1
|
Ali ML, Roky AH, Azad SAK, Shaikat AH, Meem JN, Hoque E, Ahasan AMF, Islam MM, Arif MSR, Mostaq MS, Mahmud MZ, Amin MN, Mahmud MA. Autophagy as a targeted therapeutic approach for skin cancer: Evaluating natural and synthetic molecular interventions. CANCER PATHOGENESIS AND THERAPY 2024; 2:231-245. [PMID: 39371094 PMCID: PMC11447340 DOI: 10.1016/j.cpt.2024.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/22/2024] [Accepted: 01/28/2024] [Indexed: 10/08/2024]
Abstract
Skin cancer, a prevalent malignancy worldwide, poses significant health concerns owing to its increasing incidence. Autophagy, a natural cellular process, is a pivotal event in skin cancer and has advantageous and detrimental effects. This duality has prompted extensive investigations into medical interventions targeting autophagy modulation for their substantial therapeutic potential. This systematic review aimed to investigate the relationship between skin cancer and autophagy and the contribution and mechanism of autophagy modulators in skin cancer. We outlined the effectiveness and safety of targeting autophagy as a promising therapeutic strategy for the treatment of skin cancer. This comprehensive review identified a diverse array of autophagy modulators with promising potential for the treatment of skin cancer. Each of these compounds demonstrates efficacy through distinct physiological mechanisms that have been elucidated in detail. Interestingly, findings from a literature search indicated that none of the natural, synthetic, or semisynthetic compounds exhibited notable adverse effects in either human or animal models. Consequently, this review offers novel mechanistic and therapeutic perspectives on the targeted modulation of autophagy in skin cancer.
Collapse
Affiliation(s)
- Md. Liakot Ali
- Department of Pharmacy, University of Chittagong, Chattogram 4331, Bangladesh
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
| | - Amdad Hossain Roky
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - S.M. Asadul Karim Azad
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Abdul Halim Shaikat
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Jannatul Naima Meem
- Department of Pharmacy, University of Chittagong, Chattogram 4331, Bangladesh
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
| | - Emtiajul Hoque
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Abu Mohammed Fuad Ahasan
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Mohammed Murshedul Islam
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, Daffodil International University, Dhaka 1216, Bangladesh
| | - Md. Saifur Rahaman Arif
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, BGC Trust University Bangladesh, Chattogram 4381, Bangladesh
| | - Md. Saqline Mostaq
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | | | - Mohammad Nurul Amin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Md. Ashiq Mahmud
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| |
Collapse
|
2
|
Ryan T, Roberts JD. Stem cell models of inherited arrhythmias. NATURE CARDIOVASCULAR RESEARCH 2024; 3:420-430. [PMID: 39196215 DOI: 10.1038/s44161-024-00451-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/29/2024] [Indexed: 08/29/2024]
Abstract
Inherited arrhythmias are a heterogeneous group of conditions that confer risk of sudden death. Many inherited arrhythmias have been linked to pathogenic genetic variants that result in ion channel dysfunction, although current genetic testing panels fail to identify variants in many patients, potentially secondary to their underlying substrates being oligogenic or polygenic. Here we review the current state of knowledge surrounding the cellular mechanisms of inherited arrhythmias generated from stem cell models with a focus on integrating genetic and mechanistic data. The utility and limitations of human induced pluripotent stem cell models in disease modeling and drug development are also explored with a particular focus on examples of pharmacogenetics and precision medicine. We submit that progress in understanding inherited arrhythmias is likely to be made by using human induced pluripotent stem cells to model probable polygenic cases as well as to interrogate the diverse and potentially complex molecular networks implicated by genome-wide association studies.
Collapse
Affiliation(s)
- Tammy Ryan
- McMaster University, Hamilton, Ontario, Canada.
| | - Jason D Roberts
- McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute and Hamilton Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
3
|
Scheidemann ER, Demas DM, Hou C, Ma J, He W, Sharma G, Schultz E, Weilbaecher KN, Shajahan-Haq AN. Resistance to abemaciclib is associated with increased metastatic potential and lysosomal protein deregulation in breast cancer cells. Mol Carcinog 2024; 63:209-223. [PMID: 37818798 DOI: 10.1002/mc.23646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Cyclin dependent kinase 4 and 6 inhibitors such as abemaciclib are routinely used to treat metastatic estrogen receptor positive (ER+) breast cancer. However, adaptive mechanisms inhibit their effectiveness and allow for disease progression. Using ER+ breast cancer cell models, we show that acquired resistance to abemaciclib is accompanied by increase in metastatic potential. Mass spectrometry-based proteomics from abemaciclib sensitive and resistant cells showed that lysosomal proteins including CTSD (cathepsin D), cathepsin A and CD68 were significantly increased in resistant cells. Combination of abemaciclib and a lysosomal destabilizer, such as hydroxychloroquine (HCQ) or bafilomycin A1, resensitized resistant cells to abemaciclib. Also, combination of abemaciclib and HCQ decreased migration and invasive potential and increased lysosomal membrane permeability in resistant cells. Prosurvival B cell lymphoma 2 (BCL2) protein levels were elevated in resistant cells, and a triple treatment with abemaciclib, HCQ, and BCL2 inhibitor, venetoclax, significantly inhibited cell growth compared to treatment with abemaciclib and HCQ. Furthermore, resistant cells showed increased levels of Transcription Factor EB (TFEB), a master regulator of lysosomal-autophagy genes, and siRNA mediated knockdown of TFEB decreased invasion in resistant cells. TFEB was found to be mutated in a subset of invasive human breast cancer samples, and overall survival analysis in ER+, lymph node-positive breast cancer showed that increased TFEB expression correlated with decreased survival. Collectively, we show that acquired resistance to abemaciclib leads to increased metastatic potential and increased levels of protumorigenic lysosomal proteins. Therefore, the lysosomal pathway could be a therapeutic target in advanced ER+ breast cancer.
