1
|
Qi K, Li J, Hu Y, Qiao Y, Mu Y. Research progress in mechanism of anticancer action of shikonin targeting reactive oxygen species. Front Pharmacol 2024; 15:1416781. [PMID: 39076592 PMCID: PMC11284502 DOI: 10.3389/fphar.2024.1416781] [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: 04/13/2024] [Accepted: 06/13/2024] [Indexed: 07/31/2024] Open
Abstract
Excessive buildup of highly reactive molecules can occur due to the generation and dysregulation of reactive oxygen species (ROS) and their associated signaling pathways. ROS have a dual function in cancer development, either leading to DNA mutations that promote the growth and dissemination of cancer cells, or triggering the death of cancer cells. Cancer cells strategically balance their fate by modulating ROS levels, activating pro-cancer signaling pathways, and suppressing antioxidant defenses. Consequently, targeting ROS has emerged as a promising strategy in cancer therapy. Shikonin and its derivatives, along with related drug carriers, can impact several signaling pathways by targeting components involved with oxidative stress to induce processes such as apoptosis, necroptosis, cell cycle arrest, autophagy, as well as modulation of ferroptosis. Moreover, they can increase the responsiveness of drug-resistant cells to chemotherapy drugs, based on the specific characteristics of ROS, as well as the kind and stage of cancer. This research explores the pro-cancer and anti-cancer impacts of ROS, summarize the mechanisms and research achievements of shikonin-targeted ROS in anti-cancer effects and provide suggestions for designing further anti-tumor experiments and undertaking further experimental and practical research.
Collapse
Affiliation(s)
- Ke Qi
- Department of Diagnostic Clinical Laboratory Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jiayi Li
- Department of Clinical Test Center, Medical Laboratory, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yang Hu
- Department of Diagnostic Clinical Laboratory Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yiyun Qiao
- Department of Clinical Test Center, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yongping Mu
- Department of Clinical Test Center, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Huang Z, Zhou L, Duan J, Qin S, Jiang J, Chen H, Wang K, Liu R, Yuan M, Tang X, Nice EC, Wei Y, Zhang W, Huang C. Oxidative Stress Promotes Liver Cancer Metastasis via RNF25-Mediated E-Cadherin Protein Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306929. [PMID: 38286671 PMCID: PMC10987140 DOI: 10.1002/advs.202306929] [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: 09/21/2023] [Revised: 12/25/2023] [Indexed: 01/31/2024]
Abstract
Loss of E-cadherin (ECAD) is required in tumor metastasis. Protein degradation of ECAD in response to oxidative stress is found in metastasis of hepatocellular carcinoma (HCC) and is independent of transcriptional repression as usually known. Mechanistically, protein kinase A (PKA) senses oxidative stress by redox modification in its β catalytic subunit (PRKACB) at Cys200 and Cys344. The activation of PKA kinase activity subsequently induces RNF25 phosphorylation at Ser450 to initiate RNF25-catalyzed degradation of ECAD. Functionally, RNF25 repression induces ECAD protein expression and inhibits HCC metastasis in vitro and in vivo. Altogether, these results indicate that RNF25 is a critical regulator of ECAD protein turnover, and PKA is a necessary redox sensor to enable this process. This study provides some mechanistic insight into how oxidative stress-induced ECAD degradation promotes tumor metastasis of HCC.
