151
|
Wang C, Li F, Zhang T, Yu M, Sun Y. Recent advances in anti-multidrug resistance for nano-drug delivery system. Drug Deliv 2022; 29:1684-1697. [PMID: 35616278 PMCID: PMC9154776 DOI: 10.1080/10717544.2022.2079771] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy for tumors occasionally results in drug resistance, which is the major reason for the treatment failure. Higher drug doses could improve the therapeutic effect, but higher toxicity limits the further treatment. For overcoming drug resistance, functional nano-drug delivery system (NDDS) has been explored to sensitize the anticancer drugs and decrease its side effects, which are applied in combating multidrug resistance (MDR) via a variety of mechanisms including bypassing drug efflux, controlling drug release, and disturbing metabolism. This review starts with a brief report on the major MDR causes. Furthermore, we searched the papers from NDDS and introduced the recent advances in sensitizing the chemotherapeutic drugs against MDR tumors. Finally, we concluded that the NDDS was based on several mechanisms, and we looked forward to the future in this field.
Collapse
Affiliation(s)
- Changduo Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Fashun Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Tianao Zhang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Min Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| |
Collapse
|
152
|
Huang Y, Yuan C, Liu Q, Wang L. KIF23 promotes autophagy-induced imatinib resistance in chronic myeloid leukaemia through activating Wnt/β-catenin pathway. Clin Exp Pharmacol Physiol 2022; 49:1334-1341. [PMID: 36066385 DOI: 10.1111/1440-1681.13718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/10/2022] [Accepted: 09/01/2022] [Indexed: 01/31/2023]
Abstract
Imatinib, an inhibitor of tyrosine kinase, shows remarkable efficacy in chronic myeloid leukaemia (CML). Autophagy protects tumour cells against chemotherapeutic stimulation and contributes to imatinib resistance in CML. Kinesin family member 23 (KIF23) is involved in cytokinesis and associated with autophagy. The role of KIF23 in autophagy-induced imatinib resistance in CML was investigated. First, to induce drug resistance, CML cells were exposed to increasing concentrations of imatinib. The concentration of imatinib resistance in CML cells was screened through upregulation of 50% inhibitory concentration (IC50 ) values. KIF23 was elevated in imatinib-resistant tissues and cells of CML. Second, knockdown of KIF23 reduced IC50 values of imatinib-resistant CML cells to imatinib. Moreover, silence of KIF23 also suppressed cell proliferation and promoted apoptosis of imatinib-resistant CML cells. Third, immunofluorescence analysis showed that the number of LC3 bright spots in imatinib-resistant CML cells was reduced by silence of KIF23. Knockdown of KIF23 upregulated p62 expression and downregulated the expression ratio of LC3-II to LC3-I in imatinib-resistant CML cells. Last, silence of KIF23 decreased nuclear β-catenin and increased cytoplasmic β-catenin in imatinib-resistant CML cells. Activator of Wnt/β-catenin attenuated KIF23 silence-induced increase of apoptosis and decrease of autophagy in imatinib-resistant CML cells. In conclusion, loss of KIF23 repressed autophagy-induced imatinib resistance in CML cells through inactivation of Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yong Huang
- Department of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunyan Yuan
- Department of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiwei Liu
- Department of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lingying Wang
- Department of A Dietary, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
153
|
Niemann B, Puleo A, Stout C, Markel J, Boone BA. Biologic Functions of Hydroxychloroquine in Disease: From COVID-19 to Cancer. Pharmaceutics 2022; 14:pharmaceutics14122551. [PMID: 36559044 PMCID: PMC9787624 DOI: 10.3390/pharmaceutics14122551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
Abstract
Chloroquine (CQ) and Hydroxychloroquine (HCQ), initially utilized in the treatment of malaria, have now developed a long list of applications. Despite their clinical relevance, their mechanisms of action are not clearly defined. Major pathways by which these agents are proposed to function include alkalinization of lysosomes and endosomes, downregulation of C-X-C chemokine receptor type 4 (CXCR4) expression, high-mobility group box 1 protein (HMGB1) inhibition, alteration of intracellular calcium, and prevention of thrombus formation. However, there is conflicting data present in the literature. This is likely the result of the complex overlapping pathways between these mechanisms of action that have not previously been highlighted. In fact, prior research has focused on very specific portions of particular pathways without describing these in the context of the extensive CQ/HCQ literature. This review summarizes the detailed data regarding CQ/HCQ's mechanisms of action while also providing insight into the overarching themes. Furthermore, this review provides clinical context to the application of these diverse drugs including their role in malaria, autoimmune disorders, cardiovascular disease, thrombus formation, malignancies, and viral infections.
Collapse
Affiliation(s)
- Britney Niemann
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
- Correspondence: ; Tel.: +1-304-293-1254
| | - Amanda Puleo
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Conley Stout
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Justin Markel
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Brian A. Boone
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
154
|
Zhang J, Kong X, Sun W, Wang L, Shen T, Chen M, Chen X. The RNA-binding protein RBM24 regulates lipid metabolism and SLC7A11 mRNA stability to modulate ferroptosis and inflammatory response. Front Cell Dev Biol 2022; 10:1008576. [PMID: 36478739 PMCID: PMC9720322 DOI: 10.3389/fcell.2022.1008576] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022] Open
Abstract
Lipids play a critical role in many cellular processes by serving as structural components of cell membranes or functioning as energy fuel and signaling molecules. The RNA-binding proteins RBM24 and RBM38 share an identical RNA-binding domain and thereby, regulate a group of same targets, such as p21. However, it is not certain whether RBM24 and RBM38 participates in lipid homeostasis. Here, lipidomic analysis showed that a deficiency in RBM24 or RBM38 leads to altered lipid metabolism, with more profound alteration by loss of RBM24 in MCF7 cells. We also showed that mice deficient in RBM24 were prone to chronic inflammation and liver steatosis, but not spontaneous tumors. These data let us speculate whether RBM24 regulates ferroptosis, a programmed cell death that links inflammation and liver steatosis via lipid peroxidation. Indeed, we found that over-expression of RBM24 protected, whereas knockout of RBM24 sensitized, cells to Erastin-induced ferroptosis by modulating the mRNA stability of SLC7A11, a ferroptosis inhibitor. Moreover, we showed that knockdown of SLC7A11 reversed the effect of RBM24 on ferroptosis. Together, our study revealed that RBM24 regulates lipid metabolism and SLC7A11 mRNA stability to modulate ferroptosis and inflammatory response.
Collapse
Affiliation(s)
- Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States,*Correspondence: Jin Zhang, ; Xinbin Chen,
| | - Xiangmudong Kong
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States
| | | | - Leyi Wang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States
| | - Tong Shen
- West Coast Metabolomics Center, UC, Davis, CA, United States
| | - Mingyi Chen
- Department of Pathology, Southwestern Medical Center, University of Texas, Austin, TX, United States
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States,*Correspondence: Jin Zhang, ; Xinbin Chen,
| |
Collapse
|
155
|
Yang H, Xu S, Tang L, Gong J, Fang H, Wei J, Su D. Targeting of non-apoptotic cancer cell death mechanisms by quercetin: Implications in cancer therapy. Front Pharmacol 2022; 13:1043056. [PMID: 36467088 PMCID: PMC9708708 DOI: 10.3389/fphar.2022.1043056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2023] Open
Abstract
The ultimate goal of cancer treatment is to kill cancer cells, based on the use of various therapeutic agents, such as chemotherapy, radiotherapy, or targeted therapy drugs. Most drugs exert their therapeutic effects on cancer by targeting apoptosis. However, alterations in apoptosis-related molecules and thus assisting cells to evade death, eventually lead to tumor cell resistance to therapeutic drugs. The increased incidence of non-apoptotic cell death modes such as induced autophagy, mitotic catastrophe, senescence, and necrosis is beneficial to overcoming multidrug resistance mediated by apoptosis resistance in tumor cells. Therefore, investigating the function and mechanism of drug-induced non-apoptotic cell death modes has positive implications for the development of new anti-cancer drugs and therapeutic strategies. Phytochemicals show strong potential as an alternative or complementary medicine for alleviating various types of cancer. Quercetin is a flavonoid compound widely found in the daily diet that demonstrates a significant role in inhibiting numerous human cancers. In addition to direct pro-tumor cell apoptosis, both in vivo and in vitro experiments have shown that quercetin exerts anti-tumor properties by triggering diverse non-apoptotic cell death modes. This review summarized the current status of research on the molecular mechanisms and targets through which quercetin-mediated non-apoptotic mode of cancer cell death, including autophagic cell death, senescence, mitotic catastrophe, ferroptosis, necroptosis, etc.
Collapse
Affiliation(s)
- Hao Yang
- Department of Pharmacy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Shan Xu
- Department of Pharmacy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Lidan Tang
- Department of Pharmacy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jinhong Gong
- Department of Pharmacy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Hufeng Fang
- Department of Pharmacy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jifu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Dan Su
- Department of Pharmacy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
156
|
New Iron Metabolic Pathways and Chelation Targeting Strategies Affecting the Treatment of All Types and Stages of Cancer. Int J Mol Sci 2022; 23:ijms232213990. [PMID: 36430469 PMCID: PMC9696688 DOI: 10.3390/ijms232213990] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
There is new and increasing evidence from in vitro, in vivo and clinical studies implicating the pivotal role of iron and associated metabolic pathways in the initiation, progression and development of cancer and in cancer metastasis. New metabolic and toxicity mechanisms and pathways, as well as genomic, transcription and other factors, have been linked to cancer and many are related to iron. Accordingly, a number of new targets for iron chelators have been identified and characterized in new anticancer strategies, in addition to the classical restriction of/reduction in iron supply, the inhibition of transferrin iron delivery, the inhibition of ribonucleotide reductase in DNA synthesis and high antioxidant potential. The new targets include the removal of excess iron from iron-laden macrophages, which affects anticancer activity; the modulation of ferroptosis; ferritin iron removal and the control of hyperferritinemia; the inhibition of hypoxia related to the role of hypoxia-inducible factor (HIF); modulation of the function of new molecular species such as STEAP4 metalloreductase and the metastasis suppressor N-MYC downstream-regulated gene-1 (NDRG1); modulation of the metabolic pathways of oxidative stress damage affecting mitochondrial function, etc. Many of these new, but also previously known associated iron metabolic pathways appear to affect all stages of cancer, as well as metastasis and drug resistance. Iron-chelating drugs and especially deferiprone (L1), has been shown in many recent studies to fulfill the role of multi-target anticancer drug linked to the above and also other iron targets, and has been proposed for phase II trials in cancer patients. In contrast, lipophilic chelators and their iron complexes are proposed for the induction of ferroptosis in some refractory or recurring tumors in drug resistance and metastasis where effective treatments are absent. There is a need to readdress cancer therapy and include therapeutic strategies targeting multifactorial processes, including the application of multi-targeting drugs involving iron chelators and iron-chelator complexes. New therapeutic protocols including drug combinations with L1 and other chelating drugs could increase anticancer activity, decrease drug resistance and metastasis, improve treatments, reduce toxicity and increase overall survival in cancer patients.