Collapse
Affiliation(s)
- Erin R Scheidemann
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Diane M Demas
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Chunyan Hou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Wei He
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | | | - Eric Schultz
- Ocean Genomics Inc., Pittsburgh, Pennsylvania, USA
| | | | - Ayesha N Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
4
|
Huang H, Yan J, Xu X, Feng Y, Liu H, Liu J, Xie M, Chen L, Xiang D, Peng W, Zeng L, Zeng Y, Chen F, Zhang S, Liu Q. Everolimus inhibits hepatoblastoma by inducing autophagy-dependent ferroptosis. Drug Dev Res 2024; 85:e22140. [PMID: 38349263 DOI: 10.1002/ddr.22140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 02/15/2024]
Abstract
Everolimus, a known inhibitor of the mammalian target of rapamycin (mTOR), has shown uncertain efficacy in treating hepatoblastoma. This study delves into the potential anti-hepatoblastoma properties of everolimus and its intricate relationship with autophagy and ferroptosis, both in vitro and in vivo. In vivo, tumor tissue from hepatoblastoma patient and human hepatoblastoma cell line HuH-6 were xenografted into nude mice to establish xenograft models for observing the effect of everolimus on tumor growth. In vitro, HuH-6 cells were cultured to evaluate the anti-hepatoblastoma activity of everolimus. Transmission electron microscopy and microtubule-associated proteins 1 light chain 3 (LC3), beclin 1, and p62 protein expressions were employed to investigate autophagy. Additionally, indicators of cell apoptosis, reactive oxygen species (ROS) and proteins associated with ferroptosis were measured to evaluate ferroptosis. The results demonstrate that everolimus treatment effectively induced the formation of autophagosomes in hepatoblastoma cells, upregulated the LC3II/I ratio and beclin 1 expression, and downregulated p62 expression, indicating an enhanced autophagy level both in vitro and in vivo. Furthermore, everolimus treatment induced cell apoptosis, increased ROS level, elevated concentrations of malondialdehyde, 4-hydroxynonenal, and iron content, while reducing the ratio of glutathione/oxidized glutathione, and downregulating the protein expression of glutathione peroxidase 4 and solute carrier family 7 member 11, suggesting its ability to induce ferroptosis in hepatoblastoma cells. Importantly, the induction of ferroptosis by everolimus was significantly reversed in the presence of autophinib, an autophagy inhibitor, indicating the autophagy-dependent of everolimus-induced ferroptosis. Taken together, these findings suggest that everolimus holds promise as an effective anti-hepatoblastoma drug, with its mechanism of action potentially involving the induction of autophagy-dependent ferroptosis in hepatoblastoma cells.
Collapse
Affiliation(s)
- Haijin Huang
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi Provincial Clinical Research Center for Vascular Anomalies, The First Affiliated Hospital of GanNan Medical University, Ganzhou, Jiangxi, China
| | - Jinlong Yan
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xianyun Xu
- Department of Clinical Laboratory, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Yanping Feng
- Department of Neurological Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Haijin Liu
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jianping Liu
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Mingfeng Xie
- Jiangxi Provincial Clinical Research Center for Vascular Anomalies, The First Affiliated Hospital of GanNan Medical University, Ganzhou, Jiangxi, China
- Chinese & Western Integrative Medicine Discipline, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of TCM for Prevention and Treatment on Hemangioma, Nanchang, Jiangxi, China
- Integrated Chinese and Western Medicine Institute for Children Health & Drug Innovation, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Leifeng Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Deng Xiang
- Department of General Surgery, The Affiliated Children's Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Wei Peng
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Linshan Zeng
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yong Zeng
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Feng Chen
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shouhua Zhang
- Department of General Surgery, The Affiliated Children's Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Qian Liu
- Jiangxi Provincial Clinical Research Center for Vascular Anomalies, The First Affiliated Hospital of GanNan Medical University, Ganzhou, Jiangxi, China
- Chinese & Western Integrative Medicine Discipline, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of TCM for Prevention and Treatment on Hemangioma, Nanchang, Jiangxi, China
- Integrated Chinese and Western Medicine Institute for Children Health & Drug Innovation, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
5
|
Chen Y, Wu C, Zhao X, Tan H, Li C, Deng Y, Chen X, Wu Y, Tian N, Zhang X, Zhou Y, Sun L. 20-Deoxyingenol alleviates intervertebral disc degeneration by activating TFEB in nucleus pulposus cells. Biochem Pharmacol 2023; 218:115865. [PMID: 37863322 DOI: 10.1016/j.bcp.2023.115865] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent degenerative disease with significant adverse implications for patients' quality of life and socioeconomic status. Although the precise etiology of IVDD remains elusive, the senescence of nucleus pulposus cells is recognized as the primary pathogenic factor of IVDD; however, drugs that may targetedly inhibit senescence are still lacking. In the current study, we evaluated the small-molecule active drug 20-Deoxyingenol(20-DOI) for its effects on combating senescence and delaying the progression of IVDD. In vitro experiments revealed that the administration of 20-DOI displayed inhibitory effects on senescence and the senescence-related cGAS-STING pathway of nucleus pulposus cells. Additionally, it exhibited the ability to enhance lysosome activity and promote autophagy flux within nucleus pulposus cells. Subsequent investigations elucidated that the inhibitory impact of 20-DOI on nucleus pulposus cell senescence was mediated through the autophagy-lysosome pathway. This effect was diminished in the presence of transcription factor EB (TFEB) small hairpin RNA (shRNA), thereby confirming the regulatory role of 20-DOI on the autophagy-lysosome pathway and senescence through TFEB. In vivo experiments demonstrated that 20-DOI effectively impeded the progression ofIVDD in rats. These findings collectively illustrate that 20-DOI may facilitate the autophagy-lysosomal pathway by activating TFEB, thereby suppressing the senescence in nucleus pulposus cells, thus suggesting 20-DOI as a promising therapeutic approach for IVDD.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chenyu Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiaoying Zhao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hongye Tan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chenchao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuxin Deng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ximiao Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yaosen Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Liaojun Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
