1
|
Liu X, Zhang R, Liu L, Zhi S, Feng X, Shen Y, Wang L, Zhang Q, Chen Y, Hao J. Sohlh2 Promotes the Progression of Hepatocellular Carcinoma via TGM2-Mediated Autophagy. Mol Carcinog 2025; 64:138-151. [PMID: 39436118 DOI: 10.1002/mc.23832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/02/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
Liver cancer is the third leading cause of cancer-related deaths worldwide, with hepatocellular carcinoma (HCC) accounting for 85% of liver cancer-related deaths. Autophagy controls HCC cell growth, invasion, metastasis, drug resistance, and stemness. Spermatogenesis and oogenesis basic helix-loop-helix transcription factor 2 (Sohlh2) can bind to the E-boxes in the promoter regions of target genes, which are involved in multiple neoplasms. In this study, Sohlh2 was highly expressed in HCC tissues and was related to poor prognosis. Moreover, Sohlh2 overexpression promoted the proliferation, migration, invasion, and metastasis of HCC cells in vivo and in vitro. However, Sohlh2 silencing inhibited proliferation, migration, invasion, and metastasis of HCC cells in vivo and in vitro. Mechanistically, Sohlh2 could bind to the promoter of TGM2 and enhance its transcriptional activity, thereby enhancing the autophagy of HCC cells. Furthermore, Sohlh2 protein levels were positively associated with TGM2 expression in HCC tissues. Taken together, these results demonstrate that Sohlh2 can promote HCC progression via TGM2-mediated autophagy, implying that Sohlh2 is a promising candidate for HCC treatment.
Collapse
Affiliation(s)
- Xuyue Liu
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ruihong Zhang
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lanlan Liu
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Sujuan Zhi
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaoning Feng
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ying Shen
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Liyan Wang
- Research Center for Medical and Structural Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qi Zhang
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yanru Chen
- Liver Transplantation Center, Clinical Research Center for Pediatric Liver Transplantation, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jing Hao
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
2
|
Huang G, Zhao X, Bai Y, Liu J, Li W, Wu Y. Regulation of mitochondrial autophagy by lncRNA MALAT1 in sepsis-induced myocardial injury. Eur J Med Res 2024; 29:524. [PMID: 39487520 PMCID: PMC11531147 DOI: 10.1186/s40001-024-02098-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/08/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Sepsis-induced myocardial injury (SIMI) is a severe complication of sepsis, contributing significantly to mortality. Mitochondrial dysfunction and dysregulated autophagy are implicated in SIMI pathogenesis. Long non-coding RNA MALAT1 has been associated with various diseases, including sepsis, but its role in SIMI remains unclear. OBJECTIVE This study aimed to investigate the role of lncRNA MALAT1 in SIMI, specifically in the regulation of mitochondrial autophagy. METHODS A sepsis-induced cardiomyopathy model was established in mice, and the cardiac tissues were analyzed. The expression of lncRNA MALAT1 was modulated and its effects on mitochondrial autophagy, myocardial injury, inflammation, and apoptosis were assessed. Furthermore, the interaction between MALAT1 and miR-146a was explored, as well as the involvement of the TLR4/NF-kB/MAPK signaling pathway. RESULTS Activation of mitochondrial autophagy by urolithin A (UA) alleviated SIMI, inflammation, and cardiac dysfunction. Downregulation of MALAT1 enhanced mitochondrial autophagy, stabilized the mitochondrial membrane potential, and inhibited mitochondrial reactive oxygen species (ROS) production, leading to improved cell viability and reduced myocardial injury. Furthermore, MALAT1 interacted with miR-146a, and their modulation influenced mitochondrial autophagy, myocardial injury, and inflammation. The TLR4/NF-kB/MAPK signaling pathway was implicated in these processes. CONCLUSION Our findings suggest that lncRNA MALAT1 plays a crucial role in SIMI by modulating miR-146a-mediated mitochondrial autophagy and the TLR4/NF-kB/MAPK signaling pathway. These results provide new insights into the pathogenesis of SIMI and potential therapeutic targets.