Collapse
|
157
|
Li J, Yuan J, Li Y, Wang J, Xie Q, Ma R, Wang J, Ren M, Lu D, Xu Z. d-Borneol enhances cisplatin sensitivity via autophagy dependent EMT signaling and NCOA4-mediated ferritinophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154411. [PMID: 36030746 DOI: 10.1016/j.phymed.2022.154411] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND d-Borneol has been widely used as a drug absorption enhancer, but there are few studies on the anti-resistance ability of d-borneol combined with cisplatin in cisplatin-resistant non-small cell lung cancer cells. Ferroptosis, autophagy and epithelial-mesenchymal transition (EMT) have been reported to be associated with drug resistance. PURPOSE To investigate the molecular mechanisms and sensitizing effects of d-borneol combined with cisplatin to against drug cisplatin resistance from the perspective of ferroptosis, autophagy and EMT resistance. METHODS H460/CDDP xenograft tumor model was established to verify the antitumor activity and safety in vivo. RNA sequencing was used to predict target molecules and signaling pathways. Reactive oxygen species (ROS) were used as marker of ferroptosis, and its level was determined by a dichlorodihydrofluorescein diacetate fluorescent probe and flow cytometry. Levels of glutathione (GSH), malondialdehyde (MDA), and antioxidants such as superoxide dismutase (SOD) and thioredoxin (Trx) involved in the balance of oxidative stress were measured by an assay kit or enzyme-linked immunosorbent assay. Western blotting and real-time polymerase chain reaction were used to assess the regulatory mechanism of EMT markers, autophagy, and ferroptosis signaling pathways. RESULTS d-Borneol in combination with cisplatin reduced tumor volume and weight, enhanced tumor-inhibiting effects, and alleviated cisplatin-induced damage to the liver and kidney in vivo. RNA-sequencing showed that differentially expressed genes were enriched in ferroptosis. d-Borneol in combination with cisplatin promoted ROS accumulation, increased the content of MDA levels, and decreased GSH, SOD, Trx, and heme oxygenase-1 expression to induce oxidative damage. d-Borneol combination with cisplatin induced ferroptosis by promoting nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and regulating intracellular iron ion transport via upregulating PRNP and downregulating PCBP2. In addition, d-borneol combined with cisplatin promoted autophagy by upregulating expression of LC3II/ATG5/Beclin-1 and inhibited the EMT by increasing the expression of epithelial marker E-cadherin and decreasing mesenchymal markers (N-cadherin and vimentin) and transcription factors (Snail and ZEB1). CONCLUSION For the first time, our study implies that d-borneol enhanced cisplatin sensitivity by inducing ferroptosis, promoting autophagy and inhibiting EMT progression, thereby enhancing antitumor activity. It suggests that d-borneol could be developed as a novel chemosensitizers.
Collapse
Affiliation(s)
- Jinxiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianmei Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Qian Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajun Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Danni Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
158
|
Todosenko N, Yurova K, Khaziakhmatova O, Malashchenko V, Khlusov I, Litvinova L. Heparin and Heparin-Based Drug Delivery Systems: Pleiotropic Molecular Effects at Multiple Drug Resistance of Osteosarcoma and Immune Cells. Pharmaceutics 2022; 14:pharmaceutics14102181. [PMID: 36297616 PMCID: PMC9612132 DOI: 10.3390/pharmaceutics14102181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/23/2022] Open
Abstract
One of the main problems of modern health care is the growing number of oncological diseases both in the elderly and young population. Inadequately effective chemotherapy, which remains the main method of cancer control, is largely associated with the emergence of multidrug resistance in tumor cells. The search for new solutions to overcome the resistance of malignant cells to pharmacological agents is being actively pursued. Another serious problem is immunosuppression caused both by the tumor cells themselves and by antitumor drugs. Of great interest in this context is heparin, a biomolecule belonging to the class of glycosaminoglycans and possessing a broad spectrum of biological activity, including immunomodulatory and antitumor properties. In the context of the rapid development of the new field of “osteoimmunology,” which focuses on the collaboration of bone and immune cells, heparin and delivery systems based on it may be of intriguing importance for the oncotherapy of malignant bone tumors. Osteosarcoma is a rare but highly aggressive, chemoresistant malignant tumor that affects young adults and is characterized by constant recurrence and metastasis. This review describes the direct and immune-mediated regulatory effects of heparin and drug delivery systems based on it on the molecular mechanisms of (multiple) drug resistance in (onco) pathological conditions of bone tissue, especially osteosarcoma.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Vladimir Malashchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Igor Khlusov
- Department of Morphology and General Pathology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Correspondence:
| |
Collapse
|
159
|
Liu Q, Yang Y, Cheng M, Cheng F, Chen S, Zheng Q, Sun Y, Chen L. The marine natural product, dicitrinone B, induces apoptosis through autophagy blockade in breast cancer. Int J Mol Med 2022; 50:130. [PMID: 36052845 PMCID: PMC9448296 DOI: 10.3892/ijmm.2022.5186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Being a highly conserved catabolic process, autophagy is induced by various forms of cellular stress, and its modulation has considerable potential as a cancer therapeutic approach. In the present study, it was demonstrated that dicitrinone B (DB), a rare carbon-bridged citrinin dimer, may exert anticancer effects by blocking autophagy at a late stage, without disrupting lysosomal function in MCF7 breast cancer and MDA-MB-231 triple-negative breast cancer cells. Furthermore, it was discovered that DB significantly enhanced intracellular reactive oxygen species (ROS) production and that the removal of ROS was followed by the attenuation of autophagy inhibition. In addition, DB exerted notable inhibitory effects on the proliferation and promoting effects on the apoptosis of MCF7 and MDA-MB-231 cells. In combination with conventional chemotherapeutic drugs, DB exhibited a further enhanced synergistic effect than when used as a single agent. Overall, the data of the present study demonstrate that DB may prove to be a promising autophagy inhibitor with anticancer activity against breast cancer.
Collapse
Affiliation(s)
- Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Yi Yang
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, P.R. China
| | - Miaomiao Cheng
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, P.R. China
| | - Fangting Cheng
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, P.R. China
| | - Shanshan Chen
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Qiuhong Zheng
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Yang Sun
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Li Chen
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, P.R. China
| |
Collapse
|
160
|
Guo L, Wu Z. FOXM1-mediated NUF2 expression confers temozolomide resistance to human glioma cells by regulating autophagy via the PI3K/AKT/mTOR signaling pathway. Neuropathology 2022; 42:430-446. [PMID: 35701983 DOI: 10.1111/neup.12824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Glioma is the most common malignant tumor in the central nervous system and has a high mortality rate. Temozolomide (TMZ) is a widely used chemotherapeutic drug for glioma. NDC80 kinetochore complex (NUF2) is suggested to play a regulatory role in different cancers, but its specific function and mechanism in glioblastoma TMZ resistance remain unknown. NUF2, assessed by reverse transcription quantitative polymerase chain reaction (RT-qPCR), was highly expressed in glioma cell lines. TMZ was used to treat cells to establish a TMZ-resistant cell line. The potential functions of NUF2 in glioma were assessed using cell counting kit-8 (CCK-8) assays, colony formation assays, 5-Ethynyl-2'-deoxyuridine (EdU) assays, flow cytometry, Western blotting, and a tumor xenograft model. The results showed that NUF2 knockdown attenuated malignant phenotypes of TMZ-resistant cells and prevented tumor growth. Mechanistically, as luciferase reporter assays and chromatin immunoprecipitation (ChIP) as showed, Fox transcription factor M1 (FOXM1) had binding sites on the NUF2 promoter. Rescue assays demonstrated that FOXM1 upregulation counteracted the inhibitory effects of NUF2 depletion on the malignancies of TMZ-resistant cells. This study demonstrates that FOXM1-activated NUF2 promotes TMZ to human glioma cells by regulating proliferation, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Liang Guo
- Department of Neurosurgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhangyi Wu
- Department of Neurosurgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
161
|
Fei Y, Xu J, Ge L, Chen L, Yu H, Pan L, Chen P. Establishment and validation of individualized clinical prognostic markers for LUAD patients based on autophagy-related genes. Aging (Albany NY) 2022; 14:7328-7347. [PMID: 36178365 PMCID: PMC9550247 DOI: 10.18632/aging.204097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/13/2022] [Indexed: 12/24/2022]
Abstract
There is considerable heterogeneity in the genomic drivers of lung adenocarcinoma, which has a dismal prognosis. Bioinformatics analysis was performed on lung adenocarcinoma (LUAD) datasets to establish a multi-autophagy gene model to predict patient prognosis. LUAD data were downloaded from The Cancer Genome Atlas (TCGA) database as a training set to construct a LUAD prognostic model. According to the risk score, a Kaplan-Meier cumulative curve was plotted to evaluate the prognostic value. Furthermore, a nomogram was established to predict the three-year and five-year survival of patients with LUAD based on their prognostic characteristics. Two genes (ITGB1 and EIF2AK3) were identified in the autophagy-related prognostic model, and the multivariate Cox proportional risk model showed that risk score was an independent predictor of prognosis in LUAD patients (HR=3.3, 95%CI= 2.3 to 4.6, P< 0.0001). The Kaplan-Meier cumulative curve showed that low-risk patients had significantly better overall (P<0.0001). The validation dataset GSE68465 further confirmed the nomogram’s robust ability to assess the prognosis of LUAD patients. A prognosis model of autophagy-related genes based on a LUAD dataset was constructed and exhibited diagnostic value in the prognosis of LUAD patients. Moreover, real-time qPCR confirmed the expression patterns of EIF2AK3 and ITGB1 in LUAD cell lines. Two key autophagy-related genes have been suggested as prognostic markers for lung adenocarcinoma.