6
|
Smith M, Zhang L, Jin Y, Yang M, Bade A, Gillis KD, Jana S, Bypaneni RN, Glass TE, Lin H. A Turn-On Fluorescent Amino Acid Sensor Reveals Chloroquine's Effect on Cellular Amino Acids via Inhibiting Cathepsin L. ACS CENTRAL SCIENCE 2023; 9:980-991. [PMID: 37252359 PMCID: PMC10214525 DOI: 10.1021/acscentsci.2c01325] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Indexed: 05/31/2023]
Abstract
Maintaining homeostasis of metabolites such as amino acids is critical for cell survival. Dysfunction of nutrient balance can result in human diseases such as diabetes. Much remains to be discovered about how cells transport, store, and utilize amino acids due to limited research tools. Here we developed a novel, pan-amino acid fluorescent turn-on sensor, NS560. It detects 18 of the 20 proteogenic amino acids and can be visualized in mammalian cells. Using NS560, we identified amino acids pools in lysosomes, late endosomes, and surrounding the rough endoplasmic reticulum. Interestingly, we observed amino acid accumulation in large cellular foci after treatment with chloroquine, but not with other autophagy inhibitors. Using a biotinylated photo-cross-linking chloroquine analog and chemical proteomics, we identified Cathepsin L (CTSL) as the chloroquine target leading to the amino acid accumulation phenotype. This study establishes NS560 as a useful tool to study amino acid regulation, identifies new mechanisms of action of chloroquine, and demonstrates the importance of CTSL regulation of lysosomes.
Collapse
Affiliation(s)
- Michael
R. Smith
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Le Zhang
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Yizhen Jin
- Graduate
Program of Biochemistry, Molecular and Cell Biology, Department of
Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United
States
| | - Min Yang
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Anusha Bade
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Kevin D. Gillis
- Dalton
Cardiovascular Research Center, Department of Bioengineering and Department
of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65211, United States
| | - Sadhan Jana
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Ramesh Naidu Bypaneni
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Timothy E. Glass
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Hening Lin
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
- Howard
Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
7
|
Zuo Y, Chai Y, Liu X, Gao Z, Jin X, Wang F, Bai Y, Zheng Z. A ratiometric fluorescent probe based on spiropyran in situ switching for tracking dynamic changes of lysosomal autophagy and anticounterfeiting. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 291:122338. [PMID: 36657288 DOI: 10.1016/j.saa.2023.122338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/28/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Autophagy is the controlled breakdown of cellular components that dysfunctional or nonessential, and the decomposition products are further recycled and synthesized for the normal physiological activities of cells. Lysosomal autophagy has been implicated in cancer, neurological disorders, Parkinson's disease, etc. Therefore, it is necessary to develop a fluorescent probe that can clearly describe the process of lysosomal autophagy. However, there are currently limited fluorescent probes for ratiometric monitoring of the autophagic process in dual channels. To solve this problem, a fluorescent probe based on spiropyran with lysosomal targeting and pH response for ratiometric monitoring the autophagy process of lysosomes were designed. The sensitive response of the probe to pH in vitro was verified by UV and fluorescence spectrum tests. Meanwhile, the probe demonstrated the ability to monitor the intracellular pH fluctuations. In addition, the application of Lyso-SD in the field of anti-counterfeiting has been proposed based on the obvious photoluminescence ability of Lyso-SD under UV irradiation.
Collapse
Affiliation(s)
- Yujing Zuo
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China; Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Yanfu Chai
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China; School of Mechanical and Electrical Engineering, Shaoxing University, Shaoxing 312000, China; Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China.
| | - Xiaofei Liu
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Zhiming Gao
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Xiaofeng Jin
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Feng Wang
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Yongjie Bai
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Zhijun Zheng
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| |
Collapse
|
8
|
Bohusné Barta B, Simon Á, Nagy L, Dankó T, Raffay RE, Petővári G, Zsiros V, Sebestyén A, Sipos F, Műzes G. Survival of HT29 cancer cells is influenced by hepatocyte growth factor receptor inhibition through modulation of self-DNA-triggered TLR9-dependent autophagy response. PLoS One 2022; 17:e0268217. [PMID: 35551547 PMCID: PMC9098092 DOI: 10.1371/journal.pone.0268217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
HGFR activation drives the malignant progression of colorectal cancer, and its inhibition displays anti-autophagic activity. The interrelated role of HGFR inhibition and TLR9/autophagy signaling in HT29 cancer cells subjected to modified self-DNA treatments has not been clarified. We analyzed this complex interplay with cell metabolism and proliferation measurements, TLR9, HGFR and autophagy inhibitory assays and WES Simple Western blot-based autophagy flux measurements, gene expression analyses, immunocytochemistry, and transmission electron microscopy. The overexpression of MyD88 and caspase-3 was associated with enhanced HT29 cell proliferation, suggesting that incubation with self-DNAs could suppress the apoptosis-induced compensatory cell proliferation. HGFR inhibition blocked the proliferation-reducing effect of genomic and hypermethylated, but not that of fragmented DNA. Lowest cell proliferation was achieved with the concomitant use of genomic DNA, HGFR inhibitor, and chloroquine, when the proliferation stimulating effect of STAT3 overexpression could be outweighed by the inhibitory effect of LC3B, indicating the putative involvement of HGFR-mTOR-ULK1 molecular cascade in HGFR inhibitor-mediated autophagy. The most intense cell proliferation was caused by the co-administration of hypermethylated DNA, TLR9 and HGFR inhibitors, when decreased expression of both canonical and non-canonical HGFR signaling pathways and autophagy-related genes was present. The observed ultrastructural changes also support the context-dependent role of HGFR inhibition and autophagy on cell survival and proliferation. Further investigation of the influence of the studied signaling pathways and cellular processes can provide a basis for novel, individualized anti-cancer therapies.