Collapse
Affiliation(s)
- Guangqing Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing, 100029, China
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xu Zhao
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yong Bai
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jie Liu
- Maternal and Child Health Care Hospital Affiliated to Hubei University of Medicine, Shiyan, Hubei, China
| | - Wei Li
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yongquan Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
3
|
Manuja A, Kumar B, Chhabra D, Brar B, Riyesh T, Pal Y, Bhattacharya TK, Prasad M. Antiviral and Cytoprotective Effect of Zinc (Yasad Bhasma) Based Nanoformulations Against Bovine Coronavirus. Indian J Microbiol 2024; 64:1123-1131. [PMID: 39282188 PMCID: PMC11399353 DOI: 10.1007/s12088-024-01255-0] [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: 11/23/2023] [Accepted: 03/04/2024] [Indexed: 09/18/2024] Open
Abstract
Ayurvedic medicine utilizes metal-based preparations, known as bhasmas, to treat various health conditions. Yasad bhasma (YB), a zinc-based ayurvedic preparation, shows promise as a potential candidate for developing zinc-based nanomedicines with anti-inflammatory and antioxidant properties. In this study, we synthesized a formulation combining YB and hydroxychloroquine (HC) as a zinc ionophore (YBHC) and investigated its biocompatibility and antiviral effects against buffalo calf coronavirus (BCoV) in Vero cells. Our results demonstrated that the formulation exhibited good conformity and enhanced cell proliferation compared to untreated cells. Additionally, no cytopathic effects were observed in BCoV-infected Vero cells treated with YBHC and YB, while infected control cells exhibited cytopathic effects. YB showed cytoprotection by promoting epithelial tissue turnover. We further explored whether YB/YBHC exerted a lysosomotropic effect to produce antiviral effects on coronavirus-adapted Vero cells, but no cell internalization was observed. In addition to the synergistic antiviral effect of YB and HC, YB may play a vital role in rejuvenating affected tissues.
Collapse
Affiliation(s)
- Anju Manuja
- ICAR-National Research Centre On Equines, Hisar, Haryana 125001 India
| | - Balvinder Kumar
- ICAR-National Research Centre On Equines, Hisar, Haryana 125001 India
| | - Dharvi Chhabra
- ICAR-National Research Centre On Equines, Hisar, Haryana 125001 India
| | - Basanti Brar
- Lala Lajpat Rai, University of Veterinary and Animal Sciences, Hisar, India
| | - T Riyesh
- ICAR-National Research Centre On Equines, Hisar, Haryana 125001 India
| | - Yash Pal
- ICAR-National Research Centre On Equines, Hisar, Haryana 125001 India
| | - T K Bhattacharya
- ICAR-National Research Centre On Equines, Hisar, Haryana 125001 India
| | - Minakshi Prasad
- ICAR-National Research Centre On Equines, Hisar, Haryana 125001 India
- Lala Lajpat Rai, University of Veterinary and Animal Sciences, Hisar, India
| |
Collapse
|
4
|
Liu Y, Meng Y, Zhang J, Gu L, Shen S, Zhu Y, Wang J. Pharmacology Progresses and Applications of Chloroquine in Cancer Therapy. Int J Nanomedicine 2024; 19:6777-6809. [PMID: 38983131 PMCID: PMC11232884 DOI: 10.2147/ijn.s458910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/07/2024] [Indexed: 07/11/2024] Open
Abstract
Chloroquine is a common antimalarial drug and is listed in the World Health Organization Standard List of Essential Medicines because of its safety, low cost and ease of use. Besides its antimalarial property, chloroquine also was used in anti-inflammatory and antivirus, especially in antitumor therapy. A mount of data showed that chloroquine mainly relied on autophagy inhibition to exert its antitumor effects. However, recently, more and more researches have revealed that chloroquine acts through other mechanisms that are autophagy-independent. Nevertheless, the current reviews lacked a comprehensive summary of the antitumor mechanism and combined pharmacotherapy of chloroquine. So here we focused on the antitumor properties of chloroquine, summarized the pharmacological mechanisms of antitumor progression of chloroquine dependent or independent of autophagy inhibition. Moreover, we also discussed the side effects and possible application developments of chloroquine. This review provided a more systematic and cutting-edge knowledge involved in the anti-tumor mechanisms and combined pharmacotherapy of chloroquine in hope of carrying out more in-depth exploration of chloroquine and obtaining more clinical applications.