Collapse
Affiliation(s)
- Yuchang Fei
- Department of Integrated Chinese and Western Medicine, The First People’s Hospital of Jiashan, Jiaxing, Zhejiang, China
| | - Junyi Xu
- Information Center, The First People’s Hospital of Jiashan, Jiaxing, Zhejiang, China
| | - Liping Ge
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Xuhui, Shanghai, China
| | - Luting Chen
- Department of Integrated Chinese and Western Medicine, The First People’s Hospital of Wenling, Taizhou, Zhejiang, China
| | - Huan Yu
- Ningbo Yinzhou Second Hospital, Ningbo, Zhejiang, China
| | - Lei Pan
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Peifeng Chen
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
162
|
Zhu Y, Wang A, Zhang S, Kim J, Xia J, Zhang F, Wang D, Wang Q, Wang J. Paclitaxel-loaded ginsenoside Rg3 liposomes for drug-resistant cancer therapy by dual targeting of the tumor microenvironment and cancer cells. J Adv Res 2022:S2090-1232(22)00209-0. [PMID: 36167294 DOI: 10.1016/j.jare.2022.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Inherent or acquired resistance to paclitaxel (PTX) is a pivotal challenge for chemotherapy treatment of multidrug-resistant (MDR) breast cancer. Although various targeted drug-delivery systems, including nanoparticles and liposomes, are effective for MDR cancer treatment, their efficacy is restricted by immunosuppressive tumor microenvironment (TME). METHODS Ginsenosides Rg3 was used to formulate unique Rg3-based liposomes loaded with PTX to establish Rg3-PTX-LPs, which were prepared by the thin-film hydration method. The stability of the Rg3-PTX-LPs was evaluated by particle size analysis through dynamic light scattering. The active targeting effect of Rg3-based liposomes was examined in an MCF-7/T xenograft model by an in a vivo imaging system. To evaluate the antitumor activity and mechanism of Rg3-PTX-LP, MTT, apoptosis assays, TAM regulation, and TME remodeling were performed in MCF-7/T cells in vitro and in vivo. RESULTS Rg3-PTX-LPs could specifically distribute to MCF7/T cancer cells and TME simultaneously, mainly through the recognition of GLUT-1. The drug resistance reversing capability and in vivo antitumor effect of Rg3-PTX-LPs were significantly improved compared with conventional cholesterol liposomes. The TME remodeling mechanisms of Rg3-PTX-LPs included inhibiting IL-6/STAT3/p-STAT3 pathway activation to repolarize protumor M2 macrophages to antitumor M1 phenotype, suppressing myeloid-derived suppressor cells (MDSCs), decreasing tumor-associated fibroblasts (TAFs) and collagen fibers in TME, and promoting apoptosis of tumor cells. Hence, through the dual effects of targeting tumor cells and TME remodeling, Rg3-PTX-LPs achieved a high tumor inhibition rate of 90.3%. CONCLUSION Our multifunctional Rg3-based liposome developed in the present study offered a promising strategy for rescuing the drug resistance tumor treatment.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, PR China; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Anni Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, PR China
| | - Shuya Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, PR China
| | - Jisu Kim
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, PR China
| | - Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, PR China
| | - Fengxue Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Dan Wang
- Xiamen Ginposome Pharmaceutical Co., Ltd., Xiamen 361026, People's Republic of China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, PR China; Institute of Integrated Chinese and Western Medicine, Fudan University, Shanghai 200040, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
163
|
Wang X, Yu J, Liu X, Luo D, Li Y, Song L, Jiang X, Yin X, Wang Y, Chai L, Luo T, Jing J, Shi H. PSMG2-controlled proteasome-autophagy balance mediates the tolerance for MEK-targeted therapy in triple-negative breast cancer. Cell Rep Med 2022; 3:100741. [PMID: 36099919 PMCID: PMC9512673 DOI: 10.1016/j.xcrm.2022.100741] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/21/2022] [Accepted: 08/23/2022] [Indexed: 05/29/2023]
Abstract
Although the MAPK pathway is aberrantly activated in triple-negative breast cancers (TNBCs), the clinical outcome of MEK-targeted therapy is still poor. Through a genome-wide CRISPR-Cas9 library screening, we find that inhibition of PSMG2 sensitizes TNBC cells BT549 and MB468 to the MEK inhibitor AZD6244. Mechanistically, PSMG2 knockdown impairs proteasome function, which in turn activates autophagy-mediated PDPK1 degradation. The PDPK1 degradation significantly enhances AZD6244-induced tumor cell growth inhibition by interrupting the negative feedback signals toward the AKT pathway. Consistently, co-targeting proteasomes and MEK with inhibitors synergistically suppresses tumor cell growth. The autophagy inhibitor chloroquine partially relieves the PDPK1 degradation and reverses the growth inhibition induced by combinatorial inhibition of MEK and proteasome. The combination regimen with the proteasome inhibitor MG132 plus AZD6244 synergistically inhibits tumor growth in a 4T1 xenograft mouse model. In summary, our study not only unravels the mechanism of MEK inhibitor resistance but also provides a combinatorial therapeutic strategy for TNBC in clinics.
Collapse
Affiliation(s)
- Xueyan Wang
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jing Yu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Xiaowei Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Dan Luo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Yanchu Li
- West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linlin Song
- Department of Ultrasound and Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Xian Jiang
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Xiaomeng Yin
- Department of Biotherapy, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Wang
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Chai
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ting Luo
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jing Jing
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China.
| | - Hubing Shi
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China.
| |
Collapse
|
164
|
Hu S, Liu Y, Guan S, Qiu Z, Liu D. Natural products exert anti-tumor effects by regulating exosomal ncRNA. Front Oncol 2022; 12:1006114. [PMID: 36203417 PMCID: PMC9530706 DOI: 10.3389/fonc.2022.1006114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Currently, more than 60% of the approved anti-cancer drugs come from or are related to natural products. Natural products and exosomal non-coding RNAs (ncRNAs) exert anti-cancer effects through various regulatory mechanisms, which are of great research significance. Exosomes are a form of intercellular communication and contain ncRNAs that can act as intercellular signaling molecules involved in the metabolism of tumor cells. This review exemplifies some examples of natural products whose active ingredients can play a role in cancer prevention and treatment by regulating exosomal ncRNAs, with the aim of illustrating the mechanism of action of exosomal ncRNAs in cancer prevention and treatment. Meanwhile, the application of exosomes as natural drug delivery systems and predictive disease biomarkers in cancer prevention and treatment is introduced, providing research ideas for the development of novel anti-tumor drugs.
Collapse
Affiliation(s)
| | | | | | | | - Da Liu
- *Correspondence: Zhidong Qiu, ; Da Liu,
| |
Collapse
|
165
|
Protein tyrosine kinase inhibitor resistance in malignant tumors: molecular mechanisms and future perspective. Signal Transduct Target Ther 2022; 7:329. [PMID: 36115852 PMCID: PMC9482625 DOI: 10.1038/s41392-022-01168-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/08/2022] [Accepted: 08/26/2022] [Indexed: 02/07/2023] Open
Abstract
AbstractProtein tyrosine kinases (PTKs) are a class of proteins with tyrosine kinase activity that phosphorylate tyrosine residues of critical molecules in signaling pathways. Their basal function is essential for maintaining normal cell growth and differentiation. However, aberrant activation of PTKs caused by various factors can deviate cell function from the expected trajectory to an abnormal growth state, leading to carcinogenesis. Inhibiting the aberrant PTK function could inhibit tumor growth. Therefore, tyrosine kinase inhibitors (TKIs), target-specific inhibitors of PTKs, have been used in treating malignant tumors and play a significant role in targeted therapy of cancer. Currently, drug resistance is the main reason for limiting TKIs efficacy of cancer. The increasing studies indicated that tumor microenvironment, cell death resistance, tumor metabolism, epigenetic modification and abnormal metabolism of TKIs were deeply involved in tumor development and TKI resistance, besides the abnormal activation of PTK-related signaling pathways involved in gene mutations. Accordingly, it is of great significance to study the underlying mechanisms of TKIs resistance and find solutions to reverse TKIs resistance for improving TKIs efficacy of cancer. Herein, we reviewed the drug resistance mechanisms of TKIs and the potential approaches to overcome TKI resistance, aiming to provide a theoretical basis for improving the efficacy of TKIs.
Collapse
|
166
|
Fang J, Ma Y, Li Y, Li J, Zhang X, Han X, Ma S, Guan F. CCT4 knockdown enhances the sensitivity of cisplatin by inhibiting glycolysis in human esophageal squamous cell carcinomas. Mol Carcinog 2022; 61:1043-1055. [DOI: 10.1002/mc.23460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/20/2022] [Accepted: 08/29/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Jiarui Fang
- School of Life Sciences Zhengzhou University Zhengzhou Henan China
| | - Yingchao Ma
- School of Life Sciences Zhengzhou University Zhengzhou Henan China
| | - Ya Li
- School of Life Sciences Zhengzhou University Zhengzhou Henan China
| | - Jianhui Li
- Department of Pathology Xuchang Central Hospital Affiliated to Henan University of Science and Technology Xuchang China
| | - Xishen Zhang
- School of Life Sciences Zhengzhou University Zhengzhou Henan China
| | - Xiao Han
- School of Life Sciences Zhengzhou University Zhengzhou Henan China
| | - Shanshan Ma
- School of Life Sciences Zhengzhou University Zhengzhou Henan China
| | - Fangxia Guan
- School of Life Sciences Zhengzhou University Zhengzhou Henan China
| |
Collapse
|
167
|
Wang C, Li Y, Tian Y, Ma W, Sun Y. Effects of polymer carriers on the occurrence and development of autophagy in drug delivery. NANOSCALE ADVANCES 2022; 4:3676-3688. [PMID: 36133340 PMCID: PMC9470016 DOI: 10.1039/d2na00355d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/18/2022] [Indexed: 06/16/2023]
Abstract
Autophagy is an evolutionarily conserved catabolic process that can degrade cytoplasmic materials and recycle energy to maintain metabolite homeostasis in cells. Autophagy is closely related to various physiological or pathological processes. Macromolecular materials are widely used in drug delivery systems and disease treatments due to their intrinsic effects, such as altered pharmacokinetics and biodistribution. Interaction of autophagic flux or the signal pathway with macromolecules may cause autophagy inhibition or autophagy cell death. This review covers autophagy regulation pathways and macromolecular materials (including functional micelles, biodegradable and pH-sensitive polymers, biomacromolecules, dendrimers, coordination polymers, and hybrid nanoparticles) mediated autophagy modulation.
Collapse
Affiliation(s)
- Changduo Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| | - Yang Li
- Department of Pharmacy, Qingdao Municipal Hospital Qingdao 266000 China
| | - Yu Tian
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| | - Wenyuan Ma
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| |
Collapse
|
168
|
Embelin and Its Derivatives: Design, Synthesis, and Potential Delivery Systems for Cancer Therapy. Pharmaceuticals (Basel) 2022; 15:ph15091131. [PMID: 36145352 PMCID: PMC9505931 DOI: 10.3390/ph15091131] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Embelin is a naturally occurring benzoquinone that inhibits the growth of cancer cells, making it a potent anticancer drug. However, the low water solubility of embelin restricts its clinical applicability. This review provides a concise summary and in-depth analysis of the published literature on the design and synthesis of embelin derivatives possessing increased aqueous solubility and superior therapeutic efficacy. In addition, the potential of drug delivery systems to improve the anticancer capabilities of embelin and its derivatives is discussed.
Collapse
|
169
|
Potential Therapeutic Agents against Paclitaxel—And Sorafenib-Resistant Papillary Thyroid Carcinoma. Int J Mol Sci 2022; 23:ijms231810378. [PMID: 36142303 PMCID: PMC9499486 DOI: 10.3390/ijms231810378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/24/2022] Open
Abstract
Thyroid carcinoma, a disease in which malignant cells form in the thyroid tissue, is the most common endocrine carcinoma, with papillary thyroid carcinoma (PTC) accounting for nearly 80% of total thyroid carcinoma cases. However, the management of metastatic or recurrent therapy-refractory PTC is challenging and requires complex carcinoma therapy. In this study, we proposed a new clinical approach for the treatment of therapy-refractory PTC. We identified sarco/endoplasmic reticulum calcium ATPase (SERCA) as an essential factor for the survival of PTC cells refractory to the treatment with paclitaxel or sorafenib. We validated its use as a potential target for developing drugs against resistant PTC, by using patient-derived paclitaxel- or sorafenib-resistant PTC cells. We further discovered novel SERCA inhibitors, candidates 7 and 13, using the evolutionary chemical binding similarity method. These novel SERCA inhibitors determined a substantial reduction of tumors in a patient-derived xenograft tumor model developed using paclitaxel- or sorafenib-resistant PTC cells. These results could provide a basis for clinically meaningful progress in the treatment of refractory PTC by identifying a novel therapeutic strategy: using a combination therapy between sorafenib or paclitaxel and specific SERCA inhibitors for effectively and selectively targeting extremely malignant cells such as antineoplastic-resistant and carcinoma stem-like cells.
Collapse
|
170
|
Zhang F, Chen Q, Chen P, Liu C, Wang H, Zhao L. The lncRNA CRNDE is regulated by E2F6 and sensitizes gastric cancer cells to chemotherapy by inhibiting autophagy. J Cancer 2022; 13:3061-3072. [PMID: 36046639 PMCID: PMC9414014 DOI: 10.7150/jca.65871] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy is an important treatment for gastric cancer (GC), but the primary and secondary drug resistance of tumours to chemotherapy seriously affects its curative effect. In recent years, the relationship between long noncoding RNAs (lncRNAs) and malignant tumours has received increasing attention. Based on accumulating evidence, lncRNAs are involved in the chemoresistance of GC, but the underlying mechanisms remain unclear. In this study, we identified the lncRNA colorectal neoplasia differentially expressed (CRNDE) as an important regulator of autophagy-associated chemoresistance in GC. Mechanistically, overexpression of CRNDE inhibits autophagy and induces apoptosis, thereby sensitizing GC cells to chemotherapy drugs. Moreover, E2F6, a classical transcriptional inhibitor, is confirmed to be upregulated in GC and represses the expression of CRNDE. The E2F6-CRNDE axis is clinically related to chemoresistant GC and poor outcomes in patients with advanced GC. Our findings suggest that the E2F6-CRNDE axis is a viable therapeutic target to protect against chemoresistance in GC.