Collapse
Affiliation(s)
- Zhao Huang
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Jiufei Duan
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Siyuan Qin
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth HospitalSichuan UniversityChengdu610041China
| | - Haining Chen
- Colorectal Cancer CenterDepartment of General SurgeryWest China HospitalSichuan UniversityChengdu610041China
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic MedicineState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Rui Liu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesResearch Unit of Oral Carcinogenesis and ManagementChinese Academy of Medical SciencesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Minlan Yuan
- Mental Health Center and Psychiatric LaboratoryThe State Key Laboratory of BiotherapyWest China Biomedical Big Data CenterWest China Hospital of Sichuan UniversityChengdu610041China
| | - Xiangdong Tang
- Sleep Medicine CenterDepartment of Respiratory and Critical Care MedicineMental Health CenterTranslational Neuroscience CenterState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVIC3167Australia
| | - Yuquan Wei
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Wei Zhang
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengdu610212China
- Medical Big Data CenterSichuan UniversityChengdu610041China
| | - Canhua Huang
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengdu610212China
| |
Collapse
|
3
|
Li K, Deng Z, Lei C, Ding X, Li J, Wang C. The Role of Oxidative Stress in Tumorigenesis and Progression. Cells 2024; 13:441. [PMID: 38474405 DOI: 10.3390/cells13050441] [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: 01/11/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Oxidative stress refers to the imbalance between the production of reactive oxygen species (ROS) and the endogenous antioxidant defense system. Its involvement in cell senescence, apoptosis, and series diseases has been demonstrated. Advances in carcinogenic research have revealed oxidative stress as a pivotal pathophysiological pathway in tumorigenesis and to be involved in lung cancer, glioma, hepatocellular carcinoma, leukemia, and so on. This review combs the effects of oxidative stress on tumorigenesis on each phase and cell fate determination, and three features are discussed. Oxidative stress takes part in the processes ranging from tumorigenesis to tumor death via series pathways and processes like mitochondrial stress, endoplasmic reticulum stress, and ferroptosis. It can affect cell fate by engaging in the complex relationships between senescence, death, and cancer. The influence of oxidative stress on tumorigenesis and progression is a multi-stage interlaced process that includes two aspects of promotion and inhibition, with mitochondria as the core of regulation. A deeper and more comprehensive understanding of the effects of oxidative stress on tumorigenesis is conducive to exploring more tumor therapies.
Collapse
Affiliation(s)
- Kexin Li
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Zhangyuzi Deng
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Chunran Lei
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Xiaoqing Ding
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Jing Li
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Changshan Wang
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| |
Collapse
|
4
|
Yang B, Yao L, Yang L, Zhao F, Zhou W. Inhibition of miR-578 through SOCS2-dependent manner reverses gefitinib resistance in NSCLC cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:1283-1293. [PMID: 37948135 DOI: 10.1002/tox.24004] [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: 04/22/2023] [Revised: 08/30/2023] [Accepted: 10/07/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Nonsmall-cell lung cancer (NSCLC) has emerged as one of the dreadful lung cancers globally due to its increased mortality rates. Concerning chemotherapy, gefitinib has been employed as an effective first-line treatment drug for NSCLC. Nonetheless, the acquired resistance to gefitinib has remained one of the treatment obstacles of NSCLC, requiring improvement in the therapeutic effect of gefitinib. METHODS Initially, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and Western blotting (WB) analyses were conducted to measure micro-ribose nucleic acid (miRNA, specifically miR-578) and suppressor of cytokine signaling 2 (SOCS2) levels in the clinical samples. Further, NSCLC cell lines resistance to gefitinib, established in vitro, were transfected by miR-578 inhibitor, miR-578 mimic, and si-SOCS2. Similarly, the xenograft mouse model in vivo was constructed to validate the reversing effect of miR-578. RESULTS Our findings indicated the increased miR-578 expression levels in the gefitinib resistance group. Further, inhibiting the miR-578 expression substantially reversed the gefitinib resistance. In addition, the miR-578 effect was modulated via the SOCS2 expression level. The decreased gefitinib resistance effect of miR-578 was weakened by inhibiting the SOCS2 expression. CONCLUSION These findings demonstrated that miR-578 effectively abolished gefitinib resistance by regulating the SOCS2 expression within NSCLC cells in vitro and in vivo. Together, these results will undoubtedly support a reference to provide potential molecular therapeutic targets and clinical treatments for treating NSCLC patients.