Collapse
Affiliation(s)
- Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Liwei Gu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Shengnan Shen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
- Department of Pharmacological Sciences, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| |
Collapse
|
5
|
Zhou Y, Qiu T, Wang T, Yu B, Xia K, Guo J, Liu Y, Ma X, Zhang L, Zou J, Chen Z, Zhou J. Research progress on the role of mitochondria in the process of hepatic ischemia-reperfusion injury. Gastroenterol Rep (Oxf) 2024; 12:goae066. [PMID: 38912038 PMCID: PMC11193119 DOI: 10.1093/gastro/goae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/08/2023] [Accepted: 02/26/2024] [Indexed: 06/25/2024] Open
Abstract
During liver ischemia-reperfusion injury, existing mechanisms involved oxidative stress, calcium overload, and the activation of inflammatory responses involve mitochondrial injury. Mitochondrial autophagy, a process that maintains the normal physiological activity of mitochondria, promotes cellular metabolism, improves cellular function, and facilitates organelle renewal. Mitochondrial autophagy is involved in oxidative stress and apoptosis, of which the PINK1-Parkin pathway is a major regulatory pathway, and the deletion of PINK1 and Parkin increases mitochondrial damage, reactive oxygen species production, and inflammatory response, playing an important role in mitochondrial quality regulation. In addition, proper mitochondrial permeability translational cycle regulation can help maintain mitochondrial stability and mitigate hepatocyte death during ischemia-reperfusion injury. This mechanism is also closely related to oxidative stress, calcium overload, and the aforementioned autophagy pathway, all of which leads to the augmentation of the mitochondrial membrane permeability transition pore opening and cause apoptosis. Moreover, the release of mitochondrial DNA (mtDNA) due to oxidative stress further aggravates mitochondrial function impairment. Mitochondrial fission and fusion are non-negligible processes required to maintain the dynamic renewal of mitochondria and are essential to the dynamic stability of these organelles. The Bcl-2 protein family also plays an important regulatory role in the mitochondrial apoptosis signaling pathway. A series of complex mechanisms work together to cause hepatic ischemia-reperfusion injury (HIRI). This article reviews the role of mitochondria in HIRI, hoping to provide new therapeutic clues for alleviating HIRI in clinical practice.
Collapse
Affiliation(s)
- Yujie Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Bo Yu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Kang Xia
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Yiting Liu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Xiaoxiong Ma
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Long Zhang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Jilin Zou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Zhongbao Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| |
Collapse
|
6
|
Hong Z, Wang H, Zhang T, Xu L, Zhai Y, Zhang X, Zhang F, Zhang L. The HIF-1/ BNIP3 pathway mediates mitophagy to inhibit the pyroptosis of fibroblast-like synoviocytes in rheumatoid arthritis. Int Immunopharmacol 2024; 127:111378. [PMID: 38141408 DOI: 10.1016/j.intimp.2023.111378] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Synovial hypoxia, a critical pathological characteristic of rheumatoid arthritis (RA), significantly contributes to synovitis and synovial hyperplasia. In response to hypoxic conditions, fibroblast-like synoviocytes (FLS) undergo adaptive changes involving gene expression modulation, with hypoxia-inducible factors (HIF) playing a pivotal role. The regulation of BCL2/adenovirus e1B 19 kDa protein interacting protein 3 (BNIP3) and nucleotide-binding oligomerization segment-like receptor family 3 (NLRP3) expression has been demonstrated to be regulated by HIF-1. The objective of this study was to examine the molecular mechanism that contributes to the aberrant activation of FLS in response to hypoxia. Specifically, the interaction between BNIP3-mediated mitophagy and NLRP3-mediated pyroptosis was conjointly highlighted. METHODS The research methodology employed Western blot and immunohistochemistry techniques to identify the occurrence of mitophagy in synovial tissue affected by RA. Additionally, the levels of mitophagy under hypoxic conditions were assessed using Western blot, immunofluorescence, quantitative polymerase chain reaction (qPCR), and CUT&Tag assays. Pyroptosis was observed through electron