Collapse
Affiliation(s)
- Feifei Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qian Chen
- Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Peng Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoqun Liu
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Wang
- Department of Medical Oncology, Affiliated Tumour Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liang Zhao
- Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
171
|
Abu Rabah RR, Sebastian A, Vunnam S, Sultan S, Tarazi H, Anbar HS, Shehata MK, Zaraei SO, Elgendy SM, Al Shamma SA, Omar HA, Al-Tel TH, El-Gamal MI. Design, synthesis, and biological evaluation of a new series of pyrazole derivatives: Discovery of potent and selective JNK3 kinase inhibitors. Bioorg Med Chem 2022; 69:116894. [DOI: 10.1016/j.bmc.2022.116894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 11/02/2022]
|
172
|
Xiong Y, Xiong C, Li P, Shan X. Rutaecarpine prevents the malignant biological properties of breast cancer cells by the miR-149-3p/S100A4 axis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:930. [PMID: 36172090 PMCID: PMC9511192 DOI: 10.21037/atm-22-3765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Abstract
Background Breast cancer (BC) is a frequent malignancy that endangers women's health, and its fatality rate ranks 1st among female malignancies. Research has shown that rutaecarpine (RUT), which is a Chinese herbal medicine, blocks the proliferation of cancer cells by a variety of molecular mechanisms. However, the possible effects and mechanism of RUT in the autophagy and angiogenesis of BC cells has not been clearly articulated. Methods MiR-149-3p and S100A4 expression levels were assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and the optimal concentration and time of RUT was confirmed by Cell Counting Kit-8 (CCK-8) assays of the BC cells. After treatment, changes in cell proliferation and the cell cycle were evaluated by CCK-8 assays, clone formation assays, and flow cytometry, and the levels of apoptosis, autophagy, and angiogenesis-related proteins were identified by Western blot. The targeted regulation of miR-149-3p on S100A4 was also examined by luciferase reporter assays. Results We found that RUT inhibited cell growth and upregulated miR-149-3p in MDA-MB-231 cells. In relation to the biological function activity, RUT attenuated proliferation and angiogenesis, and induced cell-cycle arrest and autophagy by miR-149-3p in the MDA-MB-231 cells. Additionally, miR-149-3p downregulated S100A4 by targeting binding to S100A4, and S100A4 was required for miR-149-3p to play a role in BC progression. We also discovered that an autophagy agonist (rapamycin) or an angiogenesis inhibitor (TNP-470) changed BC progression mediated by the RUT/miR-149-3p/S100A4 axis. Conclusions RUT blocks the malignant behaviors of BC cells through the miR-149-3p/S100A4 axis and thus alters autophagy and angiogenesis. Thus, the RUT-mediated miR-149-3p/S100A4 axis might be an underlying therapeutic agent and target for BC.
Collapse
Affiliation(s)
- Yi Xiong
- General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.,General Surgery, Wuhan Asia General Hospital, Wuhan, China
| | - Chao Xiong
- General Surgery, Wuhan Asia General Hospital, Wuhan, China
| | - Peng Li
- General Surgery, Wuhan Asia General Hospital, Wuhan, China
| | - Xuehua Shan
- General Surgery, Wuhan Asia General Hospital, Wuhan, China
| |
Collapse
|
173
|
Eptaminitaki GC, Stellas D, Bonavida B, Baritaki S. Long Non-coding RNAs (lncRNAs) signaling in Cancer Chemoresistance: From Prediction to Druggability. Drug Resist Updat 2022; 65:100866. [DOI: 10.1016/j.drup.2022.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/03/2022]
|
174
|
Runlin W, Xiang P, Juan L, Shu Y. Soybean Isoflavones Activating Autophagy and Improving the Chemosensitivity of Carboplatin to Ovarian Cancer Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objection: To discuss soybean isoflavones activating autophagy and improving the chemosensitivity of carboplatin to ovarian cancer cells. Materials and Methods: Using SKOV3 and A2780 cell lines as research object, dividing into Normal, Carb (treated with 50 μmol/L
carboplatin) and Carb+Soy (treated with 50 μmol/L carboplatin and 80 μmol/L soybean isoflavone). Evaluating cell proliferation by EdU and MTT assay; measuring cell apoptosis rate by flow cytometry; observation cells’ autophagy by transmission electron microscope (TEM);
LC 3B protein expression were evaluated by cellular immunofluorescence and using WB assay to evaluate autophagy relative proteins expression. Results: Compared with Normal group, the cell proliferation were significantly depressed with cell apoptosis rates significantly increasing (P
< 0.01, respectively); and autophagy enhancing; with LC 3B, LC 3II/LC 3I ratio and Beclin 1 significantly up-regulation and P62 protein significantly down-regulation (P < 0.01, respectively) in Carb and Carb+Soy groups. And there were significantly differences between Carb and
Carb+Soy groups in EdU cell number, cell proliferation, apoptosis rate, autophagy, LC 3B, LC 3II/LC 3I ratio, Beclin 1 and P62 protein expression. Conclusion: Soy activating autophagy and improving the chemosensitivity of carboplatin to ovarian cancer cells.
Collapse
Affiliation(s)
- Wu Runlin
- Wuhu Hospital Affiliated to East China Normal University, Wuhu, 241000, China
| | - Pan Xiang
- Wuhu Hospital Affiliated to East China Normal University, Wuhu, 241000, China
| | - Lu Juan
- Wuhu Hospital Affiliated to East China Normal University, Wuhu, 241000, China
| | - Yao Shu
- Wuhu Hospital Affiliated to East China Normal University, Wuhu, 241000, China
| |
Collapse
|
175
|
Shao BZ, Chai NL, Yao Y, Li JP, Law HKW, Linghu EQ. Autophagy in gastrointestinal cancers. Front Oncol 2022; 12:975758. [PMID: 36091106 PMCID: PMC9459114 DOI: 10.3389/fonc.2022.975758] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022] Open
Abstract
Gastrointestinal cancers are a group of cancers occurred in gastrointestinal tissues with high morbidity and mortality rate. Although numerous studies were conducted on the investigation of gastrointestinal cancers, the real mechanisms haven’t been discovered, and no effective methods of prevention and treatment of gastrointestinal cancers have been developed. Autophagy, a vital catabolic process in organisms, have been proven to participate in various mechanisms and signaling pathways, thus producing a regulatory effect on various diseases. The role of autophagy in gastrointestinal cancers remains unclear due to its high complexity. In this review, firstly, the biological features of autophagy will be introduced. Secondly, the role of autophagy in three popular gastrointestinal cancers, namely esophageal cancer, gastric cancer, and colorectal cancer will be described and discussed by reviewing the related literature. We aimed to bring novel insights in exploring the real mechanisms for gastrointestinal cancers and developing effective and efficient therapeutic methods to treat gastrointestinal cancers.
Collapse
Affiliation(s)
- Bo-Zong Shao
- Department of Gastroenterology, General Hospital of the Chinese People’s Liberation Army, Beijing, China
- Department of Health Technology and Informatics, Faculty of Health and Social Science, The Hong Kong Polytechnic University, Hunghom, Hong Kong SAR, China
- *Correspondence: En-Qiang Linghu, ; Helen Ka Wai Law, ; Bo-Zong Shao,
| | - Ning-Li Chai
- Department of Gastroenterology, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Yi Yao
- Department of Gastroenterology, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Jin-Ping Li
- Department of Gastroenterology, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Helen Ka Wai Law
- Department of Health Technology and Informatics, Faculty of Health and Social Science, The Hong Kong Polytechnic University, Hunghom, Hong Kong SAR, China
- *Correspondence: En-Qiang Linghu, ; Helen Ka Wai Law, ; Bo-Zong Shao,
| | - En-Qiang Linghu
- Department of Gastroenterology, General Hospital of the Chinese People’s Liberation Army, Beijing, China
- *Correspondence: En-Qiang Linghu, ; Helen Ka Wai Law, ; Bo-Zong Shao,
| |
Collapse
|
176
|
Lu L, Wang T, Fang C, Song L, Qian C, Lv Z, Fang Y, Liu X, Yu X, Xu X, Su C, Chen F, Zhang K. Oncolytic Impediment/Promotion Balance Disruption by Sonosensitizer-Free Nanoplatforms Unfreezes Autophagy-Induced Resistance to Sonocatalytic Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:36462-36472. [PMID: 35939287 DOI: 10.1021/acsami.2c09443] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Autophagy as a double-edged sword features an oncolytic impediment/promotion balance, which manipulates tumor progression. From this perspective, a sonosensitizer-free targeting oncolytic nanoplatform (SFTON) consisting of chloroquine (CQ) and porphyrin-structured metal centers (PMCS) was engineered to break this balance for enhancing antitumor activity. Porphyrin structure retention in a ZIF-8-derived hydrophobic carbon skeleton retained high stability and high sonocatalytic activity, and the hydrophobic carbon skeleton capable of adsorbing air provided cavitation nuclei for further elevating sonocatalytic activity. More significantly, the encapsulated CQ as the autophagy inhibitor reprogrammed autophagy, terminated the autophagy-induced self-protection or self-detoxification, and unfroze the resistances to reactive oxygen species (ROS) therapy associated with ROS accumulation and ROS activity. Systematic experiments reveal the action principles and validate that the induced apoptosis and blockaded autophagosome escalation into the autolysosome were two activated pathways to magnify the antitumor sonocatalytic therapy. Contributed by these actions, the SFTON-unlocked oncolytic impediment/promotion balance disruption strategy acquired considerable antitumor outcomes in vivo and in vitro against liver tumor progression, especially after combining with AS1411-mediated active targeting. This impediment/promotion balance disruption enabled by the SFTON can serve as a general method to elevate ROS-based antitumor activity.
Collapse
Affiliation(s)
- Lu Lu
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Ultrasound, Affiliated Hospital of Guangdong Medical University, No. 57 Peoples Avenue, Zhanjiang 524000, Guangdong Province, P. R. China
| | - Taixia Wang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Chao Fang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Li Song
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Cheng Qian
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Zheng Lv
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Yujia Fang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Xinyu Liu
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Xin Yu
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Xiaohong Xu
- Department of Medical Ultrasound, Affiliated Hospital of Guangdong Medical University, No. 57 Peoples Avenue, Zhanjiang 524000, Guangdong Province, P. R. China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Fubo Chen
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| | - Kun Zhang
- Central Laboratory, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China
| |
Collapse
|
177
|
Long D, Chen Y, Qu L, Dong Y. Lidocaine inhibits the proliferation and migration of endometrial cancer cells, and promotes apoptosis by inducing autophagy. Oncol Lett 2022; 24:347. [PMID: 36072001 PMCID: PMC9434716 DOI: 10.3892/ol.2022.13467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/26/2022] [Indexed: 11/06/2022] Open
Abstract
As a gynecological malignancy, endometrial cancer (EC) has a high incidence and mortality rate in women. The aim of the present study was to investigate the mechanism of EC and to identify novel effective treatment methods for this disease. The viability and proliferation of the RL95-2 human endometrial cancer cell line were assessed using Cell Counting Kit-8 assays. Colony formation, wound healing, Transwell, TUNEL and immunofluorescence assays were used to assess the effects of 5, 10 and 15 mM lidocaine on the colony formation, migration, invasiveness, apoptosis and Beclin 1 protein expression of RL95-2 cells, respectively. Furthermore, western blotting was used to analyze the protein expression levels of apoptosis- and autophagy-related proteins. The results demonstrated that lidocaine inhibited the viability, proliferation and migration of EC cells, and promoted apoptosis. Furthermore, lidocaine was demonstrated to induce autophagy and Beclin 1 protein expression in EC cells. In conclusion, lidocaine inhibited the proliferation and migration of EC cells, and promoted apoptosis via autophagy induction, which indicated that lidocaine may be a potential therapeutic drug for the treatment of EC.