Collapse
Affiliation(s)
- Bo Yang
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Lei Yao
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Liankai Yang
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Fang Zhao
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Wenyan Zhou
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| |
Collapse
|
5
|
Twum Y, Marshall K, Gao W. Caffeic acid phenethyl ester surmounts acquired resistance of AZD9291 in non-small cell lung cancer cells. Biofactors 2023; 49:1143-1157. [PMID: 37555475 DOI: 10.1002/biof.1983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/19/2023] [Indexed: 08/10/2023]
Abstract
Epithelial growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are the first-line therapy for EGFR mutated non-small cell lung cancer (NSCLC); however, resistance rapidly develops. The objective of this study was therefore to establish and characterize a gefitinib resistant NSCLC line (HCC827GR) and evaluate the therapeutic effects of natural products in combination with third-generation EGFR-TKI, AZD9291. The IC50 of gefitinib and AZD9291 in HCC827GR were significantly higher than those of HCC827 (p < 0.05). Furthermore, anchorage-independent colony assay indicated that HCC827GR cells were more aggressive than their predecessors. This was reflected by the gene/protein expression changes observed in HCC827GR versus HCC827 profiled by cancer drug resistance real-time polymerase chain reaction (RT-PCR) array and Western blot. Three natural products were screened and caffeic acid phenethyl ester (CAPE) exhibited the most significant combinative cytotoxic effect with AZD9291. Specifically, flow cytometry revealed that AZD9291 + CAPE considerably increased the fraction of cell in pre-G1 of the cell cycle and caspase-Glo3/7 assay showed a dramatic increase in apoptosis when compared to AZD9291 alone. Furthermore, Western blot showed significant downregulation of p-EGFR/p-AKT in HCC827GR cells treated with AZD9291 + CAPE as compared to AZD9291. Moreover, it is evident that AZD9291 + CAPE specifically resulted in a marked reduction in the protein expressions of the cell-proliferation-related genes p21, cyclin D1, and survivin. Finally, refined RT-PCR/Western blot data indicated that AZD9291 + CAPE may at least partially exert its synergistic effects via the PLK2 pathway. Together, these results suggest that CAPE is a clinically relevant compound to aid AZD9291 in treating EGFR-TKI resistant cells through modulating critical genes/proteins involved in cancer resistance/therapy.
Collapse
Affiliation(s)
- Yaw Twum
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, West Virginia, USA
| | - Kent Marshall
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, West Virginia, USA
| | - Weimin Gao
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
6
|
Koda Y, Nagasaki Y. Newly Designed Cysteine-Based Self-Assembling Prodrugs for Sepsis Treatment. Pharmaceutics 2023; 15:1775. [PMID: 37376222 DOI: 10.3390/pharmaceutics15061775] [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: 04/27/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Reactive oxygen species (ROS) are essential signaling molecules that maintain intracellular redox balance; however, the overproduction of ROS often causes dysfunction in redox homeostasis and induces serious diseases. Antioxidants are crucial candidates for reducing overproduced ROS; however, most antioxidants are less effective than anticipated. Therefore, we designed new polymer-based antioxidants based on the natural amino acid, cysteine (Cys). Amphiphilic block copolymers, composed of a hydrophilic poly(ethylene glycol) (PEG) segment and a hydrophobic poly(cysteine) (PCys) segment, were synthesized. In the PCys segment, the free thiol groups in the side chain were protected by thioester moiety. The obtained block copolymers formed self-assembling nanoparticles (NanoCys(Bu)) in water, and the hydrodynamic diameter was 40-160 nm, as determined by dynamic light scattering (DLS) measurements. NanoCys(Bu) was stable from pH 2 to 8 under aqueous conditions, as confirmed by the hydrodynamic diameter of NanoCys(Bu). Finally, NanoCys(Bu) was applied to sepsis treatment to investigate the potential of NanoCys(Bu). NanoCys(Bu) was supplied to BALB/cA mice by free drinking for two days, and lipopolysaccharide (LPS) was intraperitoneally injected into the mice to prepare a sepsis shock model (LPS = 5 mg per kg body weight (BW)). Compared with the Cys and no-treatment groups, NanoCys(Bu) prolonged the half-life by five to six hours. NanoCys(Bu), designed in this study, shows promise as a candidate for enhancing antioxidative efficacy and mitigating the adverse effect of cysteine.