microscopy, fluorescence microscopy, and Western blot analysis. Furthermore, the quantity of reactive oxygen species (ROS) was measured. The silencing of HIF-1α and BNIP3 was achieved through the transfection of short hairpin RNA (shRNA) into cells. RESULTS In the present study, a noteworthy increase in the expression of BNIP3 and LC3B was observed in the synovial tissue of patients with RA. Upon exposure to hypoxia, FLS of RA exhibited BNIP3-mediated mitophagy and NLRP3 inflammasome-mediated pyroptosis. It appears that hypoxia regulates the expression of BNIP3 and NLRP3 through the transcription factor HIF-1. Additionally, the activation of mitophagy has been observed to effectively inhibit hypoxia-induced pyroptosis by reducing the intracellular levels of ROS. CONCLUSION In summary, the activation of FLS in RA patients under hypoxic conditions involves both BNIP3-mediated mitophagy and NLRP3 inflammasome-mediated pyroptosis. Additionally, mitophagy can suppress hypoxia-induced FLS pyroptosis by eliminating ROS and inhibiting the HIF-1α/NLRP3 pathway.
Collapse
Affiliation(s)
- Zhongyang Hong
- Department of Pharmacy, Affiliated the Jianhu People's Hospital, Yancheng 224700, China; Central Laboratory, Affiliated the Jianhu People's Hospital, Yancheng 224700, China
| | - Han Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Tianjing Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Li Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Yuanfang Zhai
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Feng Zhang
- Department of Pharmacy, Affiliated the Fuyang Hospital of Anhui Medical University, Fuyang 236000, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
7
|
Reisenauer KN, Aroujo J, Tao Y, Ranganathan S, Romo D, Taube JH. Therapeutic vulnerabilities of cancer stem cells and effects of natural products. Nat Prod Rep 2023; 40:1432-1456. [PMID: 37103550 PMCID: PMC10524555 DOI: 10.1039/d3np00002h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Covering: 1995 to 2022Tumors possess both genetic and phenotypic heterogeneity leading to the survival of subpopulations post-treatment. The term cancer stem cells (CSCs) describes a subpopulation that is resistant to many types of chemotherapy and which also possess enhanced migratory and anchorage-independent growth capabilities. These cells are enriched in residual tumor material post-treatment and can serve as the seed for future tumor re-growth, at both primary and metastatic sites. Elimination of CSCs is a key goal in enhancing cancer treatment and may be aided by application of natural products in conjunction with conventional treatments. In this review, we highlight molecular features of CSCs and discuss synthesis, structure-activity relationships, derivatization, and effects of six natural products with anti-CSC activity.
Collapse
Affiliation(s)
| | - Jaquelin Aroujo
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | - Yongfeng Tao
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | | | - Daniel Romo
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | - Joseph H Taube
- Department of Biology, Baylor University, Waco, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
8
|
Huang T, Wang Y, Yu Z, Miao X, Jiang Z, Yu K, Fu M, Lai K, Wang Y, Yang G. Effect of mitophagy in the formation of osteomorphs derived from osteoclasts. iScience 2023; 26:106682. [PMID: 37250312 PMCID: PMC10214740 DOI: 10.1016/j.isci.2023.106682] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 01/27/2023] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Osteoclasts are specialized multinucleated giant cells with unique bone-destroying capacities. A recent study revealed that osteoclasts undergo an alternative cell fate by dividing into daughter cells called osteomorphs. To date, no studies have focused on the mechanisms of osteoclast fission. In this study, we analyzed the alternative cell fate process in vitro and, herein, reported the high expression of mitophagy-related proteins during osteoclast fission. Mitophagy was further confirmed by the colocalization of mitochondria with lysosomes, as observed in fluorescence images and transmission electron microscopy. We investigated the role played by mitophagy in osteoclast fission via drug stimulation experiments. The results showed that mitophagy promoted osteoclast division, and inhibition of mitophagy induced osteoclast apoptosis. In summary, this study reveals the role played by mitophagy as the decisive link in osteoclasts' fate, providing a new therapeutic target and perspective for the clinical treatment of osteoclast-related diseases.
