1
|
Ma K, Guo F, Li R, Song G, Zhang H, Lu Q, Ma K, Gong S. Knockdown of Atg1 Impairs Brain Regeneration and Downregulates ECM-Related Genes in the Planarian Dugesia japonica. Mol Neurobiol 2025:10.1007/s12035-025-04978-3. [PMID: 40281299 DOI: 10.1007/s12035-025-04978-3] [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: 12/15/2024] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Planarian regeneration is a complex process that involves the precise orchestration of cell proliferation, differentiation, migration, and autophagy. However, the role of autophagy in planarian regeneration remains poorly understood. In this study, we identified autophagy-related gene 1 from the planarian Dugesia japonica (designated as DjAtg1) and investigated its role in planarian brain regeneration. DjAtg1 transcripts are highly expressed in the cephalic ganglia of intact planarians. Following amputation, DjAtg1 is prominently expressed in the newly regenerated brain tissues. Knockdown of DjAtg1 via RNA interference (RNAi) induces head regression, with all RNAi-treated animals regenerating a small triangular-shaped head. Neoblast-marker labeling experiments demonstrate that DjAtg1 knockdown does not affect cell proliferation but impairs neoblast behavior. Notably, RNA-seq reveals that most of these down-regulated transcripts are linked to the extracellular matrix (ECM). Based on our findings and prior literature, we propose that the DjAtg1-mediated secretory pathway is essential for ECM remodeling. DjAtg1 knockdown disrupts the secretory pathway, which feedback-inhibits the expression of ECM-related genes. Our work provides new insights into the non-canonical role of autophagy in regulating of ECM remodeling during planarian regeneration.
Collapse
Affiliation(s)
- Kexue Ma
- Department of Basic Medicine, Luohe Medical College, Luohe, 462002, China.
- Henan Province Engineering Research Center of Nutrition and Health, Luohe, 462002, China.
| | - Fangying Guo
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Rui Li
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Gege Song
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hecai Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Qiong Lu
- Department of Basic Medicine, Luohe Medical College, Luohe, 462002, China
| | - Keshi Ma
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, 466001, China
| | - Shaoqing Gong
- Department of Basic Medicine, Luohe Medical College, Luohe, 462002, China.
| |
Collapse
|
2
|
Zhao P, Yin C, Liu R, Shao S, Ke W, Song Z. Exosome-Delivered circFOXP1 Upregulates Autophagy and Promotes Hepatocellular Carcinoma Progression Through Its Encoded p196 Protein Targeting the KHDRBS3/ULK1 Axis. Int J Nanomedicine 2025; 20:5247-5265. [PMID: 40292405 PMCID: PMC12034270 DOI: 10.2147/ijn.s505157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/05/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Circular RNAs (circRNAs) are pivotal regulators in cancer, and circFOXP1 has been implicated in tumorigenesis. This study explores the exosome-mediated transfer of circFOXP1 and its functional protein product, p196, in hepatocellular carcinoma (HCC) progression. Methods HCC circRNA datasets were obtained from the Gene Expression Omnibus (GEO) databases, and circRNAs were validated via qRT-PCR and Sanger sequencing. Exosomes were isolated via ultracentrifugation and characterized by TEM/NTA. RIP, Co-IP, RNA pull-down and in vitro binding assays were employed to determine molecular interactions. Loss- and gain-of-function assays were employed to evaluate the effects of circFOXP1, KHDRBS3 and ULK1 on the proliferation, and invasion abilities of HCC cells both in vitro and in vivo. Results CircFOXP1, which encoded a 196-amino acid protein, p196, was highly expressed in HCC tissues and cells and secreted via exosomes. Overexpression of p196 enhanced HCC cell proliferation, invasion, and autophagy flux in vitro, while knockdown produced opposite effects. Mechanistically, p196 directly bound KHDRBS3 through its D2 domain, forming a complex that stabilized ULK1 mRNA, thereby increasing ULK1 protein levels, activating autophagy and accelerating tumor progression. Conclusion Our findings indicated that circFOXP1-encoded p196 plays a role as a tumor promoter, contributing to the malignant progression of HCC. Targeting the circFOXP1/p196-KHDRBS3-ULK1 axis presents a promising therapeutic strategy for HCC, with potential applications in biomarker development and combination therapies.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Ran Liu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Shuyu Shao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Wenbo Ke
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| |
Collapse
|
3
|
Shamsudin NF, Leong SW, Koeberle A, Suriya U, Rungrotmongkol T, Chia SL, Taher M, Haris MS, Alshwyeh HA, Alosaimi AA, Mediani A, Ilowefah MA, Islami D, Mohd Faudzi SM, Fasihi Mohd Aluwi MF, Wai LK, Rullah K. A novel chromone-based as a potential inhibitor of ULK1 that modulates autophagy and induces apoptosis in colon cancer. Future Med Chem 2024; 16:1499-1517. [PMID: 38949858 PMCID: PMC11370956 DOI: 10.1080/17568919.2024.2363668] [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: 08/30/2023] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
Aim: Chromones are promising for anticancer drug development.Methods & results: 12 chromone-based compounds were synthesized and tested against cancer cell lines. Compound 8 showed the highest cytotoxicity (LC50 3.2 μM) against colorectal cancer cells, surpassing 5-fluorouracil (LC50 4.2 μM). It suppressed colony formation, induced cell cycle arrest and triggered apoptotic cell death, confirmed by staining and apoptosis markers. Cell death was accompanied by enhanced reactive oxygen species formation and modulation of the autophagic machinery (autophagy marker light chain 3B (LC3B); adenosine monophosphate-activated protein kinase (AMPK); protein kinase B (PKB); UNC-51-like kinase (ULK)-1; and ULK2). Molecular docking and dynamic simulations revealed that compound 8 directly binds to ULK1.Conclusion: Compound 8 is a promising lead for autophagy-modulating anti-colon cancer drugs.
Collapse
Affiliation(s)
- Nur Farisya Shamsudin
- Drug Discovery & Synthetic Chemistry Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Bandar Indera Mahkota, Kuantan25200, Pahang, Malaysia
| | - Sze-Wei Leong
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur50603, Malaysia
| | - Andreas Koeberle
- Michael Popp Institute & Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck6020, Austria
| | - Utid Suriya
- Structural & Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok10330, Thailand
| | - Thanyada Rungrotmongkol
- Structural & Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok10330, Thailand
| | - Suet Lin Chia
- UPM – MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang43400, Selangor, Malaysia
| | - Muhammad Taher
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan25200, Pahang, Malaysia
| | - Muhammad Salahuddin Haris
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan25200, Pahang, Malaysia
| | - Hussah Abdullah Alshwyeh
- Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam31441, Saudi Arabia
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam31441, Saudi Arabia
| | - Areej A Alosaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam31441, Saudi Arabia
| | - Ahmed Mediani
- Institute of Systems Biology, Universiti Kebangsaan Malaysia (UKM), Bangi43600, Malaysia
| | | | - Deri Islami
- Faculty of Pharmacy & Health Sciences, Universitas Abdurrab, Jalan Riau Ujung, Pekanbaru28292, Riau, Indonesia
| | - Siti Munirah Mohd Faudzi
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang43400, Selangor, Malaysia
| | | | - Lam Kok Wai
- Drugs & Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur50300, Malaysia
| | - Kamal Rullah
- Drug Discovery & Synthetic Chemistry Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Bandar Indera Mahkota, Kuantan25200, Pahang, Malaysia
| |
Collapse
|
4
|
Pareek G, Kundu M. Physiological functions of ULK1/2. J Mol Biol 2024; 436:168472. [PMID: 38311233 PMCID: PMC11382334 DOI: 10.1016/j.jmb.2024.168472] [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: 12/19/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
UNC-51-like kinases 1 and 2 (ULK1/2) are serine/threonine kinases that are best known for their evolutionarily conserved role in the autophagy pathway. Upon sensing the nutrient status of a cell, ULK1/2 integrate signals from upstream cellular energy sensors such as mTOR and AMPK and relay them to the downstream components of the autophagy machinery. ULK1/2 also play indispensable roles in the selective autophagy pathway, removing damaged mitochondria, invading pathogens, and toxic protein aggregates. Additional functions of ULK1/2 have emerged beyond autophagy, including roles in protein trafficking, RNP granule dynamics, and signaling events impacting innate immunity, axon guidance, cellular homeostasis, and cell fate. Therefore, it is no surprise that alterations in ULK1/2 expression and activity have been linked with pathophysiological processes, including cancer, neurological disorders, and cardiovascular diseases. Growing evidence suggests that ULK1/2 function as biological rheostats, tuning cellular functions to intra and extra-cellular cues. Given their broad physiological relevance, ULK1/2 are candidate targets for small molecule activators or inhibitors that may pave the way for the development of therapeutics for the treatment of diseases in humans.
Collapse
Affiliation(s)
- Gautam Pareek
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mondira Kundu
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
5
|
Kurganovs NJ, Engedal N. To eat or not to eat: a critical review on the role of autophagy in prostate carcinogenesis and prostate cancer therapeutics. Front Pharmacol 2024; 15:1419806. [PMID: 38910881 PMCID: PMC11190189 DOI: 10.3389/fphar.2024.1419806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Around 1 in 7 men will be diagnosed with prostate cancer during their lifetime. Many strides have been made in the understanding and treatment of this malignancy over the years, however, despite this; treatment resistance and disease progression remain major clinical concerns. Recent evidence indicate that autophagy can affect cancer formation, progression, and therapeutic resistance. Autophagy is an evolutionarily conserved process that can remove unnecessary or dysfunctional components of the cell as a response to metabolic or environmental stress. Due to the emerging importance of autophagy in cancer, targeting autophagy should be considered as a potential option in disease management. In this review, along with exploring the advances made on understanding the role of autophagy in prostate carcinogenesis and therapeutics, we will critically consider the conflicting evidence observed in the literature and suggest how to obtain stronger experimental evidence, as the application of current findings in clinical practice is presently not viable.