Collapse
Affiliation(s)
- Yuta Koda
- Department of Materials Science, Faculty of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Faculty of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Japan
- School of Medical Sciences, School of Comprehensive Human Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Japan
- Center for Research in Radiation, Isotope and Earth System Sciences (CRiES), University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| |
Collapse
|
7
|
N-Acetyl-L-Cysteine Protects Airway Epithelial Cells during Respiratory Syncytial Virus Infection against Mucin Synthesis, Oxidative Stress, and Inflammatory Response and Inhibits HSPA6 Expression. Anal Cell Pathol 2022; 2022:4846336. [PMID: 36046596 PMCID: PMC9420614 DOI: 10.1155/2022/4846336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/19/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Objective. Respiratory syncytial virus (RSV) infection is an important cause of hospitalization of children worldwide, leading to significant morbidity and mortality. RSV infection leads to increasing inflammatory and apoptosis events in the airway epithelium through mechanisms involving ROS generation. The antioxidant N-acetyl-L-cysteine (NAC) has been shown to inhibit influenza virus replication and to reduce the secretion of inflammatory and apoptotic mediators during virus infection. The study aims to investigate the effects of NAC on human bronchial epithelial cells BEAS-2B and HSPA6 expression during RSV infection. Methods. CCK-8 assays were performed to evaluate cell survival. The production of proinflammatory factors, TNF-α, IL-6, IL-1β, IL-18, and MUC5AC was examined by quantitative real-time PCR and ELISA. Oxidative stress was determined by reactive oxygen species (ROS), superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione (GSH)/glutathione disulfide (GSSG) ratio. Immunoblotting analysis of epidermal growth factor receptor (EGFR) and its phosphorylation was performed. The antiviral effect of NAC was assessed by determining viral titers using plaque assay. Results. RSV infection reduced cell survival, promoted the release of proinflammatory factors, increased the ROS production and MDA concentration, and diminished the SOD activity and GSH/GSSG ratio, all which were attenuated by NAC treatment. Accordingly, NAC treatment inhibited the activation of EGFR and MUC5AC in BEAS-2B cells with RSV infection. Furthermore, NAC administration resulted in a marked decrease in RSV-induced HSPA6 expression in BEAS-2B cells. Concomitantly, EPB treatment led to an evident inhibition of RSV fusion gene and viral replication in RSV-infected BEAS-2B cells. Conclusion. This work supports the use of NAC to exert antimucin synthesis, anti-inflammatory, antioxidant, and antiviral effects on airway epithelium during RSV infection.
Collapse
|
8
|
Abstract
Eukaryotic cells have developed complex systems to regulate the production and response to reactive oxygen species (ROS). Different ROS control diverse aspects of cell behaviour from signalling to death, and deregulation of ROS production and ROS limitation pathways are common features of cancer cells. ROS also function to modulate the tumour environment, affecting the various stromal cells that provide metabolic support, a blood supply and immune responses to the tumour. Although it is clear that ROS play important roles during tumorigenesis, it has been difficult to reliably predict the effect of ROS modulating therapies. We now understand that the responses to ROS are highly complex and dependent on multiple factors, including the types, levels, localization and persistence of ROS, as well as the origin, environment and stage of the tumours themselves. This increasing understanding of the complexity of ROS in malignancies will be key to unlocking the potential of ROS-targeting therapies for cancer treatment.
Collapse
|
9
|
Cao L, Tong Y, Wang X, Zhang Q, Qi Y, Zhou C, Yu X, Wu Y, Miao X. Effect of Amniotic Membrane/Collagen-Based Scaffolds on the Chondrogenic Differentiation of Adipose-Derived Stem Cells and Cartilage Repair. Front Cell Dev Biol 2021; 9:647166. [PMID: 34900977 PMCID: PMC8657407 DOI: 10.3389/fcell.2021.647166] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/20/2021] [Indexed: 11/24/2022] Open
Abstract
Objectives: Repairing articular cartilage damage is challenging. Clinically, tissue engineering technology is used to induce stem cell differentiation and proliferation on biological scaffolds to repair defective joints. However, no ideal biological scaffolds have been identified. This study investigated the effects of amniotic membrane/collagen scaffolds on the differentiation of adipose-derived stem cells (ADSCs) and articular cartilage repair. Methods: Adipose tissue of New Zealand rabbits was excised, and ADSCs were isolated and induced for differentiation. An articular cartilage defect model was constructed to identify the effect of amniotic membrane/collagen scaffolds on cartilage repair. Cartilage formation was analyzed by imaging and toluene blue staining. Knee joint recovery in rabbits was examined using hematoxylin and eosin, toluidine, safranine, and immunohistochemistry at 12 weeks post-operation. Gene expression was examined using ELISA, RT-PCR, Western blotting, and immunofluorescence. Results: The adipose tissue was effectively differentiated into ADSCs, which further differentiated into chondrogenic, osteogenic, and lipogenic lineages after 3 weeks’ culture in vitro. Compared with platelet-rich plasmon (PRP) scaffolds, the amniotic membrane scaffolds better promoted the growth and differentiation of ADSCs. Additionally, scaffolds containing the PRP and amniotic membrane efficiently enhanced the osteogenic differentiation of ADSCs. The levels of COL1A1, COL2A1, COL10A1, SOX9, and ACAN in ADSCs + amniotic membrane + PRP group were significantly higher than the other groups both in vitro and in vivo. The Wakitani scores of the ADSC + amniotic membrane + PRP group were lower than that in ADSC + PRP (4.4 ± 0.44**), ADSC + amniotic membrane (2.63 ± 0.38**), and control groups (6.733 ± 0.21) at week 12 post-operation. Osteogenesis in rabbits of the ADSC + amniotic membrane + PRP group was significantly upregulated when compared with other groups. Amniotic membranes significantly promoted the expression of cartilage regeneration-related factors (SOX6, SOX9, RUNX2, NKX3-2, MEF2C, and GATA4). The ADSC + PRP + amniotic membrane group exhibited the highest levels of TGF-β, PDGF, and FGF while exhibiting the lowest level of IL-1β, IL6, and TNF-α in articular cavity. Conclusion: Amniotic membrane/collagen combination-based scaffolds promoted the proliferation and cartilage differentiation of ADSCs, and may provide a new treatment paradigm for patients with cartilage injury.