Collapse
Affiliation(s)
- Tingben Huang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Yuchen Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Zhou Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Xiaoyan Miao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Ke Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Kaichen Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Endodontics, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310006, China
- Department of Implantology, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
9
|
Lobo CL, Shetty A, M M, Dubey A, El-Zahaby SA. Non-systemic Approaches for Ductal Carcinoma In Situ: Exploring the Potential of Ultra-flexible Combisomes as a Novel Drug Delivery Strategy-a Review. AAPS PharmSciTech 2023; 24:119. [PMID: 37173545 DOI: 10.1208/s12249-023-02574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Ductal carcinoma in situ (DCIS) is currently treated through breast-conserving surgery (lumpectomy), radiation therapy, breast-removing surgery (mastectomy), and hormone therapy to prevent further progression into invasive breast cancer and recurrence. Discrepancies concerning the prognosis of DCIS have sparked controversy about adequate treatment. Considering the severe medical and psychological consequences of mastectomy, developing a treatment approach that arrests the progression of DCIS to the invasive stage without affecting the non-cancerous cells is of utmost importance. In the current review, the problems associated with the diagnosis and management of DCIS have been thoroughly discussed. A summary of the route of administration and drug delivery systems to manage DCIS was also provoked. Innovative ultra-flexible combisomes were also proposed for the effective management of DCIS. Prevention is essential in managing the risk of DCIS and reducing the risk of progression to invasive breast cancer. While prevention is vital, it is not always possible to prevent DCIS, and in some cases, treatment may be necessary. Hence, this review recommends that ultra-flexible combisomes administered as a topical gel provide a non-systemic approach for managing DCIS and thus significantly minimize the side effects and costs associated with existing therapies.
Collapse
Affiliation(s)
- Cynthia Lizzie Lobo
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Amitha Shetty
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Manohar M
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India.
| | - Sally A El-Zahaby
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, King Salman International University, South Sinai, Egypt
| |
Collapse
|
10
|
Xiao X, Chung PED, Xu M, Hu A, Ju Y, Yang X, Song J, Song J, Wang C, Zacksenhaus E, Liu S, He Z, Ben-David Y. A racemosin B derivative, C25, suppresses breast cancer growth via lysosomal membrane permeabilization and inhibition of autophagic flux. Biochem Pharmacol 2022; 201:115060. [PMID: 35513042 DOI: 10.1016/j.bcp.2022.115060] [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: 11/03/2021] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 11/02/2022]
Abstract
Breast cancer is the most common malignancy among women worldwide. As conventional therapies are only partially successful in eradicating breast cancer, the development of novel strategies is a top priority. We previously showed that C25, a new racemosin B derivative, exerts its anti-cancer activity through inhibition of autophagy, but the underlying mechanism remained unknown. Here we show that C25 inhibits the growth of diverse breast cancer cell subtypes and effectively suppresses tumor progression in a xenotransplantation model of triple negative breast cancer. C25 acts as a lysosomotropic agent to induce lysosomal membrane permeabilization and inhibit autophagic flux, resulting in cathepsin release and cell death. In accordance, RNA sequencing and gene set enrichment analysis revealed that C25 induces pathways consistent with autophagy inhibition, cell cycle arrest and senescence. Interestingly, knockdown of TFEB or SQSTM1 reduced cell death induced by C25 treatment. Finally, we show that C25 synergizes with the chemo-therapeutics etoposide and paclitaxel to further limit breast cancer cell growth. Thus, C25 alone or in combination with other anti-neoplastic agents offers a novel therapeutic strategy for aggressive forms of breast cancer and possibly other malignancies.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China
| | - Philip E D Chung
- Toronto General Research Institute, University Health Network, 67 College Street, Toronto, ON M5G 2M1, Canada; Department of Laboratory Medicine& Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mei Xu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Yangju Ju
- Toronto General Research Institute, University Health Network, 67 College Street, Toronto, ON M5G 2M1, Canada; Department of Laboratory Medicine& Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Xinmei Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Jialei Song
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Jinrui Song
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Chunlin Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Eldad Zacksenhaus
- Toronto General Research Institute, University Health Network, 67 College Street, Toronto, ON M5G 2M1, Canada; Department of Laboratory Medicine& Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Sheng Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| | - Zhixu He
- Key Laboratory of Adult Stem Cell Transformation, Chinese Academy of Medical Sciences, Guiyang 550004, PR China; Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China.