Collapse
Affiliation(s)
- Natalie Jayne Kurganovs
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Nikolai Engedal
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
6
|
Wang B, Pareek G, Kundu M. ULK/Atg1: phasing in and out of autophagy. Trends Biochem Sci 2024; 49:494-505. [PMID: 38565496 PMCID: PMC11162330 DOI: 10.1016/j.tibs.2024.03.004] [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: 12/15/2023] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
Autophagy - a highly regulated intracellular degradation process - is pivotal in maintaining cellular homeostasis. Liquid-liquid phase separation (LLPS) is a fundamental mechanism regulating the formation and function of membrane-less compartments. Recent research has unveiled connections between LLPS and autophagy, suggesting that phase separation events may orchestrate the spatiotemporal organization of autophagic machinery and cargo sequestration. The Unc-51-like kinase (ULK)/autophagy-related 1 (Atg1) family of proteins is best known for its regulatory role in initiating autophagy, but there is growing evidence that the functional spectrum of ULK/Atg1 extends beyond autophagy regulation. In this review, we explore the spatial and temporal regulation of the ULK/Atg1 family of kinases, focusing on their recruitment to LLPS-driven compartments, and highlighting their multifaceted functions beyond their traditional role.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Gautam Pareek
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mondira Kundu
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
7
|
Pavlyuchenkova AN, Smirnov MS, Chernyak BV, Chelombitko MA. The Role Played by Autophagy in FcεRI-Dependent Activation of Mast Cells. Cells 2024; 13:690. [PMID: 38667305 PMCID: PMC11049365 DOI: 10.3390/cells13080690] [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: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
The significant role of mast cells in the development of allergic and inflammatory diseases is well-established. Among the various mechanisms of mast cell activation, the interaction of antigens/allergens with IgE and the subsequent binding of this complex to the high-affinity IgE receptor FcεRI stand out as the most studied and fundamental pathways. This activation process leads to the rapid exocytosis of granules containing preformed mediators, followed by the production of newly synthesized mediators, including a diverse array of cytokines, chemokines, arachidonic acid metabolites, and more. While conventional approaches to allergy control primarily focus on allergen avoidance and the use of antihistamines (despite their associated side effects), there is increasing interest in exploring novel methods to modulate mast cell activity in modern medicine. Recent evidence suggests a role for autophagy in mast cell activation, offering potential avenues for utilizing low-molecular-weight autophagy regulators in the treatment of allergic diseases. More specifically, mitochondria, which play an important role in the regulation of autophagy as well as mast cell activation, emerge as promising targets for drug development. This review examines the existing literature regarding the involvement of the molecular machinery associated with autophagy in FcεRI-dependent mast cell activation.
Collapse
Affiliation(s)
- Anastasia N. Pavlyuchenkova
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow 119992, Russia
| | - Maxim S. Smirnov
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow 119992, Russia
| | - Boris V. Chernyak
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
| | - Maria A. Chelombitko
- Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119992, Russia; (A.N.P.)
| |
Collapse
|
8
|
Shang JN, Yu CG, Li R, Xi Y, Jian YJ, Xu N, Chen S. The nonautophagic functions of autophagy-related proteins. Autophagy 2024; 20:720-734. [PMID: 37682088 PMCID: PMC11062363 DOI: 10.1080/15548627.2023.2254664] [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: 03/09/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
ABBREVIATIONS ATG: autophagy related; BECN1: beclin 1; cAMP: cyclic adenosine monophosphate; dsDNA: double-stranded DNA; EMT: epithelial-mesenchymal transition; IFN: interferon; ISCs: intestinal stem cells; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAPK/JNK: mitogen-activated protein kinase/c-Jun N-terminal kinases; MTOR: mechanistic target of rapamycin kinase; STING1: stimulator of interferon response cGAMP interactor 1; UVRAG: UV radiation resistance associated; VPS: vacuolar protein sorting.
Collapse
Affiliation(s)
- Jia-Ni Shang
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Henan University School of Medicine, Kaifeng, Henan, PR China
| | - Chen-Ge Yu
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Henan University School of Medicine, Kaifeng, Henan, PR China
| | - Rui Li
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Henan University School of Medicine, Kaifeng, Henan, PR China
| | - Yan Xi
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Henan University School of Medicine, Kaifeng, Henan, PR China
| | - Yue Jenny Jian
- Nanjing Foreign Language School, Nanjing, Jiangsu, PR China
| | - Nan Xu
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Henan University School of Medicine, Kaifeng, Henan, PR China
| | - Su Chen
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Henan University School of Medicine, Kaifeng, Henan, PR China
| |
Collapse
|
9
|
Liang P, Zhang J, Wu Y, Zheng S, Xu Z, Yang S, Wang J, Ma S, Xiao L, Hu T, Jiang W, Huang C, Xing Q, Kundu M, Wang B. An ULK1/2-PXN mechanotransduction pathway suppresses breast cancer cell migration. EMBO Rep 2023; 24:e56850. [PMID: 37846507 PMCID: PMC10626438 DOI: 10.15252/embr.202356850] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 09/09/2023] [Accepted: 09/27/2023] [Indexed: 10/18/2023] Open
Abstract
The remodeling and stiffening of the extracellular matrix (ECM) is a well-recognized modulator of breast cancer progression. How changes in the mechanical properties of the ECM are converted into biochemical signals that direct tumor cell migration and metastasis remain poorly characterized. Here, we describe a new role for the autophagy-inducing serine/threonine kinases ULK1 and ULK2 in mechanotransduction. We show that ULK1/2 activity inhibits the assembly of actin stress fibers and focal adhesions (FAs) and as a consequence impedes cell contraction and migration, independent of its role in autophagy. Mechanistically, we identify PXN/paxillin, a key component of the mechanotransducing machinery, as a direct binding partner and substrate of ULK1/2. ULK-mediated phosphorylation of PXN at S32 and S119 weakens homotypic interactions and liquid-liquid phase separation of PXN, impairing FA assembly, which in turn alters the mechanical properties of breast cancer cells and their response to mechanical stimuli. ULK1/2 and the well-characterized PXN regulator, FAK/Src, have opposing functions on mechanotransduction and compete for phosphorylation of adjacent serine and tyrosine residues. Taken together, our study reveals ULK1/2 as important regulator of PXN-dependent mechanotransduction.
Collapse
Affiliation(s)
- Peigang Liang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Jiaqi Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Yuchen Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Shanyuan Zheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Zhaopeng Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Shuo Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Jinfang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Suibin Ma
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Li Xiao
- Department of OncologyZhongshan Hospital of Xiamen UniversityXiamenChina
| | - Tianhui Hu
- Cancer Research Center, School of MedicineXiamen UniversityXiamenChina
| | - Wenxue Jiang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio‐Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life SciencesHubei UniversityWuhanChina
| | - Chaoqun Huang
- Central LaboratoryThe Fifth Hospital of XiamenXiamenChina
| | - Qiong Xing
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio‐Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life SciencesHubei UniversityWuhanChina
| | - Mondira Kundu
- Department of Cell and Molecular BiologySt. Jude Children's Research HospitalMemphisTNUSA
| | - Bo Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
- Shenzhen Research Institute of Xiamen UniversityShenzhenChina
| |
Collapse
|
10
|
Wu R, Feng S, Li F, Shu G, Wang L, Gao P, Zhu X, Zhu C, Wang S, Jiang Q. Transcriptional and post-transcriptional control of autophagy and adipogenesis by YBX1. Cell Death Dis 2023; 14:29. [PMID: 36642732 PMCID: PMC9841012 DOI: 10.1038/s41419-023-05564-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/17/2023]
Abstract
Obesity is strongly associated with metabolic diseases, which have become a global health problem. Exploring the underlying mechanism of adipogenesis is crucial for the treatment of excess white fat. Oncogene YBX1 is a multifunctional DNA- and RNA-binding protein that regulates brown adipogenesis. However, the role of YBX1 in white adipogenesis and adipose tissue expansion remains unknown. Here, we showed that YBX1 deficiency inhibited murine and porcine adipocyte differentiation. YBX1 positively regulated adipogenesis through promoting ULK1- and ULK2-mediated autophagy. Mechanistically, we identified YBX1 serves as a 5-methylcytosine (m5C)-binding protein directly targeting m5C-containing Ulk1 mRNA by using RNA immunoprecipitation. RNA decay assay further proved that YBX1 upregulated ULK1 expression though stabilizing its mRNA. Meanwhile, YBX1 promoted Ulk2 transcription and expression as a transcription factor, thereby enhancing autophagy and adipogenesis. Importantly, YBX1 overexpression in white fat enhanced ULK1/ULK2-mediated autophagy and promoted adipose tissue expansion in mice. Collectively, these findings unveil the post-transcriptional and transcriptional mechanism and functional importance of YBX1 in autophagy and adipogenesis regulation, providing an attractive molecular target for therapies of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Ruifan Wu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shengchun Feng
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Fan Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Lina Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Ping Gao
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaotong Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Canjun Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Songbo Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
11
|
Age-related changes in tau and autophagy in human brain in the absence of neurodegeneration. PLoS One 2023; 18:e0262792. [PMID: 36701399 PMCID: PMC9879510 DOI: 10.1371/journal.pone.0262792] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 08/19/2022] [Indexed: 01/27/2023] Open
Abstract
Tau becomes abnormally hyper-phosphorylated and aggregated in tauopathies like Alzheimers disease (AD). As age is the greatest risk factor for developing AD, it is important to understand how tau protein itself, and the pathways implicated in its turnover, change during aging. We investigated age-related changes in total and phosphorylated tau in brain samples from two cohorts of cognitively normal individuals spanning 19-74 years, without overt neurodegeneration. One cohort utilised resected tissue and the other used post-mortem tissue. Total soluble tau levels declined with age in both cohorts. Phosphorylated tau was undetectable in the post-mortem tissue but was clearly evident in the resected tissue and did not undergo significant age-related change. To ascertain if the decline in soluble tau was correlated with age-related changes in autophagy, three markers of autophagy were tested but only two appeared to increase with age and the third was unchanged. This implies that in individuals who do not develop neurodegeneration, there is an age-related reduction in soluble tau which could potentially be due to age-related changes in autophagy. Thus, to explore how an age-related increase in autophagy might influence tau-mediated dysfunctions in vivo, autophagy was enhanced in a Drosophila model and all age-related tau phenotypes were significantly ameliorated. These data shed light on age-related physiological changes in proteins implicated in AD and highlights the need to study pathways that may be responsible for these changes. It also demonstrates the therapeutic potential of interventions that upregulate turnover of aggregate-prone proteins during aging.