Collapse
Affiliation(s)
- Le Cao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yuling Tong
- Department of General Practice, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Wang
- Shaoxing Shangyu Hospital of Traditional Chinese medicine, Shaoxing, China
| | - Qiang Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yiying Qi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Chenhe Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Xinning Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yongping Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Xudong Miao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| |
Collapse
|
10
|
Xu C, Zhang L, Wang D, Jiang S, Cao D, Zhao Z, Huang M, Jin J. Lipidomics reveals that sustained SREBP-1-dependent lipogenesis is a key mediator of gefitinib-acquired resistance in EGFR-mutant lung cancer. Cell Death Discov 2021; 7:353. [PMID: 34775471 PMCID: PMC8590692 DOI: 10.1038/s41420-021-00744-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/17/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022] Open
Abstract
Patients with EGFR mutations in non-small cell lung cancer (NSCLC) have been greatly benefited from gefitinib, however, the therapeutic has failed due to the presence of acquired resistance. In this study, we show that gefitinib significantly induces downregulation of Sterol Regulator Element Binding (SREBP1) in therapy-sensitive cells. However, this was not observed in EGFR mutant NSCLC cells with acquired resistance. Lipidomics analysis showed that gefitinib could differently change the proportion of saturated phospholipids and unsaturated phospholipids in gefitinib-sensitive and acquired-resistant cells. Besides, levels of ROS and MDA were increased upon SREBP1 inhibition and even more upon gefitinib treatment. Importantly, inhibition of SREBP1 sensitizes EGFR-mutant therapy-resistant NSCLC to gefitinib both in vitro and in vivo models. These data suggest that sustained de novo lipogenesis through the maintenance of active SRBEP-1 is a key feature of acquired resistance to gefitinib in EGFR mutant lung cancer. Taken together, targeting SREBP1-induced lipogenesis is a promising approach to overcome acquired resistance to gefitinib in EGFR-mutant lung cancer.
Collapse
Affiliation(s)
- Chuncao Xu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lei Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Daifei Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shiqin Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Di Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhongxiang Zhao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Jin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
11
|
Combined Treatment of Cinobufotalin and Gefitinib Exhibits Potent Efficacy against Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6612365. [PMID: 34122599 PMCID: PMC8189783 DOI: 10.1155/2021/6612365] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 01/22/2023]
Abstract
This study aimed to evaluate the efficacy of cinobufotalin combined with gefitinib in the treatment of lung cancer. A549 cells were treated with gefitinib, cinobufotalin, or cinobufotalin plus gefitinib. MTT assay, annexin-V/PI staining and flow cytometry, TUNEL staining, DCFH-DA staining, Western blot, and real-time RT-PCR were performed to investigate the synergistic inhibitory effect of cinobufotalin combined with gefitinib on the growth of A549 cells. Results showed that cinobufotalin synergized with gefitinib displayed inhibited cell viability and enhanced apoptosis in the combination group. Cinobufotalin combined with gefitinib induced a significant enhancement in reactive oxygen species (ROS) production accompanied by cell cycle arrest in the S phase arrest, characterized by upregulation of p21 and downregulation of cyclin A, cyclin E, and CDK2. Besides, cinobufotalin plus gefitinib downregulated the levels of HGF and c-Met. In summary, cinobufotalin combined with gefitinib impedes viability and facilitates apoptosis of A549 cells, indicating that the combined therapy might be a new promising treatment for lung cancer patients who are resistant to gefitinib.