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| |
Collapse
|
11
|
Zhu P, Chen Y, Wang J, Lin G, Wang R, Que Y, Zhou J, Xu G, Luo J, Du Y. Receptor-Interacting Protein Kinase 3 Suppresses Mitophagy Activation via the Yes-Associated Protein/Transcription Factor EB Pathways in Septic Cardiomyopathy. Front Cardiovasc Med 2022; 9:856041. [PMID: 35402535 PMCID: PMC8987354 DOI: 10.3389/fcvm.2022.856041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
Mitophagy, known as the main mechanism of mitochondrial quality control, determines the pathophysiology of septic cardiomyopathy, although the precise regulatory mechanisms remain elusive. Data from the present study suggested that receptor-interacting protein kinase 3 (RIPK3) expression could be enhanced in response to lipopolysaccharide (LPS) challenge. Upregulated RIPK3 expression was accompanied by severe cardiac injury and cardiac dysfunction. Further examination revealed that elevated RIPK3 expression subsequently inhibited the Yes-associated protein (YAP) pathway, which was accompanied by reduced transcription factor EB (TFEB) expression. Inhibition of TFEB would reduce mitophagy, which ultimately induced cardiomyocyte death under LPS challenge. In contrast, loss of RIPK3 induced the YAP/TFEB/mitophagy pathway alleviated the sensitivity of cardiomyocytes to LPS-induced cytotoxicity. Collectively, the RIPK3/YAP/TFEB axis was confirmed to be responsible for the pathogenesis of septic cardiomyopathy by inhibiting mitophagy. These findings have potential significance for the progression of new approaches to the treatment of septic cardiomyopathy.
Collapse
Affiliation(s)
- Pingjun Zhu
- Department of Respiratory and Critical Care Medicine, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yangxiaocao Chen
- Medical Supplies Center, Chinese PLA General Hospital, Beijing, China
| | - Junyan Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Geng Lin
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Runsheng Wang
- Department of Respiratory and Critical Care Medicine, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yifan Que
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jin Zhou
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guogang Xu
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Guogang Xu
| | - Jiang Luo
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- Jiang Luo
| | - Yingzhen Du
- Department of Disease Control and Prevention, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- Yingzhen Du
| |
Collapse
|
12
|
Li HY, Peng ZG. Targeting lipophagy as a potential therapeutic strategy for nonalcoholic fatty liver disease. Biochem Pharmacol 2022; 197:114933. [PMID: 35093393 DOI: 10.1016/j.bcp.2022.114933] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/04/2022] [Accepted: 01/21/2022] [Indexed: 02/09/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming an increasingly serious disease worldwide. Unfortunately, no specific drug has been approved to treat NAFLD. Accumulating evidence suggests that lipotoxicity, which is induced by an excess of intracellular triacylglycerols (TAGs), is a potential mechanism underlying the ill-defined progression of NAFLD. Under physiological conditions, a balance is maintained between TAGs and free fatty acids (FFAs) in the liver. TAGs are catabolized to FFAs through neutral lipolysis and/or lipophagy, while FFAs can be anabolized to TAGs through an esterification reaction. However, in the livers of patients with NAFLD, lipophagy appears to fail. Reversing this abnormal state through several lipophagic molecules (mTORC1, AMPK, PLIN, etc.) facilitates NAFLD amelioration; therefore, restoring failed lipophagy may be a highly efficient therapeutic strategy for NAFLD. Here, we outline the lipophagy phases with the relevant important proteins and discuss the roles of lipophagy in the progression of NAFLD. Additionally, the potential candidate drugs with therapeutic value targeting these proteins are discussed to show novel strategies for future treatment of NAFLD.