Collapse
|
12
|
Autophagy protein ULK1 interacts with and regulates SARM1 during axonal injury. Proc Natl Acad Sci U S A 2022; 119:e2203824119. [PMID: 36375051 PMCID: PMC9704737 DOI: 10.1073/pnas.2203824119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Autophagy is a cellular catabolic pathway generally thought to be neuroprotective. However, autophagy and in particular its upstream regulator, the ULK1 kinase, can also promote axonal degeneration. We examined the role and the mechanisms of autophagy in axonal degeneration using a mouse model of contusive spinal cord injury (SCI). Consistent with activation of autophagy during axonal degeneration following SCI, autophagosome marker LC3, ULK1 kinase, and ULK1 target, phospho-ATG13, accumulated in the axonal bulbs and injured axons. SARM1, a TIR NADase with a pivotal role in axonal degeneration, colocalized with ULK1 within 1 h after SCI, suggesting possible interaction between autophagy and SARM1-mediated axonal degeneration. In our in vitro experiments, inhibition of autophagy, including Ulk1 knockdown and ULK1 inhibitor, attenuated neurite fragmentation and reduced accumulation of SARM1 puncta in neurites of primary cortical neurons subjected to glutamate excitotoxicity. Immunoprecipitation data demonstrated that ULK1 physically interacted with SARM1 in vitro and in vivo and that SAM domains of SARM1 were necessary for ULK1-SARM1 complex formation. Consistent with a role in regulation of axonal degeneration, in primary cortical neurons ULK1-SARM1 interaction increased upon neurite damage. Supporting a role for autophagy and ULK1 in regulation of SARM1 in axonal degeneration in vivo, axonal ULK1 activation and accumulation of SARM1 were both decreased after SCI in Becn1+/- autophagy hypomorph mice compared to wild-type (WT) controls. These findings suggest a regulatory crosstalk between autophagy and axonal degeneration pathways, which is mediated through ULK1-SARM1 interaction and contributes to the ability of SARM1 to accumulate in injured axons.
Collapse
|
13
|
Sakai Y, Hanafusa H, Hisamoto N, Matsumoto K. Histidine dephosphorylation of the Gβ protein GPB-1 promotes axon regeneration in C. elegans. EMBO Rep 2022; 23:e55076. [PMID: 36278516 PMCID: PMC9724660 DOI: 10.15252/embr.202255076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
Histidine phosphorylation is an emerging noncanonical protein phosphorylation in animals, yet its physiological role remains largely unexplored. The protein histidine phosphatase (PHPT1) was recently identified for the first time in mammals. Here, we report that PHIP-1, an ortholog of PHPT1 in Caenorhabditis elegans, promotes axon regeneration by dephosphorylating GPB-1 Gβ at His-266 and inactivating GOA-1 Goα signaling, a negative regulator of axon regeneration. Overexpression of the histidine kinase NDK-1 also inhibits axon regeneration via GPB-1 His-266 phosphorylation. Thus, His-phosphorylation plays an antiregenerative role in C. elegans. Furthermore, we identify a conserved UNC-51/ULK kinase that functions in autophagy as a PHIP-1-binding protein. We demonstrate that UNC-51 phosphorylates PHIP-1 at Ser-112 and activates its catalytic activity and that this phosphorylation is required for PHIP-1-mediated axon regeneration. This study reveals a molecular link from ULK to protein histidine phosphatase, which facilitates axon regeneration by inhibiting trimeric G protein signaling.
Collapse
Affiliation(s)
- Yoshiki Sakai
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| | - Hiroshi Hanafusa
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| | - Naoki Hisamoto
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| | - Kunihiro Matsumoto
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| |
Collapse
|
14
|
Nrf2 Pathway and Autophagy Crosstalk: New Insights into Therapeutic Strategies for Ischemic Cerebral Vascular Diseases. Antioxidants (Basel) 2022; 11:antiox11091747. [PMID: 36139821 PMCID: PMC9495910 DOI: 10.3390/antiox11091747] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebrovascular disease is highly prevalent and has a complex etiology and variable pathophysiological activities. It thus poses a serious threat to human life and health. Currently, pathophysiological research on cerebrovascular diseases is gradually improving, and oxidative stress and autophagy have been identified as important pathophysiological activities that are gradually attracting increasing attention. Many studies have found some effects of oxidative stress and autophagy on cerebrovascular diseases, and studies on the crosstalk between the two in cerebrovascular diseases have made modest progress. However, further, more detailed studies are needed to determine the specific mechanisms. This review discusses nuclear factor erythroid 2-related factor 2 (Nrf2) molecules, which are closely associated with oxidative stress and autophagy, and the crosstalk between them, with the aim of providing clues for studying the two important pathophysiological changes and their crosstalk in cerebrovascular diseases as well as exploring new target treatments.
Collapse
|
15
|
Liu N, Zhu M, Zhang Y, Wang Z, Li B, Ren W. Involvement of the Autophagy Protein Atg1 in Development and Virulence in Botryosphaeria dothidea. J Fungi (Basel) 2022; 8:jof8090904. [PMID: 36135629 PMCID: PMC9501979 DOI: 10.3390/jof8090904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Botryosphaeria canker and fruit rot caused by the fungus Botryosphaeria dothidea is one of the most destructive diseases of apple worldwide. Autophagy is an evolutionarily conserved self-degradation process that is important for maintaining homeostasis to ensure cellular functionality. To date, the role of autophagy in B. dothidea is not well elucidated. In this study, we identified and characterized the autophagy-related protein Atg1 in B. dothidea. The BdAtg1 deletion mutant ΔBdAtg1 showed autophagy blockade and phenotypic defects in mycelial growth, conidiation, ascosporulation and virulence. In addition, ΔBdAtg1 exhibited an increased number of nuclei in the mycelial compartment. Comparative transcriptome analysis revealed that inactivation of BdAtg1 significantly influenced multiple metabolic pathways. Taken together, our results indicate that BdAtg1 plays an important role in vegetative differentiation and the pathogenicity of B. dothidea. The results of this study will provide a reference for the development of new target-based fungicides.
Collapse
|
16
|
Function and regulation of ULK1: From physiology to pathology. Gene 2022; 840:146772. [PMID: 35905845 DOI: 10.1016/j.gene.2022.146772] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/03/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022]
Abstract
The expression of ULK1, a core protein of autophagy, is closely related to autophagic activity. Numerous studies have shown that pathological abnormal expression of ULK1 is associated with various human diseases such as neurological disorders, infections, cardiovascular diseases, liver diseases and cancers. In addition, new advances in the regulation of ULK1 have been identified. Furthermore, targeting ULK1 as a therapeutic strategy for diseases is gaining attention as new corresponding activators or inhibitors are being developed. In this review, we describe the structure and regulation of ULK1 as well as the current targeted activators and inhibitors. Moreover, we highlight the pathological disorders of ULK1 expression and its critical role in human diseases.
Collapse
|
17
|
Meng F, Shen F, Ling H, Jin P, Zhou D, Li Q. CircARHGAP12 Triggers Mesenchymal Stromal Cell Autophagy to Facilitate its Effect on Repairing Diabetic Wounds by Sponging miR-301b-3p/ATG16L1 and miR-301b-3p/ULK2. J Invest Dermatol 2022; 142:1976-1989.e4. [PMID: 34933019 DOI: 10.1016/j.jid.2021.11.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 11/07/2021] [Accepted: 11/23/2021] [Indexed: 10/19/2022]
Abstract
Circular RNAs have been confirmed to play vital roles in the development of human diseases. Nevertheless, their effects on modulating mesenchymal stromal cells (MSCs) to heal diabetic wounds are still elusive. In this study, our data revealed that MSCs treated with high glucose displayed an evident reduction in circARHGAP12 expression, whereas autophagy mediated by circARHGAP12 suppressed high glucose-triggered apoptosis of MSCs. Mechanistically, circARHGAP12 was capable of directly interacting with miR-301b-3p and subsequently sponged microRNA to modulate the expression of the miR-301b-3p target genes ATG16L1 and ULK2 and the downstream signaling pathway. Moreover, circARHGAP12 promoted the survival of MSCs in diabetic wounds in vivo and accelerated wound healing. Collectively, these results suggest that circARHGAP12/miR-301b-3p/ATG16L1 and circARHGAP12/miR-301b-3p/ULK2 regulatory networks might be an underlying therapeutic target for MSCs in diabetic wound healing.
Collapse
Affiliation(s)
- Fandong Meng
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fengjie Shen
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hongwei Ling
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Peisheng Jin
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongmei Zhou
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qiang Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
18
|
Zou L, Liao M, Zhen Y, Zhu S, Chen X, Zhang J, Hao Y, Liu B. Autophagy and beyond: Unraveling the complexity of UNC-51-like kinase 1 (ULK1) from biological functions to therapeutic implications. Acta Pharm Sin B 2022; 12:3743-3782. [PMID: 36213540 PMCID: PMC9532564 DOI: 10.1016/j.apsb.2022.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/27/2022] [Accepted: 06/02/2022] [Indexed: 12/13/2022] Open
Abstract
UNC-51-like kinase 1 (ULK1), as a serine/threonine kinase, is an autophagic initiator in mammals and a homologous protein of autophagy related protein (Atg) 1 in yeast and of UNC-51 in Caenorhabditis elegans. ULK1 is well-known for autophagy activation, which is evolutionarily conserved in protein transport and indispensable to maintain cell homeostasis. As the direct target of energy and nutrition-sensing kinase, ULK1 may contribute to the distribution and utilization of cellular resources in response to metabolism and is closely associated with multiple pathophysiological processes. Moreover, ULK1 has been widely reported to play a crucial role in human diseases, including cancer, neurodegenerative diseases, cardiovascular disease, and infections, and subsequently targeted small-molecule inhibitors or activators are also demonstrated. Interestingly, the non-autophagy function of ULK1 has been emerging, indicating that non-autophagy-relevant ULK1 signaling network is also linked with diseases under some specific contexts. Therefore, in this review, we summarized the structure and functions of ULK1 as an autophagic initiator, with a focus on some new approaches, and further elucidated the key roles of ULK1 in autophagy and non-autophagy. Additionally, we also discussed the relationships between ULK1 and human diseases, as well as illustrated a rapid progress for better understanding of the discovery of more candidate small-molecule drugs targeting ULK1, which will provide a clue on novel ULK1-targeted therapeutics in the future.