Collapse
Affiliation(s)
- Bettina Bohusné Barta
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| | - Ágnes Simon
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| | - Lőrinc Nagy
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| | - Titanilla Dankó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Regina Eszter Raffay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Petővári
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Viktória Zsiros
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ferenc Sipos
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| | - Györgyi Műzes
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
9
|
Chloroquine and COVID-19—A systems biology model uncovers the drug’s detrimental effect on autophagy and explains its failure. PLoS One 2022; 17:e0266337. [PMID: 35390060 PMCID: PMC8989232 DOI: 10.1371/journal.pone.0266337] [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: 08/12/2021] [Accepted: 03/19/2022] [Indexed: 12/05/2022] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has resulted in an urgent need for identifying potential therapeutic drugs. In the first half of 2020 tropic antimalarial drugs, such as chloroquine (CQ) or hydroxochloroquine (HCQ) were the focus of tremendous public attention. In the initial periods of the pandemic, many scientific results pointed out that CQ/HCQ could be very effective for patients with severe COVID. While CQ and HCQ have successfully been used against several diseases (such as malaria, autoimmune disease and rheumatic illnesses); long term use of these agents are associated with serious adverse effects (i.e. inducing acute kidney injury, among many others) due to their role in blocking autophagy-dependent self-degradation. Recent experimental and clinical trial data also confirmed that there is no sufficient evidence about the efficient usage of CQ/HCQ against COVID-19. By using systems biology techniques, here we show that the cellular effect of CQ/HCQ on autophagy during endoplasmic reticulum (ER) stress or following SARS-CoV-2 infection results in upregulation of ER stress. By presenting a simple mathematical model, we claim that although CQ/HCQ might be able to ameliorate virus infection, the permanent inhibition of autophagy by CQ/HCQ has serious negative effects on the cell. Since CQ/HCQ promotes apoptotic cell death, here we confirm that addition of CQ/HCQ cannot be really effective even in severe cases. Only a transient treatment seemed to be able to avoid apoptotic cell death, but this type of therapy could not limit virus replication in the infected host. The presented theoretical analysis clearly points out the utility and applicability of systems biology modelling to test the cellular effect of a drug targeting key major processes, such as autophagy and apoptosis. Applying these approaches could decrease the cost of pre-clinical studies and facilitate the selection of promising clinical trials in a timely fashion.
Collapse
|
10
|
Combination of Ascorbic Acid and Menadione Induces Cytotoxic Autophagy in Human Glioblastoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2998132. [PMID: 35368869 PMCID: PMC8967583 DOI: 10.1155/2022/2998132] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/08/2022] [Accepted: 02/19/2022] [Indexed: 01/18/2023]
Abstract
We investigated the ability of the ascorbic acid (AA) and menadione (MD) combination, the well-known reactive oxidative species- (ROS-) generating system, to induce autophagy in human U251 glioblastoma cells. A combination of AA and MD (AA+MD), in contrast to single treatments, induced necrosis-like cell death mediated by mitochondrial membrane depolarization and extremely high oxidative stress. AA+MD, and to a lesser extent MD alone, prompted the appearance of autophagy markers such as autophagic vacuoles, autophagosome-associated LC3-II protein, degradation of p62, and increased expression of beclin-1. While both MD and AA+MD increased phosphorylation of AMP-activated protein kinase (AMPK), the well-known autophagy promotor, only the combined treatment affected its downstream targets, mechanistic target of rapamycin complex 1 (mTORC1), Unc 51-like kinase 1 (ULK1), and increased the expression of several autophagy-related genes. Antioxidant N-acetyl cysteine reduced both MD- and AA+MD-induced autophagy, as well as changes in AMPK/mTORC1/ULK1 activity and cell death triggered by the drug combination. Pharmacological and genetic autophagy silencing abolished the toxicity of AA+MD, while autophagy upregulation enhanced the toxicity of both AA+MD and MD. Therefore, by upregulating oxidative stress, inhibiting mTORC1, and activating ULK1, AA converts MD-induced AMPK-dependent autophagy from nontoxic to cytotoxic. These results suggest that AA+MD or MD treatment in combination with autophagy inducers could be further investigated as a novel approach for glioblastoma therapy.