Collapse
Affiliation(s)
- Hong-Ying Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zong-Gen Peng
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
13
|
Lin Q, Chen J, Gu L, Dan X, Zhang C, Yang Y. New insights into mitophagy and stem cells. Stem Cell Res Ther 2021; 12:452. [PMID: 34380561 PMCID: PMC8359610 DOI: 10.1186/s13287-021-02520-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/16/2021] [Indexed: 12/21/2022] Open
Abstract
Mitophagy is a specific autophagic phenomenon in which damaged or redundant mitochondria are selectively cleared by autophagic lysosomes. A decrease in mitophagy can accelerate the aging process. Mitophagy is related to health and longevity and is the key to protecting stem cells from metabolic stress damage. Mitophagy decreases the metabolic level of stem cells by clearing active mitochondria, so mitophagy is becoming increasingly necessary to maintain the regenerative capacity of old stem cells. Stem cell senescence is the core problem of tissue aging, and tissue aging occurs not only in stem cells but also in transport amplifying cell chambers and the stem cell environment. The loss of the autophagic ability of stem cells can cause the accumulation of mitochondria and the activation of the metabolic state as well as damage the self-renewal ability and regeneration potential of stem cells. However, the claim remains controversial. Mitophagy is an important survival strategy against nutrient deficiency and starvation, and mitochondrial function and integrity may affect the viability, proliferation and differentiation potential, and longevity of normal stem cells. Mitophagy can affect the health and longevity of the human body, so the number of studies in this field has increased, but the mechanism by which mitophagy participates in stem cell development is still not fully understood. This review describes the potential significance of mitophagy in stem cell developmental processes, such as self-renewal, differentiation and aging. Through this work, we discovered the role and mechanism of mitophagy in different types of stem cells, identified novel targets for killing cancer stem cells and curing cancer, and provided new insights for future research in this field.
Collapse
Affiliation(s)
- Qingyin Lin
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology of School of Basic Medicine, Ningxia Medical University, Yinchuan, 75004, Ningxia, People's Republic of China
| | - Jiaqi Chen
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology of School of Basic Medicine, Ningxia Medical University, Yinchuan, 75004, Ningxia, People's Republic of China
| | - Lifang Gu
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology of School of Basic Medicine, Ningxia Medical University, Yinchuan, 75004, Ningxia, People's Republic of China
| | - Xingang Dan
- The Agricultural College of Ningxia University, Yinchuan, 750021, Ningxia, People's Republic of China
| | - Cheng Zhang
- College of Life Science, Capital Normal University, Beijing, 100048, People's Republic of China.
| | - Yanzhou Yang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology of School of Basic Medicine, Ningxia Medical University, Yinchuan, 75004, Ningxia, People's Republic of China.
| |
Collapse
|
14
|
Ma YS, Liu JB, Yang XL, Xin R, Shi Y, Zhang DD, Wang HM, Wang PY, Lin QL, Li W, Fu D. Basic approaches, challenges and opportunities for the discovery of small molecule anti-tumor drugs. Am J Cancer Res 2021; 11:2386-2400. [PMID: 34249406 PMCID: PMC8263657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/15/2021] [Indexed: 06/13/2023] Open
Abstract
Chemotherapy is one of the main treatments for cancer, especially for advanced cancer patients. In the past decade, significant progress has been made with the research into the molecular mechanisms of cancer cells and the precision medicine. The treatment on cancer patients has gradually changed from cytotoxic chemotherapy to precise treatment strategy. Research into anticancer drugs has also changed from killing effects on all cells to targeting drugs for target genes. Besides, researchers have developed the understanding of the abnormal physiological function, related genomics, epigenetics, and proteomics of cancer cells with cancer genome sequencing, epigenetic research, and proteomic research. These technologies and related research have accelerated the development of related cancer drugs. In this review, we summarize the research progress of anticancer drugs, the current challenges, and future opportunities.