Collapse
Affiliation(s)
- Ling Zou
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongqi Zhen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiou Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiya Chen
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jin Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Corresponding authors. Tel./fax: +86 28 85503817.
| | - Yue Hao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- Corresponding authors. Tel./fax: +86 28 85503817.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Corresponding authors. Tel./fax: +86 28 85503817.
| |
Collapse
|
19
|
Discovery of a signaling feedback circuit that defines interferon responses in myeloproliferative neoplasms. Nat Commun 2022; 13:1750. [PMID: 35365653 PMCID: PMC8975834 DOI: 10.1038/s41467-022-29381-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/03/2022] [Indexed: 02/06/2023] Open
Abstract
Interferons (IFNs) are key initiators and effectors of the immune response against malignant cells and also directly inhibit tumor growth. IFNα is highly effective in the treatment of myeloproliferative neoplasms (MPNs), but the mechanisms of action are unclear and it remains unknown why some patients respond to IFNα and others do not. Here, we identify and characterize a pathway involving PKCδ-dependent phosphorylation of ULK1 on serine residues 341 and 495, required for subsequent activation of p38 MAPK. We show that this pathway is essential for IFN-suppressive effects on primary malignant erythroid precursors from MPN patients, and that increased levels of ULK1 and p38 MAPK correlate with clinical response to IFNα therapy in these patients. We also demonstrate that IFNα treatment induces cleavage/activation of the ULK1-interacting ROCK1/2 proteins in vitro and in vivo, triggering a negative feedback loop that suppresses IFN responses. Overexpression of ROCK1/2 is seen in MPN patients and their genetic or pharmacological inhibition enhances IFN-anti-neoplastic responses in malignant erythroid precursors from MPN patients. These findings suggest the clinical potential of pharmacological inhibition of ROCK1/2 in combination with IFN-therapy for the treatment of MPNs. Interferon alpha (IFNalpha) therapy is showing promising results to treat myeloproliferative neoplasms (MPNs). Here, the authors show that IFNalpha response requires ULK1 phosphorylation to induce p38-MAPK signalling but it is counteracted by ROCK1-2 activation suggesting combination therapy of IFNalpha-ROCK1-2 inhibition may improve MPNs treatment.
Collapse
|
20
|
Rajak S, Raza S, Sinha RA. ULK1 Signaling in the Liver: Autophagy Dependent and Independent Actions. Front Cell Dev Biol 2022; 10:836021. [PMID: 35252196 PMCID: PMC8894804 DOI: 10.3389/fcell.2022.836021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/04/2022] [Indexed: 12/18/2022] Open
Abstract
Liver is the primary organ for energy metabolism and detoxification in the human body. Not surprisingly, a derangement in liver function leads to several metabolic diseases. Autophagy is a cellular process, which primarily deals with providing molecules for energy production, and maintains cellular health. Autophagy in the liver has been implicated in several hepatic metabolic processes, such as, lipolysis, glycogenolysis, and gluconeogenesis. Autophagy also provides protection against drugs and pathogens. Deregulation of autophagy is associated with the development of non-alcoholic fatty liver disease (NAFLD) acute-liver injury, and cancer. The process of autophagy is synchronized by the action of autophagy family genes or autophagy (Atg) genes that perform key functions at different steps. The uncoordinated-51-like kinases 1 (ULK1) is a proximal kinase member of the Atg family that plays a crucial role in autophagy. Interestingly, ULK1 actions on hepatic cells may also involve some autophagy-independent signaling. In this review, we provide a comprehensive update of ULK1 mediated hepatic action involving lipotoxicity, acute liver injury, cholesterol synthesis, and hepatocellular carcinoma, including both its autophagic and non-autophagic functions.
Collapse
Affiliation(s)
| | | | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
21
|
Visintin R, Ray SK. Specific microRNAs for Modulation of Autophagy in Spinal Cord Injury. Brain Sci 2022; 12:247. [PMID: 35204010 PMCID: PMC8870708 DOI: 10.3390/brainsci12020247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 02/05/2023] Open
Abstract
The treatment of spinal cord injury (SCI) is currently a major challenge, with a severe lack of effective therapies for yielding meaningful improvements in function. Therefore, there is a great opportunity for the development of novel treatment strategies for SCI. The modulation of autophagy, a process by which a cell degrades and recycles unnecessary or harmful components (protein aggregates, organelles, etc.) to maintain cellular homeostasis and respond to a changing microenvironment, is thought to have potential for treating many neurodegenerative conditions, including SCI. The discovery of microRNAs (miRNAs), which are short ribonucleotide transcripts for targeting of specific messenger RNAs (mRNAs) for silencing, shows prevention of the translation of mRNAs to the corresponding proteins affecting various cellular processes, including autophagy. The number of known miRNAs and their targets continues to grow rapidly. This review article aims to explore the relationship between autophagy and SCI, specifically with the intent of identifying specific miRNAs that can be useful to modulate autophagy for neuroprotection and the improvement of functional recovery in SCI.
Collapse
Affiliation(s)
- Rhett Visintin
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA;
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| |
Collapse
|
22
|
Kang WS, Lee SM, Hwang D, Park HJ, Kim JW. Association between Unc-51-like autophagy activating kinase 2 gene polymorphisms and schizophrenia in the Korean population. Medicine (Baltimore) 2022; 101:e28745. [PMID: 35119028 PMCID: PMC8812592 DOI: 10.1097/md.0000000000028745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/08/2022] [Indexed: 01/04/2023] Open
Abstract
Accumulating evidence indicates that the autophagy process is involved in the pathogenesis of schizophrenia. Autophagy plays a fundamental role in neuronal survival and function, and autophagy-related genes have been suggested to be associated with the pathogenesis of schizophrenia. The Unc-51-like autophagy activating kinase 2 (ULK2) gene has been implicated in autophagy regulation; therefore, we hypothesized that ULK2 polymorphisms may be associated with schizophrenia susceptibility.This study explored the association between polymorphisms of ULK2 and schizophrenia.Two single nucleotide polymorphisms (SNPs) (rs55730189 and rs150122) of ULK2 were genotyped in 279 patients with schizophrenia and 403 healthy individuals using Fluidigm SNPtype assays. We analyzed the genotype distribution of 2 SNPs and haplotypes between patients with schizophrenia and control subjects.The T allele frequency of rs55730189 showed a significant association between patients with schizophrenia and control subjects (P = .003). Genotype frequencies of rs55710189 were found to be significantly different between patients with schizophrenia and control subjects (odds ratio = 6.89, 95% confidence interval = 1.91-24.90, P < .001 in the dominant model [C/T + T/T vs C/C], OR = 6.50, 95% confidence interval = 1.83-23.01, P < .001 in the log-additive model (C/T vs T/T vs C/C)]. In haplotype analysis, the TT haplotype for these 2 SNPs was significantly associated with schizophrenia (P < .001, χ2 = 12.231).Our findings suggest that specific ULK2 polymorphisms may be associated with susceptibility to schizophrenia in the Korean population.
Collapse
Affiliation(s)
- Won Sub Kang
- Department of Neuropsychiatry, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sang Min Lee
- Department of Neuropsychiatry, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dongkee Hwang
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hae Jeong Park
- Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jong Woo Kim
- Department of Neuropsychiatry, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Autophagy-related DjAtg1-1 plays critical role in planarian regeneration by regulating proliferation and cell death. Cell Tissue Res 2022; 388:273-286. [PMID: 35107621 DOI: 10.1007/s00441-022-03591-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 01/19/2022] [Indexed: 11/02/2022]
Abstract
Autophagy is an intracellular degradation process and plays key roles in energy recycle and homeostasis maintenance during planarian regeneration. Although planarians provide an ideal model organism for studying autophagy in vivo, the molecular mechanism of planarian autophagy is still unknown. Here, we identify three autophagy-related (Atg) gene 1 homologs from Dugesia japonica and study their roles in planarian regeneration. Both DjATG1-1 and DjATG1-2 proteins show homology to vertebrate unc-51 like autophagy activating kinase 1 (ULK1) and ULK2, DjATG1-3 shows homology to vertebrate ULK3. In contrast to the ubiquitously expressed DjAtg1-1 and DjAtg1-3, DjAtg1-2 is mainly expressed in the intestine branches and epidermis. All the three DjAtg1s can respond to planarian regeneration and starvation. Both DjAtg1-1 and DjAtg1-2 are expressed in the reproductive organs of the starved sexual worms. DjAtg1-1 or DjAtg1-3 RNAi leads to head lysis and death of starved planarians, accompanied by exhaustion of neoblasts. DjAtg1-1 RNAi causes autophagy and regeneration defects and decreases proliferation and cell death; both DjAtg1-2 and DjAtg1-3 RNAi cause no autophagy or regeneration defect but increase cell death during regeneration. Our findings uncover the roles of DjAtg1s in autophagy and regeneration of planarian and highlight the links between proliferation, cell death, and autophagy during regeneration.
Collapse
|
24
|
Dent P. Cell Signaling and Translational Developmental Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC7538147 DOI: 10.1016/b978-0-12-820472-6.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The relationships between drug pharmacodynamics and subsequent changes in cellular signaling processes are complex. Many in vitro cell signaling studies often use drug concentrations above physiologically safe drug levels achievable in a patient's plasma. Drug companies develop agents to inhibit or modify the activities of specific target enzymes, often without a full consideration that their compounds have additional unknown targets. These two negative sequelae, when published together, become impediments against successful developmental therapeutics and translation because this data distorts our understanding of signaling mechanisms and reduces the probability of successfully translating drug-based concepts from the bench to the bedside. This article will discuss cellular signaling in isolation and as it relates to extant single and combined therapeutic drug interventions. This will lead to a hypothetical series standardized sequential approaches describing a rigorous concept to drug development and clinical translation.