Collapse
Affiliation(s)
- Dingde Long
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Yayu Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Liangchao Qu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Yang Dong
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
178
|
Tan L, Peng D, Cheng Y. Significant position of C-myc in colorectal cancer: a promising therapeutic target. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 24:2295-2304. [PMID: 35972682 DOI: 10.1007/s12094-022-02910-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/23/2022] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is a malignant tumor initiating from the mucosa of the colorectum. According to the 2020 statistics from the World Health Organization, there are 10.0% CRC cases among all 19.3 million new cancers, followed by lung and breast cancer, and 9.4% CRC cases among all 9.9 million cancer deaths, ranking second. The population of CRC patients in China is large, and its incidence and mortality continue to increase each year. Despite the continuous development of surgical methods, chemotherapy, radiotherapy, targeted therapy and immunotherapy, the overall survival of CRC patients remains low. Past research has suggested that c-myc plays a pivotal role in the development of CRC. A higher expression level of c-Myc is a negative prognostic marker in CRC. However, there are few drugs targeting c-myc directly. Therefore, we focused on discovering the mechanism of c-myc in CRC to provide a reference for a better therapy choice for patients.
Collapse
Affiliation(s)
- Li Tan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yong Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
179
|
Tong F, Xu L, Xu S, Zhang M. Identification of an autophagy-related 12-lncRNA signature and evaluation of NFYC-AS1 as a pro-cancer factor in lung adenocarcinoma. Front Genet 2022; 13:834935. [PMID: 36105077 PMCID: PMC9466988 DOI: 10.3389/fgene.2022.834935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: To develop an autophagy-related lncRNA-based risk signature and corresponding nomogram to predict overall survival (OS) for LUAD patients and investigate the possible meaning of screened factors.Methods: Differentially expressed lncRNAs and autophagy genes were screened between normal and LUAD tumor samples from the TCGA LUAD dataset. Univariate and multivariate Cox regression analyses were performed to construct the lncRNA-based risk signature and nomogram incorporating clinical information. Then, the accuracy and sensitivity were confirmed by the AUC of ROC curves in both training and validation cohorts. qPCR, immunoblot, shRNA, and ectopic expression were used to verify the positive regulation of NFYC-AS1 on BIRC6. CCK-8, immunofluorescence, and flow cytometry were used to confirm the influence of NFYC-AS1 on cell proliferation, autophagy, and apoptosis via BIRC6.Results: A 12-lncRNA risk signature and a nomogram combining related clinical information were constructed. Furthermore, the abnormal increase of NFYC-AS1 may promote LUAD progression through the autophagy-related gene BIRC6.Conclusion: 12-lncRNA signature may function as a predictive marker for LUAD patients, and NFYC-AS1 along with BIRC6 may function as carcinogenic factors in a combinatorial manner.
Collapse
Affiliation(s)
- Fang Tong
- Department of Medical Immunology, School of Medicine, Anhui University of Science and Technology, Anhui, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Anhui, China
| | - Lifa Xu
- Department of Medical Immunology, School of Medicine, Anhui University of Science and Technology, Anhui, China
| | - Sheng Xu
- The First Affiliated Hospital, Anhui University of Science and Technology, Anhui, China
| | - Mingming Zhang
- Department of Medical Immunology, School of Medicine, Anhui University of Science and Technology, Anhui, China
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
- *Correspondence: Mingming Zhang,
| |
Collapse
|
180
|
Abaza A, Vasavada AM, Sadhu A, Valencia C, Fatima H, Nwankwo I, Anam M, Maharjan S, Amjad Z, Khan S. A Systematic Review of Apoptosis in Correlation With Cancer: Should Apoptosis Be the Ultimate Target for Cancer Treatment? Cureus 2022; 14:e28496. [PMID: 36185861 PMCID: PMC9514374 DOI: 10.7759/cureus.28496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/28/2022] [Indexed: 11/05/2022] Open
Abstract
Targeting apoptosis in cancer therapy has become increasingly popular, and there has been an increasing debate on whether apoptosis should be one of the main targets of therapy in cancer management. This study demonstrates the definition of apoptosis, the signaling pathways, and the pathogenesis behind it. We also show the correlation between apoptosis and cancer and how cancer can evade apoptosis to develop resistance to therapy. In addition, we illustrate the efficacy of adding pro-apoptotic therapy to conventional radio-chemotherapy cancer treatment. A systematic review was conducted using PubMed, PubMed Central (PMC), and ResearchGate, including papers written in English, focusing on adult and geriatric populations, in literature reviews, systematic reviews, and randomized controlled trials published in the last 25 years with relevance to the question. Based on the findings of this review, we conclude that apoptosis is a very sophisticated programmed cellular death with many signaling pathways. Its evasion should be considered one of the hallmarks of cancer and is responsible for multiple drug resistance (MDR) to cancer therapy. Targeting apoptosis seems promising, especially if combined with radio-chemotherapy.
Collapse
Affiliation(s)
- Abdelrahman Abaza
- Pathology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Advait M Vasavada
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Akhil Sadhu
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Carla Valencia
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Hameeda Fatima
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ijeoma Nwankwo
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mahvish Anam
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shrinkhala Maharjan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Zainab Amjad
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
181
|
Peña-Flores JA, Bermúdez M, Ramos-Payán R, Villegas-Mercado CE, Soto-Barreras U, Muela-Campos D, Álvarez-Ramírez A, Pérez-Aguirre B, Larrinua-Pacheco AD, López-Camarillo C, López-Gutiérrez JA, Garnica-Palazuelos J, Estrada-Macías ME, Cota-Quintero JL, Barraza-Gómez AA. Emerging role of lncRNAs in drug resistance mechanisms in head and neck squamous cell carcinoma. Front Oncol 2022; 12:965628. [PMID: 35978835 PMCID: PMC9376329 DOI: 10.3389/fonc.2022.965628] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) originates in the squamous cell lining the mucosal surfaces of the head and neck region, including the oral cavity, nasopharynx, tonsils, oropharynx, larynx, and hypopharynx. The heterogeneity, anatomical, and functional characteristics of the patient make the HNSCC a complex and difficult-to-treat disease, leading to a poor survival rate and a decreased quality of life due to the loss of important physiologic functions and aggressive surgical injury. Alteration of driver-oncogenic and tumor-suppressing lncRNAs has recently been recently in HNSCC to obtain possible biomarkers for diagnostic, prognostic, and therapeutic approaches. This review provides current knowledge about the implication of lncRNAs in drug resistance mechanisms in HNSCC. Chemotherapy resistance is a major therapeutic challenge in HNSCC in which lncRNAs are implicated. Lately, it has been shown that lncRNAs involved in autophagy induced by chemotherapy and epithelial–mesenchymal transition (EMT) can act as mechanisms of resistance to anticancer drugs. Conversely, lncRNAs involved in mesenchymal–epithelial transition (MET) are related to chemosensitivity and inhibition of invasiveness of drug-resistant cells. In this regard, long non-coding RNAs (lncRNAs) play a pivotal role in both processes and are important for cancer detection, progression, diagnosis, therapy response, and prognostic values. As the involvement of more lncRNAs is elucidated in chemoresistance mechanisms, an improvement in diagnostic and prognostic tools could promote an advance in targeted and specific therapies in precision oncology.
Collapse
Affiliation(s)
- José A. Peña-Flores
- Faculty of Odontology, Autonomous University of Chihuahua, Chihuahua, Mexico
| | - Mercedes Bermúdez
- Faculty of Odontology, Autonomous University of Chihuahua, Chihuahua, Mexico
- *Correspondence: Mercedes Bermúdez,
| | - Rosalío Ramos-Payán
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Culiacán, Mexico
| | | | - Uriel Soto-Barreras
- Faculty of Odontology, Autonomous University of Chihuahua, Chihuahua, Mexico
| | | | | | | | | | | | - Jorge A. López-Gutiérrez
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Culiacán, Mexico
- Faculty of Biology, Autonomous University of Sinaloa, Culiacán, Mexico
| | | | | | - Juan L. Cota-Quintero
- Faculty of Biology, Autonomous University of Sinaloa, Culiacán, Mexico
- Faculty of Odontology , Autonomous University of Sinaloa, Culiacán, Mexico
| | | |
Collapse
|
182
|
Wang Y, Chen B, Xiao M, Wang X, Peng Y. Brucea javanica Oil Emulsion Promotes Autophagy in Ovarian Cancer Cells Through the miR-8485/LAMTOR3/mTOR/ATG13 Signaling Axis. Front Pharmacol 2022; 13:935155. [PMID: 35959437 PMCID: PMC9358144 DOI: 10.3389/fphar.2022.935155] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Ovarian cancer is a common malignant tumor of the female reproductive tract, with the highest mortality rate. At present, no effective approaches to improve the survival rate exist. B. javanica Oil Emulsion (BJOE), an extract from B. javanica (L.) Merr. [Simaroubaceae], exhibits antitumor effects and can increase the sensitivity of radiotherapy and chemotherapy in many types of cancers. MiR-8485, a discovered miRNA, has been shown to be involved in the occurrence and development of tumors. The purpose of this study was to investigate the effect of BJOE on the regulation of mammalian rapamycin target protein (mTOR) autophagy signal pathway and related autophagy factors on ovarian cancer cells through miR-8485. Methods: The main chemical constituents of BJOE were determined by UHPLC-MS/MS. Detection of miR-8485 expression in ovarian cancer cells treated with BJOE by quantitative reverse transcription polymerase chain reaction (qRT-PCR). CCK8 experiment and flow cytometry were used to observe the effects of BJOE and overexpression of miR-8485 on cell proliferation and apoptosis. Then, monodansylcadaverine (MDC) fluorescence staining was used to observe the changes of autophagy vesicles before and after the effect of BJOE and overexpressed miR-8485 on cancer cells. Next, the binding sites between miR8485 and mammalian rapamycin target protein activator 3 (LAMTOR3) were detected by double luciferase reporter assay. Furthermore, qRT-PCR and Western blot experiments were used to explore the changes of autophagy-related factors LAMTOR3, mTOR and autophagy-related 13 (ATG13), and microtubule associated protein 1 light chain 3 beta (LC3-Ⅱ) after BJOE and overexpression of miR-8485, in addition to autophagy inhibitor (3-MA) for rescue experiment verification. Results: The qRT-PCR results showed that the expression of miR-8485 increased after BJOE treatment in the SKOV3 cell. The CCK8 assay and flow cytometry analysis revealed that both BJOE and miR-8485 overexpression inhibited the proliferation and promoted the apoptosis of the SKOV3 cell. MDC fluorescence staining showed that BJOE and miR-8485 overexpression led to a significant increase in autophagy vesicles in the SKOV3 cell. Double luciferase reporter assay confirmed the existence of binding sites between miR8485 and LAMTOR3. The results of qRT-PCR and Western blot showed that BJOE and overexpressed miR-8485 downregulated the expression of LAMTOR3 and mTOR and up-regulated the expression of ATG13 and LC3-Ⅱ. Conclusion: 1) MiR-8485 may be the key factor of BJOE in promoting autophagy and apoptosis and inhibiting cell proliferation of ovarian cancer cells; 2) BJOE may play an antitumor role by regulating LAMTOR3/mTOR/ATG13 signaling axis through miR-8485 to promote autophagy in ovarian cancer cells.