Collapse
|
11
|
Liao HY, Wang ZQ, Ran R, Zhou KS, Ma CW, Zhang HH. Biological Functions and Therapeutic Potential of Autophagy in Spinal Cord Injury. Front Cell Dev Biol 2022; 9:761273. [PMID: 34988074 PMCID: PMC8721099 DOI: 10.3389/fcell.2021.761273] [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: 08/20/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an evolutionarily conserved lysosomal degradation pathway that maintains metabolism and homeostasis by eliminating protein aggregates and damaged organelles. Many studies have reported that autophagy plays an important role in spinal cord injury (SCI). However, the spatiotemporal patterns of autophagy activation after traumatic SCI are contradictory. Most studies show that the activation of autophagy and inhibition of apoptosis have neuroprotective effects on traumatic SCI. However, reports demonstrate that autophagy is strongly associated with distal neuronal death and the impaired functional recovery following traumatic SCI. This article introduces SCI pathophysiology, the physiology and mechanism of autophagy, and our current review on its role in traumatic SCI. We also discuss the interaction between autophagy and apoptosis and the therapeutic effect of activating or inhibiting autophagy in promoting functional recovery. Thus, we aim to provide a theoretical basis for the biological therapy of SCI.
Collapse
Affiliation(s)
- Hai-Yang Liao
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhi-Qiang Wang
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Rui Ran
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Kai-Sheng Zhou
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Chun-Wei Ma
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Hai-Hong Zhang
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
12
|
Guo Y, Gao F, Wang X, Pan Z, Wang Q, Xu S, Pan S, Li L, Zhao D, Qian J. Spontaneous formation of neutrophil extracellular traps is associated with autophagy. Sci Rep 2021; 11:24005. [PMID: 34907287 PMCID: PMC8671464 DOI: 10.1038/s41598-021-03520-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/02/2021] [Indexed: 12/17/2022] Open
Abstract
Neutrophils release neutrophil extracellular traps (NETs), via NETosis, as a defense mechanism against pathogens. Neutrophils can release NETs spontaneously; however, the mechanisms underlying spontaneous NETosis remain unclear. Neutrophils isolated from healthy donors were tested for NET formation and autophagy at 1, 6, 12, and 24 h after incubation. Autophagy response was evaluated in response to various autophagy inducers and inhibitors. The relationship between autophagy and NETosis was detected in vivo using an ovalbumin-induced mouse model of asthma. We found that the increase in the proportion of spontaneous NETosis was time-dependent. The number of autophagy-positive cells also increased over time and LC3B protein played an integral role in NET formation. Trehalose (an inducer of mTOR-independent autophagy) treatment significantly increased NET formation, whereas rapamycin (an mTOR-dependent autophagy inducer) did not increase NET release by neutrophils. Compared with the control group, 3-methyladenine (an autophagy sequestration inhibitor) and hydroxychloroquine sulfate (autophagosome-lysosome fusion inhibitor) treatments significantly reduced the percentage of NET-positive cells. In vivo studies on ovalbumin-induced asthma lung sections revealed NETs and LC3B and citH3 proteins were found to co-localize with DNA. Our findings suggest that autophagy plays a crucial role in aging-related spontaneous NETosis.
Collapse
Affiliation(s)
- Yun Guo
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Fei Gao
- Department of Intensive Care Unit, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xin Wang
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Zhenzhen Pan
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Qian Wang
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Shiyao Xu
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Shanshan Pan
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Ling Li
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Deyu Zhao
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China.
| | - Jun Qian
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
13
|
Bata N, Cosford NDP. Cell Survival and Cell Death at the Intersection of Autophagy and Apoptosis: Implications for Current and Future Cancer Therapeutics. ACS Pharmacol Transl Sci 2021; 4:1728-1746. [PMID: 34927007 DOI: 10.1021/acsptsci.1c00130] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Indexed: 12/25/2022]
Abstract
Autophagy and apoptosis are functionally distinct mechanisms for cytoplasmic and cellular turnover. While these two pathways are distinct, they can also regulate each other, and central components of the apoptosis or autophagy pathway regulate both processes directly. Furthermore, several upstream stress-inducing signaling pathways can influence both autophagy and apoptosis. The crosstalk between autophagy and apoptosis has an integral role in pathological processes, including those related to cancer, homeostasis, and aging. Apoptosis is a form of programmed cell death, tightly regulated by various cellular and biochemical mechanisms, some of which have been the focus of drug discovery efforts targeting cancer therapeutics. Autophagy is a cellular degradation pathway whereby cells recycle macromolecules and organelles to generate energy when subjected to stress. Autophagy can act as either a prodeath or a prosurvival process and is both tissue and microenvironment specific. In this review we describe five groups of proteins that are integral to the apoptosis pathway and discuss their role in regulating autophagy. We highlight several apoptosis-inducing small molecules and biologics that have been developed and advanced into the clinic and discuss their effects on autophagy. For the most part, these apoptosis-inducing compounds appear to elevate autophagy activity. Under certain circumstances autophagy demonstrates cytoprotective functions and is overactivated in response to chemo- or radiotherapy which can lead to drug resistance, representing a clinical obstacle for successful cancer treatment. Thus, targeting the autophagy pathway in combination with apoptosis-inducing compounds may be a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Nicole Bata
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Nicholas D P Cosford
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
14
|
Catalani E, Giovarelli M, Zecchini S, Perrotta C, Cervia D. Oxidative Stress and Autophagy as Key Targets in Melanoma Cell Fate. Cancers (Basel) 2021; 13:cancers13225791. [PMID: 34830947 PMCID: PMC8616245 DOI: 10.3390/cancers13225791] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 01/18/2023] Open
Abstract
Melanoma originates from the malignant transformation of melanocytes and is one of the most aggressive forms of cancer. The recent approval of several drugs has increased the chance of survival although a significant subset of patients with metastatic melanoma do not show a long-lasting response to these treatments. The complex cross-talk between oxidative stress and the catabolic process autophagy seems to play a central role in all aspects of melanoma pathophysiology, from initiation to progression and metastasis, including drug resistance. However, determining the fine role of autophagy in cancer death and in response to redox disruption is still a fundamental challenge in order to advance both basic and translational aspects of this field. In order to summarize the interactions among reactive oxygen and nitrogen species, autophagy machinery and proliferation/growth/death/apoptosis/survival, we provide here a narrative review of the preclinical evidence for drugs/treatments that modulate oxidative stress and autophagy in melanoma cells. The significance and the potential for pharmacological targeting (also through multiple and combination approaches) of these two different events, which can contribute independently or simultaneously to the fate of melanoma, may help to define new processes and their interconnections underlying skin cancer biology and unravel new reliable approaches.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
- Correspondence: (C.P.); (D.C.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
- Correspondence: (C.P.); (D.C.)