Collapse
|
25
|
Tornero-Écija A, Tábara LC, Bueno-Arribas M, Antón-Esteban L, Navarro-Gómez C, Sánchez I, Vincent O, Escalante R. A Dictyostelium model for BPAN disease reveals a functional relationship between the WDR45/WIPI4 homolog Wdr45l and Vmp1 in the regulation of autophagy-associated PtdIns3P and ER stress. Autophagy 2021; 18:661-677. [PMID: 34328055 PMCID: PMC9037511 DOI: 10.1080/15548627.2021.1953262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PROPPINs are conserved PtdIns3P-binding proteins required for autophagosome biogenesis that fold into a characteristic group of seven-bladed beta-propellers. Mutations in WDR45/WIPI4, a human member of this family, lead to BPAN, a rare form of neurodegeneration. We have generated mutants for the two PROPPIN proteins present in the model system Dictyostelium discoideum (Atg18 and Wdr45l) and characterized their function. Lack of Wdr45l greatly impairs autophagy, while Atg18 only causes subtle defects in the maturation of autolysosomes. The strong phenotype of the Wdr45l mutant is strikingly similar to that observed in Dictyostelium cells lacking Vmp1, an ER protein required for omegasome formation. Common phenotypes include impaired growth in axenic medium, lack of aggregation, and local enrichment of PtdIns3P as determined by the use of lipid reporters. In addition, Vmp1 and Wdr45l mutants show a chronically active response to ER stress. For both mutants, this altered PtdIns3P localization can be prevented by the additional mutation of the upstream regulator Atg1, which also leads to recovery of axenic growth and reduction of ER stress. We propose that, in addition to an autophagy defect, local autophagy-associated PtdIns3P accumulation might contribute to the pathogenesis of BPAN by disrupting ER homeostasis. The introduction of BPAN-associated mutations in Dictyostelium Wdr45l reveals the impact of pathogenic residues on the function and localization of the protein.
Collapse
Affiliation(s)
- Alba Tornero-Écija
- C.S.I.C./U.A.M., Instituto De Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | - Luis-Carlos Tábara
- C.S.I.C./U.A.M., Instituto De Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | - Miranda Bueno-Arribas
- C.S.I.C./U.A.M., Instituto De Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | - Laura Antón-Esteban
- C.S.I.C./U.A.M., Instituto De Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | | | - Irene Sánchez
- C.S.I.C./U.A.M., Instituto De Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | - Olivier Vincent
- C.S.I.C./U.A.M., Instituto De Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | - Ricardo Escalante
- C.S.I.C./U.A.M., Instituto De Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| |
Collapse
|
26
|
Du Y, Guo H, Guo L, Miao J, Ren H, Liu K, Ren L, He J, Wang X, Chen J, Li J, Wang Y, Wang J, Huang N. The regulatory effect of acetylation of HMGN2 and H3K27 on pyocyanin-induced autophagy in macrophages by affecting Ulk1 transcription. J Cell Mol Med 2021; 25:7524-7537. [PMID: 34278675 PMCID: PMC8335688 DOI: 10.1111/jcmm.16788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/05/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022] Open
Abstract
Pyocyanin (PYO) is a major virulence factor secreted by Pseudomonas aeruginosa, and autophagy is a crucial homeostatic mechanism for the interaction between the pathogens and the host. It remains unknown whether PYO leads to autophagy in macrophages by regulating histone acetylation. The high mobility group nucleosomal binding domain 2 (HMGN2) has been reported to regulate the PYO‐induced autophagy and oxidative stress in the epithelial cells; however, the underlying molecular mechanism has not been fully elucidated. In this study, PYO was found to induce autophagy in macrophages, and the mechanism might be correlated with the up‐regulation of HMGN2 acetylation (HMGN2ac) and the down‐regulation of H3K27 acetylation (H3K27ac) by modulation of the activities of acetyltransferases and deacetylases. Moreover, we further demonstrated that the up‐regulated HMGN2ac enhances its recruitment to the Ulk1 promoter, while the down‐regulation of H3K27ac reduces its recruitment to the Ulk1 promoter, thereby promoting or inhibiting the transcription of Ulk1. In conclusion, HMGN2ac and H3K27ac play regulatory roles in the PYO‐induced autophagy in macrophages.
Collapse
Affiliation(s)
- Yu Du
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.,Department of Anesthesiology, Nanchong Central Hospital, Second Clinical Medical Institution, North Sichuan Medical College, Nanchong, China
| | - Hongjun Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijuan Guo
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junming Miao
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hongyu Ren
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Keyun Liu
- Department of Physiology, School of Medicine, Hubei University for Nationalities, Enshi, China
| | - Laibin Ren
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jinchen He
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoying Wang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junli Chen
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jingyu Li
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Wang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Ji Wang
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ning Huang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Takhtarova TG, Khismatullina ZR, Panova LD, Panova AN. Ichthyosis (concept, pathohistology, clinical picture, treatment). VESTNIK DERMATOLOGII I VENEROLOGII 2021. [DOI: 10.25208/vdv1214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Ichthyosis is a skin disease that is hereditary, has pronounced symptoms in the form of a violation of the skin, and the presence of formations resembling fish scales. It is possible to distinguish different approaches to the definition of ichthyosis, based on the modern study of this issue. Ichthyosis is classified by type: congenital and acquired. Congenital ichthyosis has its own classification depending on the manifestation of changes in the skin, the course of the disease, concomitant pathologies. Congenital ichthyosis is divided into ordinary (vulgar autosomal dominant, simple) ichthyosis, lamellar ichthyosis (dry ichthyosiform erythroderma, "collodion child", lamellar ichthyosis), X-linked ichthyosis (ichthyosis associated with the X chromosome, blackening ichthyosis), congenital bullous ichthyosiform erythroderma (erythroderma Broca's disease, ichthyosiform epidermolytic hyperkeratosis), fetal ichthyosis (intrauterine ichthyosis, universal hyperkeratosis, "Harlequin fetus", congenital keratosis), other congenital ichthyosis. Ichthyosiform conditions (the so-called acquired ichthyosis) are divided into symptomatic, age-related (senile), discoid ichthyosis. The causes of acquired ichthyosis can be various diseases, taking medications, improper skin care, unbalanced nutrition. Each form of ichthyosis differs by the type of inheritance, prevalence in the population, clinical picture, verified by histological examination of skin biopsies and electron microscopic examination of the skin. It may be accompanied by seasonality of exacerbation of the clinical picture, association with other diseases (allergic, diseases of the gastrointestinal tract, congenital malformations). There is no specific treatment for ichthyosis. In systemic therapy, derivatives of vitamin A are used, keratolytics, as well as moisturizing and emollient agents are used for external treatment. The use of therapeutic baths, general ultraviolet irradiation is effective.
Collapse
|
28
|
Luo Z, Zeng A, Chen Y, He S, He S, Jin X, Li C, Mei W, Lu Q. Ligustilide inhibited Angiotensin II induced A7r5 cell autophagy via Akt/mTOR signaling pathway. Eur J Pharmacol 2021; 905:174184. [PMID: 34004211 DOI: 10.1016/j.ejphar.2021.174184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022]
Abstract
Autophagy is essential to vessel homeostasis and function in the cardiovascular system. Ligustilide (LIG) is one of the main active ingredients extracted from traditional Chinese medicines, such as Ligusticum chuanxiong, Angelica, and other umbelliferous plants, and reported to have cardiovascular protective effects. In this study, we explore the effects and the potential mechanism of ligustilide on the Ang II-induced autophagy in A7r5 cells. Our results showed that ligustilide inhibited the Ang II-induced autophagy in A7r5 cells and down regulated the expression of autophagy-related proteins LC3, ULK1, and Beclin-1. Ligustilide exerted a protective effect on the reduction of the concentrations of reactive oxygen species and Ca2+ and upregulated the nitric oxide concentration in A7r5 cells with Ang II-induced autophagy. Additionally, the analyses of network pharmacological targets and potential signal pathways indicated that the target of ligustilide to regulate autophagy was related to the Akt/mTOR signaling pathway. Furthermore, ligustilide could upregulate the expression of p-Akt and p-mTOR and inhibit the expression of LC3II in A7r5 cells with Ang II-induced autophagy. These findings showed that ligustilide inhibited the autophagic flux in A7r5 cells induced by Ang II via the activation of the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhenhui Luo
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ao Zeng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuankun Chen
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shumiao He
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Siqing He
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chunmei Li
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenjie Mei
- Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China
| | - Qun Lu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China.
| |
Collapse
|
29
|
Crawley O, Grill B. Autophagy in axonal and presynaptic development. Curr Opin Neurobiol 2021; 69:139-148. [PMID: 33940492 DOI: 10.1016/j.conb.2021.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 10/21/2022]
Abstract
The study of autophagy in the nervous system has predominantly centered on degeneration. Evidence is now cementing crucial roles for autophagy in neuronal development and growth, especially in axonal and presynaptic compartments. A picture is emerging that autophagy typically promotes the growth of axons and reduces presynaptic stability. Nonetheless, these are not rigid principles, and it remains unclear why autophagy does not always display these relationships during axonal and presynaptic development. Recent progress has identified mechanisms underlying spatiotemporal control of autophagy in neurons and begun to unravel how autophagy is integrated with other cellular processes, such as proteasomal degradation and axon guidance. Ultimately, understanding how autophagy is regulated and its role in the developing nervous system is key to comprehending how the nervous system assembles its stereotyped yet plastic configuration. It is also likely to inform how we think about neurodevelopmental disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliver Crawley
- Unidad de Neurobiología Celular y de Sistemas, Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, 03550, Spain.
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98199, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
30
|
Li D, Vogel P, Li-Harms X, Wang B, Kundu M. ATG14 and RB1CC1 play essential roles in maintaining muscle homeostasis. Autophagy 2021; 17:2576-2585. [PMID: 33794726 DOI: 10.1080/15548627.2021.1911549] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Defects in macroautophagy/autophagy are implicated in the pathogenesis of neuromuscular and heart diseases. To precisely define the roles of autophagy-related genes in skeletal and cardiac muscles, we generated muscle-specific rb1cc1- and atg14-conditional knockout (cKO) mice by using Ckm/Ckmm2-Cre and compared their phenotypes to those of ulk1 ulk2-conditional double-knockout (cDKO) mice. atg14-cKO mice developed hypertrophic cardiomyopathy, which was associated with abnormal accumulation of autophagic cargoes in the heart and early mortality. Skeletal muscles of both atg14-cKO and rb1cc1-cKO mice showed features of autophagic vacuolar myopathy with ubiquitin+ SQSTM1+ deposits, but only those of rb1cc1-cKO mice showed TARDBP/TDP-43+ pathology and other features of the inclusion body myopathy-like disease we previously described in ulk1 ulk2-cDKO mice. Herein, we highlight tissue-specific differences between skeletal and cardiac muscles in their reliance on core autophagy proteins and unique roles for ULK1-ULK2 and RB1CC1 among these proteins in the development of TARDBP+ pathology.ABBREVIATIONS:AVM: autophagic vacuolar myopathy; cDKO: conditional double knockout; cKO: conditional knockout; H&E: hematoxylin and eosin; IBM: inclusion body myopathy; mtDNA: mitochondrial DNA; PFA: paraformaldehyde; RNP: ribonucleoprotein; TBST: Tris-buffered saline with 0.2% Triton X-100.