Collapse
Affiliation(s)
- Yihan Wang
- The Second Clinical College, Hainan Medical University, Haikou, China
| | - Bocen Chen
- Key Laboratory of Molecular Biology, School of Basic Medicine and Sciences, Hainan Medical University, Haikou, China
| | - Man Xiao
- Key Laboratory of Molecular Biology, School of Basic Medicine and Sciences, Hainan Medical University, Haikou, China
| | - Xiaoli Wang
- Hainan Women and Children’s Medical Center, Haikou, China
| | - Yunhua Peng
- The First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Yunhua Peng,
| |
Collapse
|
183
|
Fan H, Hao X, Gao Y, Yang J, Liu A, Su Y, Xia Y. Nodosin Exerts an Anti-Colorectal Cancer Effect by Inhibiting Proliferation and Triggering Complex Cell Death in Vitro and in Vivo. Front Pharmacol 2022; 13:943272. [PMID: 35935881 PMCID: PMC9353177 DOI: 10.3389/fphar.2022.943272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common digestive system cancer in the world. Its incidence and mortality are increasing annually. Presently, CRC lacks long-term effective treatment methods and drugs. Therefore, finding new treatment methods and drugs is of great significance for CRC treatment. Compounds derived from natural plants have been widely used in tumor research and treatment because of their good antitumor activity these years. This study found that nodosin, a diterpenoid extracted from the medicinal plant Rabdosia serra (Maxim.) Hara, inhibited the growth of CRC cells SW480, HT-29 and LoVo in a dose- and time-dependent manner, with inhibitory concentrations (IC50) of 7.4, 7.7, and 6.6 μM respectively. We selected highly metastatic and poorly differentiated SW480 cells for further studies. We found that nodosin could inhibit cell proliferation by inhibiting DNA synthesis and induce cell death by inducing oxidative stress, apoptosis and autophagy in cells. Through in vitro assays combined with transcriptomic analysis, it was found that nodosin could downregulate tribbles pseudokinase 3 and upregulate oxidative stress-induced growth inhibitor 1 to induce oxidative stress in cells; nodosin-induced reactive oxygen species were able to upregulate the expression of heme oxygenase 1 to induce apoptosis and the expression of cathepsin L. and light chain-3 to induce autophagy. In vivo, we found that nodosin inhibited tumor growth and induced cells to undergo apoptosis and autophagy without significant toxic effects. In conclusion, our findings suggest that nodosin exerts anti-CRC effects mainly through its ability to induce apoptosis and autophagy in vitro and in vivo. Therefore, our study contributes to the development of nodosin-based potential CRC therapeutic drugs.
Collapse
Affiliation(s)
- Huixia Fan
- School of Life Sciences, Henan University, Kaifeng, China
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
- School of Pharmacy, Henan University, Kaifeng, China
| | - Xiaopeng Hao
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Yuan Gao
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Jian Yang
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijng, China
| | - Aojun Liu
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Yarui Su
- School of Life Sciences, Henan University, Kaifeng, China
- *Correspondence: Yarui Su, ; Yong Xia,
| | - Yong Xia
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
- *Correspondence: Yarui Su, ; Yong Xia,
| |
Collapse
|
184
|
Zhao H, Yang Y, Si X, Liu H, Wang H. The Role of Pyroptosis and Autophagy in Ischemia Reperfusion Injury. Biomolecules 2022; 12:biom12071010. [PMID: 35883566 PMCID: PMC9313059 DOI: 10.3390/biom12071010] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 02/07/2023] Open
Abstract
Pyroptosis is a process of programmed cell death mediated by gasdermin (GSDM) found in recent years. In the process of pyroptosis, caspase-1 or caspase-11/4/5 is activated, which cleaves gasdermin D and separates its N-terminal pore-forming domain (PFD). The oligomers of PFD bind to the cell membrane and form macropores on the membrane, resulting in cell swelling and membrane rupture. Increasing evidence indicates that pyroptosis is involved in many diseases, including ischemia reperfusion injury. Autophagy is a highly conserved catabolic process in eukaryotic cells. It plays an important role in the survival and maintenance of cells by degrading organelles, proteins, and macromolecules in the cytoplasm and recycling degradation products. Increasing evidence shows that dysfunctional autophagy participates in many diseases. Recently, autophagy and pyroptosis have been reported to play a vital role in the process of ischemia/reperfusion injury, but the related mechanisms are not completely clear. Therefore, this article reviews the role of autophagy and pyroptosis in ischemia–reperfusion injury and analyzes the related mechanisms to provide a basis for future research.
Collapse
Affiliation(s)
- Huijie Zhao
- Institute of Chronic Disease Risks Assessment, Henan University, Jinming Avenue, Kaifeng 475004, China;
| | - Yihan Yang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
| | - Xinya Si
- School of Stomatology, Henan University, Kaifeng 475004, China;
| | - Huiyang Liu
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
| | - Honggang Wang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
- Correspondence:
| |
Collapse
|
185
|
Lin H, Wang Y, Wang P, Long F, Wang T. Mutual regulation between N6-methyladenosine (m6A) modification and circular RNAs in cancer: impacts on therapeutic resistance. Mol Cancer 2022; 21:148. [PMID: 35843942 PMCID: PMC9290271 DOI: 10.1186/s12943-022-01620-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/08/2022] [Indexed: 02/08/2023] Open
Abstract
The resistance of tumor cells to therapy severely impairs the efficacy of treatment, leading to recurrence and metastasis of various cancers. Clarifying the underlying mechanisms of therapeutic resistance may provide new strategies for overcoming cancer resistance. N6-methyladenosine (m6A) is the most prevalent RNA modification in eukaryotes, and is involved in the regulation of RNA splicing, translation, transport, degradation, stability and processing, thus affecting several physiological processes and cancer progression. As a novel type of multifunctional non-coding RNAs (ncRNAs), circular RNAs (circRNAs) have been demonstrated to play vital roles in anticancer therapy. Currently, accumulating studies have revealed the mutual regulation of m6A modification and circRNAs, and their interaction can further influence the sensitivity of cancer treatment. In this review, we mainly summarized the recent advances of m6A modification and circRNAs in the modulation of cancer therapeutic resistance, as well as their interplay and potential mechanisms, providing promising insights and future directions in reversal of therapeutic resistance in cancer.
Collapse
Affiliation(s)
- Hong Lin
- Department of Pharmacy, Sichuan Cancer Hospital & Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pinghan Wang
- Laboratory Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China.
| | - Ting Wang
- Department of Pharmacy, Sichuan Cancer Hospital & Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
186
|
Chopra H, Bibi S, Goyal R, Gautam RK, Trivedi R, Upadhyay TK, Mujahid MH, Shah MA, Haris M, Khot KB, Gopan G, Singh I, Kim JK, Jose J, Abdel-Daim MM, Alhumaydhi FA, Emran TB, Kim B. Chemopreventive Potential of Dietary Nanonutraceuticals for Prostate Cancer: An Extensive Review. Front Oncol 2022; 12:925379. [PMID: 35903701 PMCID: PMC9315356 DOI: 10.3389/fonc.2022.925379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022] Open
Abstract
There are more than two hundred fifty different types of cancers, that are diagnosed around the world. Prostate cancer is one of the suspicious type of cancer spreading very fast around the world, it is reported that in 2018, 29430 patients died of prostate cancer in the United State of America (USA), and hence it is expected that one out of nine men diagnosed with this severe disease during their lives. Medical science has identified cancer at several stages and indicated genes mutations involved in the cancer cell progressions. Genetic implications have been studied extensively in cancer cell growth. So most efficacious drug for prostate cancer is highly required just like other severe diseases for men. So nutraceutical companies are playing major role to manage cancer disease by the recommendation of best natural products around the world, most of these natural products are isolated from plant and mushrooms because they contain several chemoprotective agents, which could reduce the chances of development of cancer and protect the cells for further progression. Some nutraceutical supplements might activate the cytotoxic chemotherapeutic effects by the mechanism of cell cycle arrest, cell differentiation procedures and changes in the redox states, but in other, it also elevate the levels of effectiveness of chemotherapeutic mechanism and in results, cancer cell becomes less reactive to chemotherapy. In this review, we have highlighted the prostate cancer and importance of nutraceuticals for the control and management of prostate cancer, and the significance of nutraceuticals to cancer patients during chemotherapy.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-milat University, Islamabad, Pakistan
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, China
| | - Rajat Goyal
- Maharishi Markandeshwar (MM) School of Pharmacy, Maharishi Markandeshwar University, Sadopur-Ambala, India
- Maharishi Markandeshwar (MM) College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Rupesh K. Gautam
- Maharishi Markandeshwar (MM) School of Pharmacy, Maharishi Markandeshwar University, Sadopur-Ambala, India
| | - Rashmi Trivedi
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | - Mohd Hasan Mujahid
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | | | - Muhammad Haris
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Kartik Bhairu Khot
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Gopika Gopan
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Jin Kyu Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jobin Jose
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
187
|
Konoshenko M, Lansukhay Y, Krasilnikov S, Laktionov P. MicroRNAs as Predictors of Lung-Cancer Resistance and Sensitivity to Cisplatin. Int J Mol Sci 2022; 23:7594. [PMID: 35886942 PMCID: PMC9321818 DOI: 10.3390/ijms23147594] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Platinum-based chemotherapy, cisplatin (DDP) specifically, is the main strategy for treating lung cancer (LC). However, currently, there is a lack of predictive drug-resistance markers, and there is increased interest in the development of a reliable and sensitive panels of markers for DDP chemotherapy-effectiveness prediction. MicroRNAs represent a perspective pool of markers for chemotherapy effectiveness. OBJECTIVES Data on miRNAs associated with LC DDP chemotherapy response are summarized and analyzed. MATERIALS AND METHODS A comprehensive review of the data in the literature and an analysis of bioinformatics resources were performed. The gene targets of miRNAs, as well as their reciprocal relationships with miRNAs, were studied using several databases. RESULTS AND DISCUSSION The complex analysis of bioinformatics resources and the literature indicated that the expressions of 12 miRNAs have a high predictive potential for LC DDP chemotherapy responses. The obtained information was discussed from the point of view of the main mechanisms of LC chemoresistance. CONCLUSIONS An overview of the published data and bioinformatics resources, with respect to the predictive microRNA markers of chemotherapy response, is presented in this review. The selected microRNAs and gene panel have a high potential for predicting LC DDP sensitiveness or DDP resistance as well as for the development of a DDP co-therapy.