Collapse
|
12
|
Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative Stress in Cancer. Cancer Cell 2020; 38:167-197. [PMID: 32649885 DOI: 10.1016/jxcell.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 05/28/2023]
Abstract
Contingent upon concentration, reactive oxygen species (ROS) influence cancer evolution in apparently contradictory ways, either initiating/stimulating tumorigenesis and supporting transformation/proliferation of cancer cells or causing cell death. To accommodate high ROS levels, tumor cells modify sulfur-based metabolism, NADPH generation, and the activity of antioxidant transcription factors. During initiation, genetic changes enable cell survival under high ROS levels by activating antioxidant transcription factors or increasing NADPH via the pentose phosphate pathway (PPP). During progression and metastasis, tumor cells adapt to oxidative stress by increasing NADPH in various ways, including activation of AMPK, the PPP, and reductive glutamine and folate metabolism.
Collapse
Affiliation(s)
- John D Hayes
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland.
| | - Albena T Dinkova-Kostova
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
13
|
Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative Stress in Cancer. Cancer Cell 2020; 38:167-197. [PMID: 32649885 PMCID: PMC7439808 DOI: 10.1016/j.ccell.2020.06.001] [Citation(s) in RCA: 1241] [Impact Index Per Article: 310.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Contingent upon concentration, reactive oxygen species (ROS) influence cancer evolution in apparently contradictory ways, either initiating/stimulating tumorigenesis and supporting transformation/proliferation of cancer cells or causing cell death. To accommodate high ROS levels, tumor cells modify sulfur-based metabolism, NADPH generation, and the activity of antioxidant transcription factors. During initiation, genetic changes enable cell survival under high ROS levels by activating antioxidant transcription factors or increasing NADPH via the pentose phosphate pathway (PPP). During progression and metastasis, tumor cells adapt to oxidative stress by increasing NADPH in various ways, including activation of AMPK, the PPP, and reductive glutamine and folate metabolism.
Collapse
Affiliation(s)
- John D Hayes
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland.
| | - Albena T Dinkova-Kostova
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
14
|
Luo X, Zhou J, Quan L, Liang Y, Huang P, Chen F, Liu S. LncRNA NCK1-AS1 promotes non-small cell lung cancer progression via regulating miR-512-5p/p21 axis. Pathol Res Pract 2020; 216:153157. [PMID: 32858372 DOI: 10.1016/j.prp.2020.153157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 01/22/2023]
Abstract
OBJECTIVE This study aimed at probing into the effect of lncRNA NCK1-AS1 on proliferation, migration and invasion of non-small cell lung cancer (NSCLC) cells and its regulatory function on miR-512-5p/p21 molecular axis. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was used to assess the expressions of NCK1-AS1 and miR-512-5p in NSCLC tissues and cell lines. The alterations of cell proliferation, migration, invasion and cell cycle were examined by cell counting kit-8 (CCK-8) assay, BrdU experiment, Transwell experiment and flow cytometry, respectively. The dual-luciferase reporter assay and RNA immunoprecipitation experiment were performed to validate the binding relationships between miR-512-5p and NCK1-AS1, and miR-512-5p the 3'UTR of p21 mRNA. Western blot was used to determine the effects of NCK1-AS1 and miR-512-5p on p21 protein expression. RESULTS NCK1-AS1 expression was up-regulated in NSCLC tissues and cells, and its high expression was correlated with shorter overall survival time and faster progression of patients. Overexpression of NCK1-AS1 promoted NSCLC cell proliferation, migration and invasion, and accelerated the cell cycle, whereas NCK1-AS1 siRNA inhibited these malignant biological behaviors, and arrested cell cycle. NCK1-AS1 could bind to miR-512-5p, p21 was verified as a target gene of miR-512-5p, and NCK1-AS1 could up-regulate the expression of p21 in NSCLC cells via repressing miR-512-5p expression. CONCLUSION NCK1-AS1 promotes NSCLC progression by regulating miR-512-5p/p21 molecular axis.