Collapse
Affiliation(s)
- Dongfang Li
- Departments of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter Vogel
- Departments of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiujie Li-Harms
- Departments of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bo Wang
- Departments of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.,State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian Province, Fujian Province, China
| | - Mondira Kundu
- Departments of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
31
|
Long noncoding RNA H19 contributes to the proliferation and autophagy of glioma cells through mTOR/ULK1 pathway. Neuroreport 2021; 32:352-358. [PMID: 33661803 DOI: 10.1097/wnr.0000000000001602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Long noncoding RNA (LncRNA) H19 has been proven to be involved in many kinds of cancers including glioma, and a previous study has shown an autophagy regulation of H19. The mammalian target of rapamycin (mTOR) signaling pathway plays a key role in autophagy and Unc-51 like autophagy activating kinase 1 (ULK1) is also thought to be involved in autophagy signaling. In our study, we investigated the role of mTOR/ULK1 autophagy signaling in the H19-mediated promotion of glioma proliferation. Human glioma cells U87 and U251 and normal human astrocytes HA1800 were used in the study. First, the expression of H19 was determined in U87, U251, and HA1800 cells. Then, the cell proliferation and migration of glioma cells were detected, while the protein levels of main molecules of the mTOR/ULK1 pathway and autophagy-related proteins were also examined. Rapamycin, an inhibitor of mTOR, was used to further study the role of H19 in autophagy. We observed that overexpressed H19 promoted the proliferation and migration in glioma cells. The autophagy of U87 cells was suppressed when H19 was overexpressed and enhanced when H19 was silenced. H19 overexpression inhibited mTOR phosphorylation and promoted ULK1 phosphorylation. H19 promoted proliferation, migration, and autophagy by regulating mTOR signaling. In conclusion, we validate that H19 contributes to the proliferation and autophagy of glioma cells through the mTOR/ULK1 pathway.
Collapse
|
32
|
Kojima W, Yamano K, Kosako H, Imai K, Kikuchi R, Tanaka K, Matsuda N. Mammalian BCAS3 and C16orf70 associate with the phagophore assembly site in response to selective and non-selective autophagy. Autophagy 2021; 17:2011-2036. [PMID: 33499712 PMCID: PMC8386740 DOI: 10.1080/15548627.2021.1874133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Macroautophagy/autophagy is an intracellular degradation process that delivers cytosolic materials and/or damaged organelles to lysosomes. De novo synthesis of the autophagosome membrane occurs within a phosphatidylinositol-3-phosphate-rich region of the endoplasmic reticulum, and subsequent expansion is critical for cargo encapsulation. This process is complex, especially in mammals, with many regulatory factors. In this study, by utilizing PRKN (parkin RBR E3 ubiquitin protein ligase)-mediated mitochondria autophagy (mitophagy)-inducing conditions in conjunction with chemical crosslinking and mass spectrometry, we identified human BCAS3 (BCAS3 microtubule associated cell migration factor) and C16orf70 (chromosome 16 open reading frame 70) as novel proteins that associate with the autophagosome formation site during both non-selective and selective autophagy. We demonstrate that BCAS3 and C16orf70 form a complex and that their association with the phagophore assembly site requires both proteins. In silico structural modeling, mutational analyses in cells and in vitro phosphoinositide-binding assays indicate that the WD40 repeat domain in human BCAS3 directly binds phosphatidylinositol-3-phosphate. Furthermore, overexpression of the BCAS3-C16orf70 complex affects the recruitment of several core autophagy proteins to the phagophore assembly site. This study demonstrates regulatory roles for human BCAS3 and C16orf70 in autophagic activity. Abbreviations: AO: antimycin A and oligomycin; Ash: assembly helper; ATG: autophagy-related; BCAS3: BCAS3 microtubule associated cell migration factor; C16orf70: chromosome 16 open reading frame 70; DAPI: 4‘,6-diamidino-2-phenylindole; DKO: double knockout; DMSO: dimethyl sulfoxide; ER: endoplasmic reticulum; fluoppi: fluorescent-based technology detecting protein-protein interactions; FIS1: fission, mitochondrial 1; FKBP: FKBP prolyl isomerase family member 1C; FRB: FKBP-rapamycin binding; hAG: humanized azami-green; IP: immunoprecipitation; IRES: internal ribosome entry site; KO: knockout; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MFN2: mitofusin 2; MS: mass spectrometry; MT-CO2: mitochondrially encoded cytochrome c oxidase II; mtDNA: mitochondrial DNA; OPTN: optineurin; PFA: paraformaldehyde; PE: phosphatidylethanolamine; PtdIns3K: phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns(3,5)P2: phosphatidylinositol-3,5-bisphosphate; PINK1: PTEN induced kinase 1; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; PROPPIN: β-propellers that bind polyphosphoinositides; RB1CC1/FIP200: RB1 inducible coiled-coil 1; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like autophagy activating kinase 1; WDR45B/WIPI3: WD repeat domain 45B; WDR45/WIPI4: WD repeat domain 45; WIPI: WD repeat domain, phosphoinositide interacting; WT: wild type; ZFYVE1/DFCP1: zinc finger FYVE-type containing 1
Collapse
Affiliation(s)
- Waka Kojima
- Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Japan
| | - Koji Yamano
- Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Kenichiro Imai
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan.,Molecular Profiling Research Center for Drug Discovery (Molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Reika Kikuchi
- Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Keiji Tanaka
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Noriyuki Matsuda
- Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
33
|
Yang W, Li Y, Liu S, Sun W, Huang H, Zhang Q, Yan J. Inhibition of ULK1 promotes the death of leukemia cell in an autophagy irrelevant manner and exerts the antileukemia effect. Clin Transl Med 2021; 11:e282. [PMID: 33463048 PMCID: PMC7803353 DOI: 10.1002/ctm2.282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Wei Yang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.,Institute of Biomedical Engineering, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China
| | - Yunlei Li
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Shuai Liu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.,Department of Laboratory, Shenzhen Sami International Medical Center (Shenzhen Fourth People's Hospital), Shenzhen, China
| | - Weimin Sun
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Hualan Huang
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiqing Zhang
- Institute of Biomedical Engineering, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China
| | - Jie Yan
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
34
|
Lechauve C, Keith J, Khandros E, Fowler S, Mayberry K, Freiwan A, Thom CS, Delbini P, Romero EB, Zhang J, Motta I, Tillman H, Cappellini MD, Kundu M, Weiss MJ. The autophagy-activating kinase ULK1 mediates clearance of free α-globin in β-thalassemia. Sci Transl Med 2020; 11:11/506/eaav4881. [PMID: 31434755 DOI: 10.1126/scitranslmed.aav4881] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/26/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
In β-thalassemia, accumulated free α-globin forms intracellular precipitates that impair erythroid cell maturation and viability. Protein quality control systems mitigate β-thalassemia pathophysiology by degrading toxic free α-globin, although the associated mechanisms are poorly understood. We show that loss of the autophagy-activating Unc-51-like kinase 1 (Ulk1) gene in β-thalassemic mice reduces autophagic clearance of α-globin in red blood cell precursors and exacerbates disease phenotypes, whereas inactivation of the canonical autophagy-related 5 (Atg5) gene has relatively minor effects. Systemic treatment with the mTORC1 inhibitor rapamycin reduces α-globin precipitates and lessens pathologies in β-thalassemic mice via an ULK1-dependent pathway. Similarly, rapamycin reduces free α-globin accumulation in erythroblasts derived from CD34+ cells of β-thalassemic individuals. Our findings define a drug-regulatable pathway for ameliorating β-thalassemia.
Collapse
Affiliation(s)
- Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Julia Keith
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Eugene Khandros
- Department of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephanie Fowler
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kalin Mayberry
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abdullah Freiwan
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher S Thom
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paola Delbini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Emilio Boada Romero
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jingjing Zhang
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Irene Motta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Heather Tillman
- Departments of Pathology and Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M Domenica Cappellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Mondira Kundu
- Departments of Pathology and Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
35
|
Hwang DY, Eom JI, Jang JE, Jeung HK, Chung H, Kim JS, Cheong JW, Min YH. ULK1 inhibition as a targeted therapeutic strategy for FLT3-ITD-mutated acute myeloid leukemia. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:85. [PMID: 32393312 PMCID: PMC7212592 DOI: 10.1186/s13046-020-01580-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022]
Abstract
Background In acute myeloid leukemia (AML), internal tandem duplication mutations in the FLT3 tyrosine kinase receptor (FLT3-ITD) are associated with a dismal outcome. Although uncoordinated 51-like kinase 1 (ULK1), which plays a central role in the autophagy pathway, has emerged as a novel therapeutic target for various cancers, its role in FLT3-ITD AML remains elusive. In this study, we evaluated the effects of ULK1 inhibition on leukemia cell death in FLT3-ITD AML. Method We evaluated ULK1 expression and the levels of apoptosis and autophagy following ULK1 inhibition in FLT3-ITD AML cell lines and investigated the mechanism underlying apoptosis induced by ULK1 inhibition. Statistical analysis was performed using GraphPad Prism 4.0 (GraphPad Software Inc). Results FLT3-ITD AML cells showed significantly higher ULK1 expression than FLT3-wild-type (WT) AML cells. Two ULK1 inhibitors, MRT 68921 and SBI-0206965, induced apoptosis in FLT3-ITD AML cells, with relatively minimal effects on FLT3-WT AML cells and normal CD34-positive cells. Apoptosis induction by ULK1 inhibition was associated with caspase pathway activation. Interestingly, ULK1 inhibition paradoxically also induced autophagy, showing synergistic interaction with autophagy inhibitors. Hence, autophagy may act as a prosurvival mechanism in FLT3-ITD AML cells. FLT3-ITD protein degradation and inhibition of the ERK, AKT, and STAT5 pathways were also observed in FLT3-ITD AML cells following treatment with ULK1 inhibitors. Conclusion ULK1 is a viable drug target and ULK1 inhibition may represent a promising therapeutic strategy against FLT3-ITD AML.