| |
Collapse
|
15
|
Ristic B, Harhaji-Trajkovic L, Bosnjak M, Dakic I, Mijatovic S, Trajkovic V. Modulation of Cancer Cell Autophagic Responses by Graphene-Based Nanomaterials: Molecular Mechanisms and Therapeutic Implications. Cancers (Basel) 2021; 13:cancers13164145. [PMID: 34439299 PMCID: PMC8392723 DOI: 10.3390/cancers13164145] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Graphene-based nanomaterials (GNM) are one-to-several carbon atom-thick flakes of graphite with at least one lateral dimension <100 nm. The unique electronic structure, high surface-to-volume ratio, and relatively low toxicity make GNM potentially useful in cancer treatment. GNM such as graphene, graphene oxide, graphene quantum dots, and graphene nanofibers are able to induce autophagy in cancer cells. During autophagy the cell digests its own components in organelles called lysosomes, which can either kill cancer cells or promote their survival, as well as influence the immune response against the tumor. However, a deeper understanding of GNM-autophagy interaction at the mechanistic and functional level is needed before these findings could be exploited to increase GNM effectiveness as cancer therapeutics and drug delivery systems. In this review, we analyze molecular mechanisms of GNM-mediated autophagy modulation and its possible implications for the use of GNM in cancer therapy. Abstract Graphene-based nanomaterials (GNM) are plausible candidates for cancer therapeutics and drug delivery systems. Pure graphene and graphene oxide nanoparticles, as well as graphene quantum dots and graphene nanofibers, were all able to trigger autophagy in cancer cells through both transcriptional and post-transcriptional mechanisms involving oxidative/endoplasmic reticulum stress, AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and Toll-like receptor signaling. This was often coupled with lysosomal dysfunction and subsequent blockade of autophagic flux, which additionally increased the accumulation of autophagy mediators that participated in apoptotic, necrotic, or necroptotic death of cancer cells and influenced the immune response against the tumor. In this review, we analyze molecular mechanisms and structure–activity relationships of GNM-mediated autophagy modulation, its consequences for cancer cell survival/death and anti-tumor immune response, and the possible implications for the use of GNM in cancer therapy.
Collapse
Affiliation(s)
- Biljana Ristic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Ljubica Harhaji-Trajkovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia;
| | - Mihajlo Bosnjak
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Dakic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Srdjan Mijatovic
- Clinic for Emergency Surgery, Clinical Centre of Serbia, 11000 Belgrade, Serbia;
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
- Correspondence:
| |
Collapse
|
16
|
Shao M, Shi R, Gao ZX, Gao SS, Li JF, Li H, Cui SZ, Hu WM, Chen TY, Wu GR, Zhang J, Xu J, Sy MS, Li C. Crizotinib and Doxorubicin Cooperatively Reduces Drug Resistance by Mitigating MDR1 to Increase Hepatocellular Carcinoma Cells Death. Front Oncol 2021; 11:650052. [PMID: 34094940 PMCID: PMC8170002 DOI: 10.3389/fonc.2021.650052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/16/2021] [Indexed: 11/24/2022] Open
Abstract
As the sixth most lethal cancers worldwide, hepatocellular carcinoma (HCC) has been treated with doxorubicin (Dox) for decades. However, chemotherapy resistance, especially for Dox is an even more prominent problem due to its high cardiotoxicity. To find a regimen to reduce Dox resistance, and identify the mechanisms behind it, we tried to identify combination of drugs that can overcome drug resistance by screening tyrosine kinase inhibitor(s) with Dox with various HCC cell lines in vitro and in vivo. We report here that combination of Crizo and Dox has a synergistic effect on inducing HCC cell death. Accordingly, Crizo plus Dox increases Dox accumulation in nucleus 3-16 times compared to Dox only; HCC cell death enhanced at least 50% in vitro and tumor weights reduced ranging from 35 to 65%. Combining these two drugs reduces multiple drug resistance 1 (MDR1) protein as a result of activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK), which phosphorylates eIF2α, leading to protein translational repression. Additionally, PERK stimulation activates C-Jun terminal kinase (JNK), resulting in accumulation of unfused autophagosome to enhance autophagic cell death via Poly-ADP-ribosyltransferase (PARP-1) cleavage. When the activity of PERK or JNK is blocked, unfused autophagosome is diminished, cleaved PARP-1 is reduced, and cell death is abated. Therefore, Crizo plus Dox sensitize HCC drug resistance by engaging PERK-p- eIF2α-MDR1, and kill HCC cells by engaging PERK-JNK- autophagic cell death pathways. These newly discovered mechanisms of Crizo plus Dox not only provide a potential treatment for HCC but also point to an approach to overcome MDR1 related drug resistance in other cancers.