Collapse
Affiliation(s)
- Xiping Luo
- Pulmonary and Critical Care Medicine 1, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412000, China
| | - Juan Zhou
- Pulmonary and Critical Care Medicine 1, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412000, China
| | - Lingli Quan
- Pulmonary and Critical Care Medicine 1, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412000, China.
| | - Yanchao Liang
- Pulmonary and Critical Care Medicine 1, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412000, China.
| | - Peng Huang
- Pulmonary and Critical Care Medicine 1, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412000, China
| | - Fangwei Chen
- Pulmonary and Critical Care Medicine 1, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412000, China
| | - Shuangbo Liu
- Pulmonary and Critical Care Medicine 1, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412000, China
| |
Collapse
|
15
|
Fu J, Wang X, Yue Q. Functional loss of TAGLN inhibits tumor growth and increases chemosensitivity of non-small cell lung cancer. Biochem Biophys Res Commun 2020; 529:1086-1093. [PMID: 32819569 DOI: 10.1016/j.bbrc.2020.06.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/14/2020] [Indexed: 01/18/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of tumor mortality worldwide. However, the mechanisms underlying NSCLC tumorigenesis are incompletely understood. TAGLN, also named SM22, as a member of the calponin family, is highly expressed in many types of tumors. Nevertheless, its effects on NSCLC progression remain unclear. In this study, we found that TAGLN was over-expressed in tumor tissues of NSCLC patients and cell lines. Additionally, NSCLC patients with high expression showed worse overall survival rate. Then, gene silencing results indicated that TAGLN knockdown markedly inhibited proliferation and induced apoptosis in NSCLC cells, while rescue study exhibited opposite results. Moreover, suppressing TAGLN significantly reduced migration and invasion of NSCLC cells, and its over-expression promoted the migratory and invasive activities of NSCLC cells. The in vivo studies confirmed the oncogenic roles of TAGLN in NSCLC, along with clearly elevated metastasis. Notably, these effects were abrogated in mice with TAGLN deletion. Finally, we found that TAGLN knockdown could improve the sensitivity of NSCLC cells to sorafenib (SFB) and 5-FU treatment, further suppressing the proliferation, migration and invasion of NSCLC cells. Consistently, TAGLN deletion attenuated tumor xenografts growth and metastasis of NSCLC in mouse models by enhancing the anti-cancer effects of SFB and 5-FU. Altogether, these findings demonstrated that TAGLN functioned as an oncogene as well as a chemotherapeutic regulator during NSCLC development, which suggested a potential therapeutic strategy for NSCLC treatment mainly through repressing TAGLN expression.
Collapse
Affiliation(s)
- Juanjuan Fu
- Department of Pathology, The Fifth People's Hospital of Wuxi, Wuxi City, 214013, China
| | - Xiaoguang Wang
- Department of Pulmonary Medicine, Chinese People's Liberation Army 92493 Military Hospital, Huludao City, Liaoning Province, 125000, China
| | - Qingfang Yue
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| |
Collapse
|
16
|
Li J, Wang XH, Hu J, Shi M, Zhang L, Chen H. Combined treatment with N-acetylcysteine and gefitinib overcomes drug resistance to gefitinib in NSCLC cell line. Cancer Med 2019; 9:1495-1502. [PMID: 31891230 PMCID: PMC7013061 DOI: 10.1002/cam4.2610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 09/01/2019] [Accepted: 09/13/2019] [Indexed: 12/24/2022] Open
Abstract
We aimed to explore the molecular substrate underlying EGFR‐TKI resistance and investigate the effects of N‐acetylcysteine (NAC) on reversing EGFR‐TKI resistance. In the current research, the effects of NAC in combination with gefitinib on reversing gefitinib resistance were examined using CCK‐8 assay, combination index (CI) method, matrigel invasion assay, wound‐healing assay, flow cytometry, western blot, and quantitative real‐time PCR (qRT‐PCR). CCK8 assay showed that NAC plus gefitinib combination overcame EGFR‐TKI resistance in non‐small cell lung cancer (NSCLC) cells by lowering the value of half maximal inhibitory concentration (IC50). CI calculations demonstrated a synergistic effect between the two drugs (CI < 1). Matrigel invasion assay and wound healing assay demonstrated a decrease in migration and invasion ability of PC‐9/GR cells after NAC and gefitinib treatment. Flow cytometry displayed enhanced apoptosis in the combination group. Western blot and qRT‐PCR revealed that increased E‐cadherin and decreased vimentin in the combination group. When PP2 was administered with gefitinib, the same effects were seen. Our findings suggest that NAC could restore the sensitivity of gefitinib‐resistant NSCLC cells to gefitinib via suppressing Src activation and reversing epithelial‐mesenchymal transition.
Collapse
Affiliation(s)
- Jun Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Hui Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Hu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meng Shi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|