Collapse
Affiliation(s)
- Doh Yu Hwang
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Ju-In Eom
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Ji Eun Jang
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Hoi-Kyung Jeung
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Yoo Hong Min
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
36
|
Singha B, Laski J, Ramos Valdés Y, Liu E, DiMattia GE, Shepherd TG. Inhibiting ULK1 kinase decreases autophagy and cell viability in high-grade serous ovarian cancer spheroids. Am J Cancer Res 2020; 10:1384-1399. [PMID: 32509386 PMCID: PMC7269771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/27/2020] [Indexed: 06/11/2023] Open
Abstract
Metastasis in high-grade serous ovarian cancer (HGSOC) occurs through an unconventional route that involves exfoliation of cancer cells from primary tumors and peritoneal dissemination via multicellular clusters or spheroids. Previously, we demonstrated autophagy induction in HGSOC spheroids grown in vitro and in spheroids collected from ovarian cancer patient ascites; thus, we speculate that autophagy may contribute to spheroid cell survival and overall disease progression. Hence, in this study we sought to evaluate whether ULK1 (unc-51-like kinase-1), a serine-threonine kinase critical for stress-induced autophagy, is important for autophagy regulation in HGSOC spheroids. We demonstrate that HGSOC spheroids have increased ULK1 protein expression that parallels autophagy activation. ULK1 knockdown increased p62 accumulation and decreased LC3-II/I ratio in HGSOC spheroids. In addition, knocking down ATG13, a protein that regulates ULK1 activity via complex formation, phenocopied our ULK1 knockdown results. HGSOC spheroids were blocked in autophagic flux due to ULK1 and ATG13 knockdown as determined by an mCherry-eGFP-LC3B fluorescence reporter. These observations were recapitulated when HGSOC spheroids were treated with an ULK1 kinase inhibitor, MRT68921. Autophagy regulation in normal human fallopian tube epithelial FT190 cells, however, may bypass ULK1, since MRT68921 reduced viability in HGSOC spheroids but not in FT190 cells. Interestingly, ULK1 mRNA expression is negatively correlated with patient survival among stage III and stage IV serous ovarian cancer patients. As we observed using established HGSOC cell lines, cultured spheroids using our new, patient-derived HGSOC cells were also sensitive to ULK1 inhibition and demonstrated reduced cell viability to MRT68921 treatment. These results demonstrate the importance of ULK1 for autophagy induction in HGSOC spheroids and therefore justifies further evaluation of MRT68921, and other novel ULK1 inhibitors, as potential therapeutics against metastatic HGSOC.
Collapse
Affiliation(s)
- Bipradeb Singha
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer ProgramLondon, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western UniversityLondon, Ontario, Canada
| | - Jeremi Laski
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer ProgramLondon, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western UniversityLondon, Ontario, Canada
| | - Yudith Ramos Valdés
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer ProgramLondon, Ontario, Canada
| | - Elaine Liu
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer ProgramLondon, Ontario, Canada
| | - Gabriel E DiMattia
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer ProgramLondon, Ontario, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western UniversityLondon, Ontario, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, The University of Western OntarioLondon, Ontario, Canada
| | - Trevor G Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer ProgramLondon, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western UniversityLondon, Ontario, Canada
- Department of Obstetrics & Gynaecology, Schulich School of Medicine and Dentistry, Western UniversityLondon, Ontario, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, The University of Western OntarioLondon, Ontario, Canada
| |
Collapse
|
37
|
Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B 2020; 10:569-581. [PMID: 32322463 PMCID: PMC7161711 DOI: 10.1016/j.apsb.2019.10.003] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023] Open
Abstract
Autophagy, defined as a scavenging process of protein aggregates and damaged organelles mediated by lysosomes, plays a significant role in the quality control of macromolecules and organelles. Since protein kinases are integral to the autophagy process, it is critically important to understand the role of kinases in autophagic regulation. At present, intervention of autophagic processes by small-molecule modulators targeting specific kinases has becoming a reasonable and prevalent strategy for treating several varieties of human disease, especially cancer. In this review, we describe the role of some autophagy-related kinase targets and kinase-mediated phosphorylation mechanisms in autophagy regulation. We also summarize the small-molecule kinase inhibitors/activators of these targets, highlighting the opportunities of these new therapeutic agents.
Collapse
Key Words
- 4E-BP1, eukaryotic translation initiation factor 4E-binding protein
- AKT1, AKT serine/threonine kinase 1
- AMBRA1, autophagy/beclin-1 regulator 1
- AMPK, AMP-activated protein kinase
- ARF, auxin response factor gene
- ATG, autophagy-related protein
- Autophagy
- Autophagy-related kinase
- CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2
- DAPK, death associated protein kinase
- FIP200, FAK family kinase-interacting protein of 200 kDa
- GAP, GTPase-activating protein
- GO, gene ontology
- GSK3α, glycogen synthase kinase 3 alpha
- HMGB1, high mobility group protein B1
- Human disease therapy
- JNK1, C-Jun N-terminal kinase
- LC3, microtubule-associated protein 1 light chain 3
- LKB1, serine/threonine-protein kinase stk11
- LPS, lipopolysaccharide
- LRRK2, leucine rich repeat kinase 2
- PD, Parkinson's disease
- PI, phosphatidylinositol
- PI3 kinase, phosphoinositide 3-kinase
- PI3P, phosphatidylinositol triphosphate
- PIM2, proviral insertion in murine lymphomas 2
- PINK1, PTEN-induced putative kinase 1
- PIP2, phosphatidylinositol-4,5-bisphosphate
- PKACα, a protein kinase cAMP-activated catalytic subunit alpha
- PKCα, protein kinase C alpha type
- PKD1, polycystin-1
- PPIs, protein–protein interactions
- PROTAC, proteolysis targeting chimeras
- PTMs, post-translational modifications
- Phosphorylation
- Protein kinases
- Rheb, the RAS homolog enriched in brain
- Small-molecule kinase inhibitors/activators
- TAK1, transforming growth factor activated kinase-1
- TFEB, transcription factor EB
- TNBC, triple-negative breast cancer
- TSC1/2, tuberous sclerosis complex proteins 1/2
- ULK complex, ULK1–mATG13–FIP200–ATG101 complex
- ULK1, unc-51-like kinase 1
- UVRAG, ultraviolet resistance-associated gene
- mTOR, mammalian target of rapamycin
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Honggang Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
38
|
Tyra LK, Nandi N, Tracy C, Krämer H. Yorkie Growth-Promoting Activity Is Limited by Atg1-Mediated Phosphorylation. Dev Cell 2020; 52:605-616.e7. [PMID: 32032548 DOI: 10.1016/j.devcel.2020.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/26/2019] [Accepted: 01/09/2020] [Indexed: 01/31/2023]
Abstract
The expression of multiple growth-promoting genes is coordinated by the transcriptional co-activator Yorkie with its major regulatory input provided by the Hippo-Warts kinase cascade. Here, we identify Atg1/ULK1-mediated phosphorylation of Yorkie as an additional inhibitory input independent of the Hippo-Warts pathway. Two serine residues in Yorkie, S74 and S97, are Atg1/ULK1 consensus target sites and are phosphorylated by ULK1 in vitro, thereby preventing its binding to Scalloped. In vivo, gain of function of Atg1, or its activator Acinus, caused elevated Yorkie phosphorylation and inhibited Yorkie's growth-promoting activity. Loss of function of Atg1 or Acinus raised expression of Yorkie target genes and increased tissue size. Unlike Atg1's role in autophagy, Atg1-mediated phosphorylation of Yorkie does not require Atg13. Atg1 is activated by starvation and other cellular stressors and therefore can impose temporary stress-induced constraints on the growth-promoting gene networks under the control of Hippo-Yorkie signaling.
Collapse
Affiliation(s)
- Lauren K Tyra
- Peter O'Donnell Jr. Brain Institute, Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Nilay Nandi
- Peter O'Donnell Jr. Brain Institute, Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Charles Tracy
- Peter O'Donnell Jr. Brain Institute, Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Helmut Krämer
- Peter O'Donnell Jr. Brain Institute, Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
39
|
Yu F, Hao P, Ye C, Feng Y, Pang K, Yu X. NlATG1 Gene Participates in Regulating Autophagy and Fission of Mitochondria in the Brown Planthopper, Nilaparvata lugens. Front Physiol 2020; 10:1622. [PMID: 32082181 PMCID: PMC7004972 DOI: 10.3389/fphys.2019.01622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/24/2019] [Indexed: 01/06/2023] Open
Abstract
Autophagy plays multiple roles in regulating various physiological processes in cells. However, we currently lack a systematic analysis of autophagy and the autophagy-related gene 1 ATG1 in the brown planthopper (BPH, Nilaparvata lugens), one of the most destructive of the insect pests of rice. In this study, the full-length cDNA of an autophagy-related gene, NlATG1, was cloned from BPH. Real-time qPCR (RT-qPCR) revealed that this NlATG1 gene was expressed differently across developmental stages, at higher levels in nymphs but lower levels in adults. RNA interference with dsNlATG1 significantly decreased the mRNA level of the target gene to 14.6% at day 4 compared with that of the dsGFP control group. The survival of the dsNlATG1-treated group decreased significantly from day 4 onward, dropping to 48.3% on day 8. Examination using transmission electron microscopy (TEM) showed that epithelial cells of the BPH’s midgut in the dsNlATG1-treated group had less autophagic vacuoles than did the dsGFP control, and knockdown of NlATG1 clearly inhibited the starvation-induced autophagy response in this insect. RNA interference of NlATG1 upregulated the NlFis1 gene involved in mitochondrial fission, leading to reductions in mitochondrial width and area. Furthermore, knockdown of NlATG1 also decreased the ATP content and accumulation of glycogen. Together, these results demonstrate that the NlATG1 gene participates in regulating autophagy and fission of mitochondria in the brown planthopper, making it a potentially promising target for pest control given its key role in autophagy, including maintaining the normal structure and function of mitochondria.