Collapse
Affiliation(s)
- Ming Shao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Run Shi
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhen-Xing Gao
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Shan-Shan Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jing-Feng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Huan Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Wei-Min Hu
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Tian-Yun Chen
- Department of Stomatology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gui-Ru Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jie Zhang
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Cao W, Li J, Yang K, Cao D. An overview of autophagy: Mechanism, regulation and research progress. Bull Cancer 2021; 108:304-322. [PMID: 33423775 DOI: 10.1016/j.bulcan.2020.11.004] [Citation(s) in RCA: 197] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
Autophagy refers to the formation of autophagosomes by membrane wrapping part of the cytoplasm and the organelles and proteins that need to be degraded in the cells. Autophagosomes are fused with lysosomes to form autophagolysosome, which degrade the contents of the inclusions, to achieve cell homeostasis and organelle renewal. The regulatory mechanism of autophagy is complex, and its upstream signaling pathway mainly involves mTOR dependent pathway and mTOR independent pathway (AMPK, PI3K, Ras-MAPK, p53, PTEN, endoplasmic reticulum stress). Autophagy is a phenomenon of "self-eating" in cells. Apoptosis is a phenomenon of "self-killing". Both of them share the same stimulating factors and regulatory proteins, but the threshold of induction is different. How to transform and coordinate is not clear at present. This paper summarizes the history of autophagy discovery, the structure and function of related molecules, the biological function of autophagy, the regulatory mechanism and the research results of the relationship between autophagy and apoptosis.
Collapse
Affiliation(s)
- Weiya Cao
- Anhui University of Science & Technology, Medical school, Huainan 232001, China.
| | - Jinhong Li
- Juancheng Hospital of Shandong Provincial Hospital Group, Heze 274100, China
| | - Kepeng Yang
- Anhui University of Science & Technology, Medical school, Huainan 232001, China
| | - Dongli Cao
- Anhui University of Science & Technology, Medical school, Huainan 232001, China
| |
Collapse
|
18
|
Paunovic V, Kosic M, Misirkic-Marjanovic M, Trajkovic V, Harhaji-Trajkovic L. Dual targeting of tumor cell energy metabolism and lysosomes as an anticancer strategy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118944. [PMID: 33383091 DOI: 10.1016/j.bbamcr.2020.118944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 12/04/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023]
Abstract
To sustain their proliferative and metastatic capacity, tumor cells increase the activity of energy-producing pathways and lysosomal compartment, resorting to autophagolysosomal degradation when nutrients are scarce. Consequently, large fragile lysosomes and enhanced energy metabolism may serve as targets for anticancer therapy. A simultaneous induction of energy stress (by caloric restriction and inhibition of glycolysis, oxidative phosphorylation, Krebs cycle, or amino acid/fatty acid metabolism) and lysosomal stress (by lysosomotropic detergents, vacuolar ATPase inhibitors, or cationic amphiphilic drugs) is an efficient anti-cancer strategy demonstrated in a number of studies. However, the mechanisms of lysosomal/energy stress co-amplification, apart from the protective autophagy inhibition, are poorly understood. We here summarize the established and suggest potential mechanisms and candidates for anticancer therapy based on the dual targeting of lysosomes and energy metabolism.
Collapse
Affiliation(s)
- Verica Paunovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia
| | - Milica Kosic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia
| | - Maja Misirkic-Marjanovic
- Department of Neurophysiology, Institute for Biological Research, "Sinisa Stankovic"- National Institute of Republic of Serbia, University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia
| | - Ljubica Harhaji-Trajkovic
- Department of Neurophysiology, Institute for Biological Research, "Sinisa Stankovic"- National Institute of Republic of Serbia, University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia.
| |
Collapse
|
19
|
Kong Z, Zhou C, Kang J, Tan Z. Comparison of the Effects of Nonprotein and Protein Nitrogen on Apoptosis and Autophagy of Rumen Epithelial Cells in Goats. Animals (Basel) 2020; 10:E2079. [PMID: 33182520 PMCID: PMC7696569 DOI: 10.3390/ani10112079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/05/2020] [Indexed: 12/04/2022] Open
Abstract
Protein nutrition is particularly important for the self-renewal processes of gastrointestinal epithelial cells. The self-renewal of cells is inseparable from the interaction between apoptosis and autophagy. However, there are few reports on the relationship between different nitrogen sources and apoptosis/autophagy. In this study, the relative protein expression of Bcl-2-associated X protein(Bax), caspase-3, and p62 was significantly higher (p < 0.05), while that of Bcl-xl, Bcl-2, Beclin1, and Microtuble-associated protein light chain 3 (LC3-II) was significantly lower (p < 0.05), in the NH4Cl group in comparison with the NH4Cl + 4-phenylbutyric acid (4PBA) group. In addition, the relative protein expression of Bax and caspase-3 was significantly higher (p < 0.05), while that of Bcl-2 and Bcl-xl was decreased significantly (p < 0.05), in the NH4Cl + 3-Methyladenine (3-MA) group and the methionine (Met) + 3-MA group in comparison with the NH4Cl group. Furthermore, the relative protein expression of Beclin1 and LC3B-II was significantly lower (p < 0.05), while that of p62 was significantly higher (p < 0.05), in the NH4Cl + Z-VAD-FMK group and the Met + Z-VAD-FMK group in comparison with the NH4Cl group. In conclusion, our results suggested that endoplasmic reticulum (ER) stress played a critical role in the crosstalk between apoptosis and autophagy induced by NH4Cl and Met. Autophagy had a more obvious ameliorative effect on ruminal epithelial cell apoptosis after treatment with nonprotein nitrogen than after treatment with protein nitrogen. These findings may reveal the molecular mechanism of apoptosis and autophagy induced by nonprotein nitrogen and protein nitrogen.