Collapse
Affiliation(s)
- Yu-Shui Ma
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Ji-Bin Liu
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Xiao-Li Yang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Rui Xin
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Yi Shi
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Dan-Dan Zhang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Hui-Min Wang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Pei-Yao Wang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Qin-Lu Lin
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
| | - Wen Li
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
| | - Da Fu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| |
Collapse
|
15
|
Hajimolaali M, Mohammadian H, Torabi A, Shirini A, Khalife Shal M, Barazandeh Nezhad H, Iranpour S, Baradaran Eftekhari R, Dorkoosh F. Application of chloroquine as an endosomal escape enhancing agent: new frontiers for an old drug. Expert Opin Drug Deliv 2021; 18:877-889. [PMID: 33455479 DOI: 10.1080/17425247.2021.1873272] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Adequate transfection efficiency is indispensable to safe and effective delivery of therapeutically active agents, particularly in cancer. Endosomal escape is regarded as a critical and determining step devoted a significant number of studies of the drug/gene delivery field. AREAS COVERED This paper critically reviews the fundamental properties of chloroquine (CQ), its pharmacokinetics, pharmacodynamics, and clinical applications and the present knowledge of CQ application as an endosomal escape enhancing agent. Different approaches to enhance the endosomal escape process of nanoparticles have been introduced including use of endosomal escape enhancing agents. Application of CQ as either a pre-treatment modality in which cells or animals are exposed to CQ prior to the main treatment or a component of co-delivery systems where CQ and other anti-cancer agents are simultaneously entered the cancer cells, is discussed with recent studies. EXPERT OPINION CQ is founded to intervene with the natural process of endosomal maturation. Moreover, CQ seems to increase the effectiveness of gene delivery by its electrostatic interaction with negatively charged components of the transferred genetic molecules. Endosomal escape might be regarded as the bottleneck of efficient gene delivery and CQ as an effective and available endosomal escape enhancing agent deserves more sophisticated studies.
Collapse
Affiliation(s)
- Mohammad Hajimolaali
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Pátrai, Greece
| | - Hosein Mohammadian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Torabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Shirini
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Khalife Shal
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sheida Iranpour
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Baradaran Eftekhari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Al‐Bari MAA. Co-targeting of lysosome and mitophagy in cancer stem cells with chloroquine analogues and antibiotics. J Cell Mol Med 2020; 24:11667-11679. [PMID: 32935427 PMCID: PMC7578893 DOI: 10.1111/jcmm.15879] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
The catabolic autophagy eliminates cytoplasmic components and organelles via lysosomes. Non-selective bulk autophagy and selective autophagy (mitophagy) are linked in intracellular homeostasis both normal and cancer cells. Autophagy has complex and paradoxical dual role in cancers; it can play either tumour suppressor or tumour promoter depending on the tumour type, stage, microenvironment and genetic context. Cancer stem cells (CSCs) cause tumour recurrence and promote resistant to therapy for driving poor clinical consequences. Thus, new healing strategies are urgently needed to annihilate and eradicate CSCs. As chloroquine (CQ) analogues show positive clinical outcome in several clinical trials either standalone or combination with several chemotherapies. Moreover, CQ analogues are known to eliminate CSCs via altering DNA methylation. However, several obstacles such as higher concentrations and dose-dependent toxicity are noticeable in the treatment of cancers. As tumour cells predominantly rely on mitochondrial actions, mitochondrial targeting FDA-approved antibiotics are reported to effectively eradicate CSCs alone or combination with chemotherapy. However, antibiotics cause metabolic glycolytic shift in cancer cells for survival and repopulation. This review will provide a sketch of the inhibiting roles of current chloroquine analogues and antibiotic combination in CSC autophagy process and discuss the possibility that pre-clinical and clinical potential therapeutic strategy for anticancer therapy.
Collapse
|