Collapse
Affiliation(s)
- Maria Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Yuriy Lansukhay
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Sergey Krasilnikov
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Pavel Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| |
Collapse
|
188
|
Zhang B, Li D, Wang R. Transcriptome Profiling of N7-Methylguanosine Modification of Messenger RNA in Drug-Resistant Acute Myeloid Leukemia. Front Oncol 2022; 12:926296. [PMID: 35865472 PMCID: PMC9294171 DOI: 10.3389/fonc.2022.926296] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological tumor caused by the malignant transformation of myeloid progenitor cells. Although intensive chemotherapy leads to an initial therapeutic response, relapse due to drug resistance remains a significant challenge. In recent years, accumulating evidence has suggested that post-transcriptional methylation modifications are strongly associated with tumorigenesis. However, the mRNA profile of m7G modification in AML and its role in drug-resistant AML are unknown. In this study, we used MeRIP-seq technology to establish the first transcriptome-wide m7G methylome profile for AML and drug-resistant AML cells, and differences in m7G between the two groups were analyzed. In addition, bioinformatics analysis was conducted to explore the function of m7G-specific methylated transcripts. We found significant differences in m7G mRNA modification between AML and drug-resistant AML cells. Furthermore, bioinformatics analysis revealed that differential m7G-modified mRNAs were associated with a wide range of cellular functions. Importantly, down-methylated m7G modification was significantly enriched in ABC transporter-related mRNAs, which are widely recognized to play a key role in multidrug resistance. Our results provide new insights into a novel function of m7G methylation in drug resistance progression of AML.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Shandong, China
| | - Dong Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Shandong, China
| | - Ran Wang
- Department of Hematology, Qilu Hospital of Shandong University, Shandong, China
- *Correspondence: Ran Wang,
| |
Collapse
|
189
|
Po WW, Choi WS, Khing TM, Lee JY, Lee JH, Bang JS, Min YS, Jeong JH, Sohn UD. Benzyl Isothiocyanate-Induced Cytotoxicity via the Inhibition of Autophagy and Lysosomal Function in AGS Cells. Biomol Ther (Seoul) 2022; 30:348-359. [PMID: 35768332 PMCID: PMC9252883 DOI: 10.4062/biomolther.2022.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/05/2022] Open
Abstract
Gastric adenocarcinoma is among the top causes of cancer-related death and is one of the most commonly diagnosed carcinomas worldwide. Benzyl isothiocyanate (BITC) has been reported to inhibit the gastric cancer metastasis. In our previous study, BITC induced apoptosis in AGS cells. The purpose of the present study was to investigate the effect of BITC on autophagy mechanism in AGS cells. First, the AGS cells were treated with 5, 10, or 15 μM BITC for 24 h, followed by an analysis of the autophagy mechanism. The expression level of autophagy proteins involved in different steps of autophagy, such as LC3B, p62/SQSTM1, Atg5-Atg12, Beclin1, p-mTOR/mTOR ratio, and class III PI3K was measured in the BITC-treated cells. Lysosomal function was investigated using cathepsin activity and Bafilomycin A1, an autophagy degradation stage inhibitor. Methods including qPCR, western blotting, and immunocytochemistry were employed to detect the protein expression levels. Acridine orange staining and omnicathepsin assay were conducted to analyze the lysosomal function. siRNA transfection was performed to knock down the LC3B gene. BITC reduced the level of autophagy protein such as Beclin 1, class III PI3K, and Atg5-Atg12. BITC also induced lysosomal dysfunction which was shown as reducing cathepsin activity, protein level of cathepsin, and enlargement of acidic vesicle. Overall, the results showed that the BITC-induced AGS cell death mechanism also comprises the inhibition of the cytoprotective autophagy at both initiation and degradation steps.
Collapse
Affiliation(s)
- Wah Wah Po
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Won Seok Choi
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Tin Myo Khing
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Ji-Yun Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jong Hyuk Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Joon Seok Bang
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Young Sil Min
- Department of Pharmaceutical Science, Jungwon University, Goesan 28024, Republic of Korea
| | - Ji Hoon Jeong
- College of Medicine, Chung-Ang University, and Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, Republic of Korea
| | - Uy Dong Sohn
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
190
|
Mahinfar P, Mansoori B, Rostamzadeh D, Baradaran B, Cho WC, Mansoori B. The Role of microRNAs in Multidrug Resistance of Glioblastoma. Cancers (Basel) 2022; 14:3217. [PMID: 35804989 PMCID: PMC9265057 DOI: 10.3390/cancers14133217] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor that develops from neuroglial stem cells and represents a highly heterogeneous group of neoplasms. These tumors are predominantly correlated with a dismal prognosis and poor quality of life. In spite of major advances in developing novel and effective therapeutic strategies for patients with glioblastoma, multidrug resistance (MDR) is considered to be the major reason for treatment failure. Several mechanisms contribute to MDR in GBM, including upregulation of MDR transporters, alterations in the metabolism of drugs, dysregulation of apoptosis, defects in DNA repair, cancer stem cells, and epithelial-mesenchymal transition. MicroRNAs (miRNAs) are a large class of endogenous RNAs that participate in various cell events, including the mechanisms causing MDR in glioblastoma. In this review, we discuss the role of miRNAs in the regulation of the underlying mechanisms in MDR glioblastoma which will open up new avenues of inquiry for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Parvaneh Mahinfar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - Behnaz Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 175-14115, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran;
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Behzad Mansoori
- The Wistar Institute, Molecular & Cellular Oncogenesis Program, Philadelphia, PA 19104, USA
| |
Collapse
|
191
|
Wang F, Xie D, Lai W, Zhou M, Wang J, Xu R, Huang J, Zhang R, Li G. Autophagy responsive intra-intercellular delivery nanoparticles for effective deep solid tumor penetration. J Nanobiotechnology 2022; 20:300. [PMID: 35752856 PMCID: PMC9233833 DOI: 10.1186/s12951-022-01514-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/14/2022] [Indexed: 11/23/2022] Open
Abstract
Deep tumor cells (cells in the center of solid tumors) play a crucial role in drug tolerance, metastasis, recurrence and microenvironment immune suppression. However, their deep location endows them with an untouched abdomen and makes them refractory to current treatments. Herein, we exploited the characteristic of higher autophagy in deep tumor cells than in superficial tumor cells and designed autophagy-responsive multifunctional nanoparticles (PGN) to enhance drug accumulation in deep tumor cells. PGNs were prepared by densely coating poly (lactic-co-glycolic acid) (PLGA) with cationic autophagy-responsive cell-penetrating peptide (GR9) and anionic 2,3-dimethylmaleic anhydride (DMA)-modified DSPE-PEG. The suitable nanoparticle size (122.4 nm) and charge-neutral surface (0.21 mV) of the NPs enabled long blood circulation. The hydrolysis of surface-anchored anionic DMA in the acidic microenvironment led to the exposure of the GR9 peptide and enhance tumor penetration. Once the PGN arrived in deep tumor cells with strong autophagy, GR9 was cut off by an autophagy shear enzyme, and the nanoparticles remained in the cells to undergo degradation. Furthermore, we prepared docetaxel (DTX) and chloroquine (CQ) loaded d-PGN. CQ inhibits autophagosome fusion with lysosomes, resulting in autophagosome accumulation, which further enhances the sensitivity of d-PGN to autophagy and their deep tumor retention. In vivo experiments showed that drug-loaded d-PGN achieved excellent antitumor efficacy with a peak inhibition rate of 82.1%. In conclusion, autophagy-responsive multifunctional nanoparticles provide a novel potential strategy for solid tumor treatment.
Collapse
Affiliation(s)
- Fengling Wang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Dandan Xie
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Min Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Jie Wang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Rufu Xu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Jingbing Huang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao Road, Chongqing, China.
| |
Collapse
|
192
|
Xu J, Shen R, Jiao Z, Chen W, Peng D, Wang L, Yu N, Peng C, Cai B, Song H, Chen F, Liu B. Current Advancements in Antitumor Properties and Mechanisms of Medicinal Components in Edible Mushrooms. Nutrients 2022; 14:nu14132622. [PMID: 35807802 PMCID: PMC9268676 DOI: 10.3390/nu14132622] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Edible and medicinal fungi, a group of eukaryotic organisms with numerous varieties, including Coriolus versicolor, Ganoderma lucidum, Cordyceps sinensis, Pleurotus ostreatus, and Grifola frondosa, have been demonstrated to possess a board range of pharmaceutical properties, including anti-virus, anti-inflammation, and neuroprotection. Moreover, edible and medicinal fungi have been traditionally consumed as food to provide multiple nutrients and as drugs owing to having the activities of invigorating blood circulation, reinforcing the healthy qi, clearing away heat, and eliminating stasis for thousands of years in China. Malignant tumors, well-known as the second leading cause of death globally, accounted for nearly 10 million deaths in 2020. Thus, in-depth exploration of strategies to prevent and treat cancer is extremely urgent. A variety of studies have reported that the main bioactive components of edible and medicinal fungi, mainly polysaccharides and triterpenoids, exhibit diverse anticancer activities via multiple mechanisms, including inhibition of cell proliferation and metastasis, induction of apoptosis and autophagy, reversing multidrug resistance, and regulation of immune responses, thus suggesting their substantial potential in the prevention and treatment of cancer. Our review summarizes the research progress on the anticancer properties of edible and medicinal fungi and the underlying molecular mechanism, which may offer a better understanding of this field. Additionally, few studies have reported the safety and efficacy of extracts from edible and medicinal fungi, which may limit their clinical application. In summary, there is a need to continue to explore the use of those extracts and to further validate their safety and efficacy.
Collapse
Affiliation(s)
- Jing Xu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (J.X.); (R.S.); (Z.J.); (B.C.)
| | - Rui Shen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (J.X.); (R.S.); (Z.J.); (B.C.)
| | - Zhuoya Jiao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (J.X.); (R.S.); (Z.J.); (B.C.)
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.C.); (D.P.); (L.W.); (N.Y.); (C.P.)
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.C.); (D.P.); (L.W.); (N.Y.); (C.P.)
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.C.); (D.P.); (L.W.); (N.Y.); (C.P.)
| | - Nianjun Yu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.C.); (D.P.); (L.W.); (N.Y.); (C.P.)
| | - Can Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.C.); (D.P.); (L.W.); (N.Y.); (C.P.)
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (J.X.); (R.S.); (Z.J.); (B.C.)
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (J.X.); (R.S.); (Z.J.); (B.C.)
- Correspondence: (B.L.); (H.S.); (F.C.)
| | - Fengyuan Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (J.X.); (R.S.); (Z.J.); (B.C.)
- Correspondence: (B.L.); (H.S.); (F.C.)
| | - Bin Liu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
- Correspondence: (B.L.); (H.S.); (F.C.)