Collapse
Affiliation(s)
- Zhiwei Kong
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- College of Food Engineering and Biotechnology, Han Shan Normal University, Chaozhou 521041, China
| | - Chuanshe Zhou
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Hunan Co-Innovation Center of Safety Animal Production, CICSAP, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410128, China
| | - Jinhe Kang
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Hunan Co-Innovation Center of Safety Animal Production, CICSAP, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410128, China
| | - Zhiliang Tan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Hunan Co-Innovation Center of Safety Animal Production, CICSAP, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410128, China
| |
Collapse
|
20
|
Fu R, Zhang L, Li Y, Li B, Ming Y, Li Z, Xing H, Chen J. Saikosaponin D inhibits autophagosome‑lysosome fusion and induces autophagy‑independent apoptosis in MDA‑MB‑231 breast cancer cells. Mol Med Rep 2020; 22:1026-1034. [PMID: 32468000 PMCID: PMC7339770 DOI: 10.3892/mmr.2020.11155] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 04/30/2020] [Indexed: 12/17/2022] Open
Abstract
The present study aimed to explore the effect of Saikosaponin D (SSD) and its underlying mechanism on apoptosis and autophagy in human breast cancer MDA‑MB‑231 cells. MTT assay, flow cytometry, western blotting and confocal fluorescence microscopy detection were employed. SSD, a kind of triterpenoid saponins extracted from Radix bupleuri, has been demonstrated to have the effects of anti‑inflammatory, antioxidative and anticancer effects and can regulate autophagy. The present study revealed that SSD induced apoptosis through the activation of the p38 mitogen‑activated protein kinase (MAPK) signaling pathway in human breast cancer MDA‑MB‑231 cells. The administration of SSD promoted the phosphorylation/activation of p38 MAPK in MDA‑MB‑231 cells, whereas pretreatment with SB203580, an effective p38 MAPK inhibitor, attenuated SSD‑mediated apoptosis, the cleavage of PARP and the activation of caspase‑3. In addition, SSD blocked autophagic degradation by inhibiting autolysosome formation, resulting in the accumulation of autophagosomes. Mechanistically, the results of the present study revealed that SSD inhibited the formation of autophagosomes by inhibiting autophagosome‑lysosome fusion, rather than by damaging lysosome function. Furthermore, blocking autophagy degradation was not associated with SSD‑mediated apoptosis. The genetic knockdown of autophagy‑related protein 5 markedly reduced SSD‑mediated LC3B‑II accumulation; however, it did not affect the SSD‑mediated phosphorylation/activation of p38, cleavage of PARP, activation of caspase‑3 or apoptosis. In conclusion, the findings of the present study suggest that SSD may induce apoptosis and block autophagic degradation, which provides further evidence of the association between the inhibition of autophagic degradation and cell death.
Collapse
Affiliation(s)
- Ruoqiu Fu
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| | - Lin Zhang
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| | - Yuanyuan Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| | - Bin Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| | - Yue Ming
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| | - Zhiwei Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| | - Haiyan Xing
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital, Army Medical University, Daping, Chongqing 400042, P.R. China
| |
Collapse
|
21
|
Qin B, Zou S, Li K, Wang H, Wei W, Zhang B, Xiao L, Choi HH, Tang Q, Huang D, Liu Q, Pan Q, Meng M, Fang L, Lee MH. CSN6-TRIM21 axis instigates cancer stemness during tumorigenesis. Br J Cancer 2020; 122:1673-1685. [PMID: 32225170 PMCID: PMC7250844 DOI: 10.1038/s41416-020-0779-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/13/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Background Cancer stem cells (CSCs) are responsible for tumour initiation, metastasis and recurrence. However, the mechanism of CSC formation, maintenance and expansion in colorectal cancer (CRC) remains poorly characterised. Methods The role of COP9 signalosome subunit 6 (CSN6) in regulating cancer stemness was evaluated by organoid formation and limited dilution analysis. The role of CSN6–TRIM21–OCT1–ALDH1A1 axis in CSC formation was evaluated in vitro and in vivo. The association of CSN6, TRIM21 and ALDH1A1 expression was validated by a tissue microarray with 267 CRC patients. Results The results showed that CSN6 is critical for sphere formation and maintaining the growth of patient-derived organoids (PDOs). We characterised the role of CSN6 in regulating cancer stemness, which involves the TRIM21 E3 ubiquitin ligase, transcription factor POU class 2 homeobox 1 (OCT1) and cancer stem cell marker aldehyde dehydrogenase 1 A1 (ALDH1A1). Our data showed that CSN6 facilitates ubiquitin-mediated degradation of TRIM21, which in turn decreases TRIM21-mediated OCT1 ubiquitination and subsequently stabilises OCT1. Consequently, OCT1 stabilisation leads to ALDH1A1expression and promotes cancer stemness. We further showed that the protein expression levels of CSN6, TRIM21 and ALDH1A1 can serve as prognostic markers for human CRC. Conclusions In conclusion, we validate a pathway for cancer stemness regulation involving ALDH1A1 levels through the CSN6–TRIM21 axis, which may be utilised as CRC molecular markers and be targeted for therapeutic intervention in cancers.
Collapse
Affiliation(s)
- Baifu Qin
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Shaomin Zou
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Kai Li
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Huashe Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Wenxia Wei
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Boyu Zhang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Lishi Xiao
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Hyun Ho Choi
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Qin Tang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Dandan Huang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Qingxin Liu
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Qihao Pan
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Manqi Meng
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Lekun Fang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China. .,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China. .,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.
| | - Mong-Hong Lee
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China. .,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China.
| |
Collapse
|