao‐Yan Yin
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Jiu‐Jiao Gao
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Xuemin Chen
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Bin Ma
- Department of Chemistry Jinan University Guangzhou 510632 P. R. China
| | - Zi‐Shu Yang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Juan Tang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Bing‐Wu Wang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Tianfeng Chen
- Department of Chemistry Jinan University Guangzhou 510632 P. R. China
| | - Chu Wang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Song Gao
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
- School of Chemistry and Chemical Engineering South China University of Technology Guangzhou 510640 P. R. China
| | - Jun‐Long Zhang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| |
Collapse
|
49
|
Facile synthesis of heterobimetallic [FeII(µ-diphosphine)RuII] and homobimetallic [FeII(µ-diphosphine)FeII] complexes and their in vitro cytotoxic activity on cisplatin-resistant cancer cells. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Yin H, Gao J, Chen X, Ma B, Yang Z, Tang J, Wang B, Chen T, Wang C, Gao S, Zhang J. A Gallium(III) Complex that Engages Protein Disulfide Isomerase A3 (PDIA3) as an Anticancer Target. Angew Chem Int Ed Engl 2020; 59:20147-20153. [DOI: 10.1002/anie.202008432] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/23/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Hao‐Yan Yin
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Jiu‐Jiao Gao
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Xuemin Chen
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Bin Ma
- Department of Chemistry Jinan University Guangzhou 510632 P. R. China
| | - Zi‐Shu Yang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Juan Tang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Bing‐Wu Wang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Tianfeng Chen
- Department of Chemistry Jinan University Guangzhou 510632 P. R. China
| | - Chu Wang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| | - Song Gao
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
- School of Chemistry and Chemical Engineering South China University of Technology Guangzhou 510640 P. R. China
| | - Jun‐Long Zhang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Rare Earth Materials Chemistry and Applications College of Chemistry and Molecular Engineering Peking University Beijing 100871 P. R. China
| |
Collapse
|