| |
Collapse
|
193
|
Shen W, Luo P, Sun Y, Zhang W, Zhou N, Zhan H, Zhang Q, Shen J, Lin A, Cheng Q, Wang Q, Zhang J, Wang HH, Wei T. NRBF2 regulates the chemoresistance of small cell lung cancer by interacting with the P62 protein in the autophagy process. iScience 2022; 25:104471. [PMID: 35712081 PMCID: PMC9194155 DOI: 10.1016/j.isci.2022.104471] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/22/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022] Open
Abstract
Reversing chemotherapy resistance in small cell lung cancer (SCLC) is crucial to improve patient prognosis. The present study aims to investigate the underlying mechanisms in SCLC chemoresistance. We see that nuclear receptor binding factor 2 (NRBF2) is a poor prognostic factor in SCLC. The effects of NRBF2 on chemoresistance were determined in SCLC. The underlying molecular mechanisms of NRBF2 in the autophagy process in SCLC were examined. NRBF2 positively regulated autophagy, leading to drug resistance in SCLC. The MIT domain of NRBF2 directly interacted with the PB1 domain of P62. This interaction increased autophagic P62 body formation, revealing the regulatory role of NRBF2 in autophagy. Notably, NRBF2 was directly modulated by the transcription factor XRCC6. The MIT domain of NRBF2 interacts with the PB1 domain of P62 to regulate the autophagy process, resulting in SCLC chemoresistance. NRBF2 is likely a useful chemotherapy response marker and therapeutic target in SCLC. NRBF2 promoted the chemoresistance of SCLC in vitro and in vivo The chemoresistance induced by NRBF2 was mediated via autophagy in SCLC NRBF2 interacting with P62 contributed to autophagic P62 bodies' formation NRBF2 was regulated by XRCC6 via direct binding to the NRBF2 gene promoter
Collapse
Affiliation(s)
- Weitao Shen
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Yueqin Sun
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Wei Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Ningning Zhou
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510282, Guangdong, People's Republic of China
| | - Hongrui Zhan
- Department of Rehabilitation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, People's Republic of China
| | - Qingxi Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Jie Shen
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Qiongyao Wang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| | - Hai-Hong Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, Guangdong, People's Republic of China
| |
Collapse
|
194
|
miR-214 Modulates the Growth and Migration of Oral Cancer before and after Chemotherapy through Mediating ULK1. J Immunol Res 2022; 2022:4589182. [PMID: 35692501 PMCID: PMC9184158 DOI: 10.1155/2022/4589182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022] Open
Abstract
The role of miRNAs as crucial components in carcinogenesis has been well documented. However, whether and how miR-214 influences oral cancer cells' drug resistance remains to be elucidated, and its downstream targets are still under investigation. Hence, this research is aimed at determining miR-214 and ULK1 expression in oral cancer before and after chemotherapy and their correlations with cancer cell growth. Human oral normal epithelial cells and human tongue squamous cell carcinoma CAL-27 cells were cultured to detect miR-214 and ULK1 levels. It was found that before chemotherapy, miR-214 was higher, while ULK1 was underexpressed in CAL-27 cells, versus normal epithelial cells. After chemotherapy, miR-214 decreased obviously in CAL-27 cells, while ULK1 level increased significantly. In addition, autophagy-related genes (Beclin 1, mTOR, and P53) in CAL-27 cells were found to be significantly inhibited before chemotherapy and were obviously increased after chemotherapy. Moreover, to further determine the impacts of miR-214 and ULK1 on oral cancer cell growth after chemotherapy, the two were overexpressed or silenced in CAL-27 cells after transfection. We found that ULK1 could effectively decrease the activity and invasion of CAL-27 cells and increase their apoptosis level, while miR-214 could antagonize its antitumor effect. Therefore, miR-214 can be used as an early prognostic biomarker for oral cancer, and ULK1 is a new candidate therapeutic target.
Collapse
|
195
|
Chiappa M, Petrella S, Damia G, Broggini M, Guffanti F, Ricci F. Present and Future Perspective on PLK1 Inhibition in Cancer Treatment. Front Oncol 2022; 12:903016. [PMID: 35719948 PMCID: PMC9201472 DOI: 10.3389/fonc.2022.903016] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Polo-like kinase 1 (PLK1) is the principle member of the well conserved serine/threonine kinase family. PLK1 has a key role in the progression of mitosis and recent evidence suggest its important involvement in regulating the G2/M checkpoint, in DNA damage and replication stress response, and in cell death pathways. PLK1 expression is tightly spatially and temporally regulated to ensure its nuclear activation at the late S-phase, until the peak of expression at the G2/M-phase. Recently, new roles of PLK1 have been reported in literature on its implication in the regulation of inflammation and immunological responses. All these biological processes are altered in tumors and, considering that PLK1 is often found overexpressed in several tumor types, its targeting has emerged as a promising anti-cancer therapeutic strategy. In this review, we will summarize the evidence suggesting the role of PLK1 in response to DNA damage, including DNA repair, cell cycle progression, epithelial to mesenchymal transition, cell death pathways and cancer-related immunity. An update of PLK1 inhibitors currently investigated in preclinical and clinical studies, in monotherapy and in combination with existing chemotherapeutic drugs and targeted therapies will be discussed.
Collapse
Affiliation(s)
- Michela Chiappa
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Serena Petrella
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Giovanna Damia
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Federica Guffanti
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Francesca Ricci
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| |
Collapse
|
196
|
Liu K, Huang J, Liu J, Klionsky DJ, Kang R, Tang D. Induction of autophagy-dependent ferroptosis to eliminate drug-tolerant human retinoblastoma cells. Cell Death Dis 2022; 13:521. [PMID: 35654783 PMCID: PMC9163041 DOI: 10.1038/s41419-022-04974-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 01/21/2023]
Abstract
Carboplatin is the most used first-line drug for the treatment of human retinoblastoma (RB), a rare form of cancer in infancy and childhood. However, the clinical application of carboplatin is restricted due to the emergence of acquired multi-drug resistance (MDR) after long-term treatment. Here, we report a new strategy to eliminate MDR RB cells by inducing autophagy-dependent ferroptosis. Compared with parent cells, carboplatin-resistant human RB cells have higher autophagy activity, which drives the formation of MDR to other chemotherapeutic drugs (e.g., etoposide and vincristine). In addition to confirming the traditional strategy of inhibiting autophagy to overcome MDR, we also establish an approach of inducing selective ferritinophagy to eliminate drug-resistant cells. We evaluate the effectiveness and safety of 4-octyl itaconate, a cell-permeable derivative of the metabolite itaconate, in inducing ferritinophagy-dependent ferroptosis in the treatment of MDR RB cells in vitro and in xenograft mouse models. These findings may provide essential clues for initiating clinical trials that target autophagy-dependent ferroptosis to kill drug-tolerant persistent cells during RB therapy.
Collapse
Affiliation(s)
- Ke Liu
- grid.216417.70000 0001 0379 7164Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Huang
- grid.216417.70000 0001 0379 7164Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Liu
- grid.410737.60000 0000 8653 1072DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Daniel J. Klionsky
- grid.214458.e0000000086837370Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Rui Kang
- grid.267313.20000 0000 9482 7121Department of Surgery, UT Southwestern Medical Center, Dallas, TX USA
| | - Daolin Tang
- grid.267313.20000 0000 9482 7121Department of Surgery, UT Southwestern Medical Center, Dallas, TX USA
| |
Collapse
|
197
|
Zhang H, Zhang L, Fan YT, Li TN, Peng LS, Wang KP, Ma J. Signature Based on Six Autophagy-related Genes to Predict Prognosis of Head and Neck Squamous Cell Carcinoma. Curr Med Sci 2022; 42:597-605. [DOI: 10.1007/s11596-022-2560-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/28/2021] [Indexed: 11/28/2022]
|
198
|
Exosomal CTCF Confers Cisplatin Resistance in Osteosarcoma by Promoting Autophagy via the IGF2-AS/miR-579-3p/MSH6 Axis. JOURNAL OF ONCOLOGY 2022; 2022:9390611. [PMID: 35693981 PMCID: PMC9175095 DOI: 10.1155/2022/9390611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/21/2022] [Indexed: 11/18/2022]
Abstract
Cancer-derived exosomes participate in carcinogenesis and progression of cancers, including metastasis and drug-resistance. Of note, CTCF has been suggested to induce drug resistance in various cancers. Herein, we aim to investigate the role of cisplatin- (CDDP-) resistant osteosarcoma- (OS-) derived exosomal CTCF in OS cell resistance to CDDP and its mechanistic basis. Differentially expressed transcription factors, long noncoding RNAs (lncRNAs), miRNAs, and genes in OS were retrieved using bioinformatics approaches. Exosomes were extracted from CDDP-resistant OS cells and then cocultured with parental OS cells, followed by lentiviral transduction to manipulate the expression of CTCF, IGF2-AS, miR-579-3p, and MSH6. We assessed the in vitro and in vivo effects on malignant phenotypes, autophagy, CDDP sensitivity, and tumor formation of OS cells. It was established that CTCF and IGF2-AS were highly expressed in CDDP-resistant OS cells, and the CDDP-resistant OS cell-derived exosomal CTCF enhanced IGF2-AS transcription. CDDP-resistant OS-derived exosomes transmitted CTCF to OS cells and increased CDDP resistance in OS cells by activating an autophagy-dependent pathway. Mechanistically, CTCF activated IGF2-AS transcription and IGF2-AS competitively bound to miR-579-3p to upregulate MSH6 expression. Additionally, the promoting function of exosomal CTCF-mediated IGF2-AS/miR-579-3p/MSH6 in OS cell resistance to CDDP was confirmed in vivo. Taken together, CDDP-resistant OS-derived exosomal CTCF enhanced resistance of OS cells to CDDP via activating the autophagy-dependent pathway, providing a potential therapeutic consideration for OS treatment.
Collapse
|
199
|
SIRT1 regulates mitotic catastrophe via autophagy and BubR1 signaling. Mol Cell Biochem 2022; 477:2787-2799. [PMID: 35639235 DOI: 10.1007/s11010-022-04470-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/04/2022] [Indexed: 10/18/2022]
Abstract
Mitotic catastrophe (MC) is a suppressive mechanism that mediates the elimination of mitosis-deficient cells through apoptosis, necrosis or senescence after M phase block. SIRT1 is involved in the regulation of several cellular processes, including autophagy. However, the relationship between SIRT1 and MC has been largely obscure. Our study highlights that SIRT1 might be involved in the regulation of MC. We have shown that degradation of the SIRT1 protein via proteasome and lysosomal pathway was accompanied by MC induced via BMH-21. Overexpression of SIRT1 alleviated MC by decreasing the proportion of apoptotic and multinuclear cells induced by G2/M block and triggered autophagy whereas knockdown of SIRT1 aggravated MC and repressed autophagy. Furthermore, we found that serum starvation triggered autophagy evidently generated lower MC whereas siRNA of ATG5/7 suppressed autophagy leading to higher MC. ChIP analysis revealed that SIRT1 could bind to the promoter of BubR1, a component of spindle assembly checkpoint (SAC), to upregulate its expression. Overexpression of BubR1 decreased MC whereas knockdown of BubR1 increased it. These results reveal that SIRT1 regulates MC through autophagy and BubR1 signaling, and provide evidence for SIRT1, autophagy and BubR1 being the potential cancer therapeutic targets.
Collapse
|
200
|
Chen Y, Liu H, Zheng X, Wang J, Wang L, Yang J. SIRT1 ameliorates renal ischemia-reperfusion injury through suppressing endoplasmic reticulum stress-mediated autophagy. Am J Transl Res 2022; 14:3419-3429. [PMID: 35702070 PMCID: PMC9185025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 04/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Renal ischemia-reperfusion (IR) injury is a therapeutic challenge for surgeons. Sirtuin 1 (SIRT1) is an NAD+-dependent deacetylase that plays a vital role in modulating cellular senescence and aging. In this study, we determined whether SIRT1 upregulation could alleviate renal IR injury and the underlying mechanism. METHODS A renal IR model was induced in male C57BL/6 mice. Blood urea nitrogen and serum creatinine were evaluated as markers of kidney function, and renal injury was assessed by pathological examination. The inflammatory milieu was analyzed by real-time RT-PCR and myeloperoxidase immunofluorescence assays. Western blotting was used to quantify SIRT1 protein expression, endoplasmic reticulum stress, and autophagy. RESULTS SIRT1 was upregulated in renal tissue after IR. Blood analysis and histopathologic examination demonstrated that SIRT1 preserved renal function and reduced renal damage. Further evaluation illustrated that IR induced autophagy and endoplasmic reticulum stress, while SIRT1 upregulation reduced endoplasmic reticulum stress-mediated autophagy levels. CONCLUSIONS SIRT1 upregulation protects the kidney against IR-induced injury by inhibiting endoplasmic reticulum stress-mediated autophagy.
Collapse
Affiliation(s)
- Yu Chen
- Department of Organ Transplantation, Shanghai Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Hao Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of MedicineShanghai, China
| | - Xueyang Zheng
- Department of Organ Transplantation, Shanghai Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Jiyuan Wang
- Department of Organ Transplantation, Shanghai Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Liming Wang
- Department of Organ Transplantation, Shanghai Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Jinghui Yang
- Department of Organ Transplantation, Shanghai Changzheng Hospital, Navy Medical UniversityShanghai, China
| |
Collapse
|