Collapse
Affiliation(s)
- Feifei Yu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Peiying Hao
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Chenglong Ye
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Yalin Feng
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Kun Pang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Xiaoping Yu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou, China
| |
Collapse
|
40
|
Abstract
Autophagy is cellular recycling process that plays a complex role in cancer. Pre-clinical studies indicating a pro-tumorigenic role of autophagy have led to the launch of dozens of clinical trials combining autophagy inhibition with other standard of care therapies in different tumor types. A recent publication utilized a novel, acute, CRISPR/Cas9 assay to identify cancer cell lines that are exquisitely sensitive to loss of core autophagy genes within the first 7 days. However, weeks later, rare populations of originally autophagy dependent cells were found that could circumvent autophagy inhibition. Analysis of these rare clones revealed that in the process of circumventing loss of autophagy, the cells upregulated NRF2 signaling to maintain protein homeostasis and consequently become more sensitive to proteasome inhibition as well as knock down of NRF2. This review highlights recent publications regarding the role of autophagy in cancer and potential mechanisms cancer cells may be able to commandeer to circumvent autophagy inhibition. We hope to make significant clinical advances by understanding if and when cancer cells will become resistant to autophagy inhibition, and pre-clinical studies may be able to provide insight into the best combinatorial therapies to prevent tumor relapse while on autophagy inhibitors.
Collapse
Affiliation(s)
- Christina G Towers
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
41
|
Autophagy is inhibited by ubiquitin ligase activity in the nervous system. Nat Commun 2019; 10:5017. [PMID: 31676756 PMCID: PMC6825199 DOI: 10.1038/s41467-019-12804-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
Autophagy is an intracellular catabolic process prominent in starvation, aging and disease. Neuronal autophagy is particularly important, as it affects the development and function of the nervous system, and is heavily implicated in neurodegenerative disease. Nonetheless, how autophagy is regulated in neurons remains poorly understood. Using an unbiased proteomics approach, we demonstrate that the primary initiator of autophagy, the UNC-51/ULK kinase, is negatively regulated by the ubiquitin ligase RPM-1. RPM-1 ubiquitin ligase activity restricts UNC-51 and autophagosome formation within specific axonal compartments, and exerts effects broadly across the nervous system. By restraining UNC-51 activity, RPM-1 inhibits autophagosome formation to affect axon termination, synapse maintenance and behavioral habituation. These results demonstrate how UNC-51 and autophagy are regulated subcellularly in axons, and unveils a mechanism for restricting initiation of autophagy across the nervous system. Our findings have important implications beyond nervous system development, given growing links between altered autophagy regulation and neurodegenerative diseases.
Collapse
|
42
|
Cancer Cells Upregulate NRF2 Signaling to Adapt to Autophagy Inhibition. Dev Cell 2019; 50:690-703.e6. [PMID: 31378590 DOI: 10.1016/j.devcel.2019.07.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/26/2019] [Accepted: 07/03/2019] [Indexed: 12/24/2022]
Abstract
While autophagy is thought to be an essential process in some cancer cells, it is unknown if or how such cancer cells can circumvent autophagy inhibition. To address this, we developed a CRISPR/Cas9 assay with dynamic live-cell imaging to measure acute effects of knockout (KO) of autophagy genes compared to known essential and non-essential genes. In some cancer cells, autophagy is as essential for cancer cell growth as mRNA transcription or translation or DNA replication. However, even these highly autophagy-dependent cancer cells evolve to circumvent loss of autophagy by upregulating NRF2, which is necessary and sufficient for autophagy-dependent cells to circumvent ATG7 KO and maintain protein homeostasis. Importantly, however, this adaptation increases susceptibly to proteasome inhibitors. These studies identify a common mechanism of acquired resistance to autophagy inhibition and show that selection to avoid tumor cell dependency on autophagy creates new, potentially actionable cancer cell susceptibilities.
Collapse
|
43
|
Saleiro D, Platanias LC. Interferon signaling in cancer. Non-canonical pathways and control of intracellular immune checkpoints. Semin Immunol 2019; 43:101299. [PMID: 31771762 PMCID: PMC8177745 DOI: 10.1016/j.smim.2019.101299] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/11/2019] [Indexed: 01/01/2023]
Abstract
The interferons (IFNs) are cytokines with important antineoplastic and immune modulatory effects. These cytokines have been conserved through evolution as important elements of the immune surveillance against cancer. Despite this, defining their precise and specific roles in the generation of antitumor responses remains challenging. Emerging evidence suggests the existence of previously unknown roles for IFNs in the control of the immune response against cancer that may redefine our understanding on how these cytokines function. Beyond the engagement of classical JAK-STAT signaling pathways that promote transcription and expression of gene products, the IFNs engage multiple other signaling cascades to generate products that mediate biological responses and outcomes. There is recent emerging evidence indicating that IFNs control the expression of both traditional immune checkpoints like the PD-L1/PD1 axis, but also less well understood "intracellular" immune checkpoints whose targeting may define new approaches for the treatment of malignancies.
Collapse
Affiliation(s)
- Diana Saleiro
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, 303 East Superior Ave., Chicago, IL 60611, USA
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, 303 East Superior Ave., Chicago, IL 60611, USA; Department of Medicine, Jesse Brown Veterans Affairs Medical Center, 820 S. Damen Ave., Chicago, IL 60612, USA.
| |
Collapse
|
44
|
Wang B, Maxwell BA, Joo JH, Gwon Y, Messing J, Mishra A, Shaw TI, Ward AL, Quan H, Sakurada SM, Pruett-Miller SM, Bertorini T, Vogel P, Kim HJ, Peng J, Taylor JP, Kundu M. ULK1 and ULK2 Regulate Stress Granule Disassembly Through Phosphorylation and Activation of VCP/p97. Mol Cell 2019; 74:742-757.e8. [PMID: 30979586 PMCID: PMC6859904 DOI: 10.1016/j.molcel.2019.03.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/08/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022]
Abstract
Disturbances in autophagy and stress granule dynamics have been implicated as potential mechanisms underlying inclusion body myopathy (IBM) and related disorders. Yet the roles of core autophagy proteins in IBM and stress granule dynamics remain poorly characterized. Here, we demonstrate that disrupted expression of the core autophagy proteins ULK1 and ULK2 in mice causes a vacuolar myopathy with ubiquitin and TDP-43-positive inclusions; this myopathy is similar to that caused by VCP/p97 mutations, the most common cause of familial IBM. Mechanistically, we show that ULK1/2 localize to stress granules and phosphorylate VCP, thereby increasing VCP's activity and ability to disassemble stress granules. These data suggest that VCP dysregulation and defective stress granule disassembly contribute to IBM-like disease in Ulk1/2-deficient mice. In addition, stress granule disassembly is accelerated by an ULK1/2 agonist, suggesting ULK1/2 as targets for exploiting the higher-order regulation of stress granules for therapeutic intervention of IBM and related disorders.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brian A Maxwell
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Joung Hyuck Joo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Youngdae Gwon
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - James Messing
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Ashutosh Mishra
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Timothy I Shaw
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amber L Ward
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Honghu Quan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sadie Miki Sakurada
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Tulio Bertorini
- Department of Neurology, University of Tennessee Heath Science Center, Memphis, TN 38163, USA
| | - Peter Vogel
- Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hong Joo Kim
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - J Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Mondira Kundu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
45
|
Shimamura S, Miyazaki T, Tashiro M, Takazono T, Saijo T, Yamamoto K, Imamura Y, Izumikawa K, Yanagihara K, Kohno S, Mukae H. Autophagy-Inducing Factor Atg1 Is Required for Virulence in the Pathogenic Fungus Candida glabrata. Front Microbiol 2019; 10:27. [PMID: 30761093 PMCID: PMC6362428 DOI: 10.3389/fmicb.2019.00027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/09/2019] [Indexed: 01/09/2023] Open
Abstract
Candida glabrata is one of the leading causes of candidiasis and serious invasive infections in hosts with weakened immune systems. C. glabrata is a haploid budding yeast that resides in healthy hosts. Little is known about the mechanisms of C. glabrata virulence. Autophagy is a ‘self-eating’ process developed in eukaryotes to recycle molecules for adaptation to various environments. Autophagy is speculated to play a role in pathogen virulence by supplying sources of essential proteins for survival in severe host environments. Here, we investigated the effects of defective autophagy on C. glabrata virulence. Autophagy was induced by nitrogen starvation and hydrogen peroxide (H2O2) in C. glabrata. A mutant strain lacking CgAtg1, an autophagy-inducing factor, was generated and confirmed to be deficient for autophagy. The Cgatg1Δ strain was sensitive to nitrogen starvation and H2O2, died rapidly in water without any nutrients, and showed high intracellular ROS levels compared with the wild-type strain and the CgATG1-reconstituted strain in vitro. Upon infecting mouse peritoneal macrophages, the Cgatg1Δ strain showed higher mortality from phagocytosis by macrophages. Finally, in vivo experiments were performed using two mouse models of disseminated candidiasis and intra-abdominal candidiasis. The Cgatg1Δ strain showed significantly decreased CFUs in the organs of the two mouse models. These results suggest that autophagy contributes to C. glabrata virulence by conferring resistance to unstable nutrient environments and immune defense of hosts, and that Atg1 is a novel fitness factor in Candida species.
Collapse
Affiliation(s)
- Shintaro Shimamura
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Taiga Miyazaki
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan.,Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masato Tashiro
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan.,Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomomi Saijo
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Kazuko Yamamoto
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Yoshifumi Imamura
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shigeru Kohno
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| |
Collapse
|
46
|
Autophagy: An Essential Degradation Program for Cellular Homeostasis and Life. Cells 2018; 7:cells7120278. [PMID: 30572663 PMCID: PMC6315530 DOI: 10.3390/cells7120278] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a lysosome-dependent cellular degradation program that responds to a variety of environmental and cellular stresses. It is an evolutionarily well-conserved and essential pathway to maintain cellular homeostasis, therefore, dysfunction of autophagy is closely associated with a wide spectrum of human pathophysiological conditions including cancers and neurodegenerative diseases. The discovery and characterization of the kingdom of autophagy proteins have uncovered the molecular basis of the autophagy process. In addition, recent advances on the various post-translational modifications of autophagy proteins have shed light on the multiple layers of autophagy regulatory mechanisms, and provide novel therapeutic targets for the treatment of the diseases.
Collapse
|
47
|
Hu L, Zhang R, Yuan Q, Gao Y, Yang MQ, Zhang C, Huang J, Sun Y, Yang W, Yang JY, Min ZL, Cheng J, Deng Y, Hu X. The emerging role of microRNA-4487/6845-3p in Alzheimer's disease pathologies is induced by Aβ25-35 triggered in SH-SY5Y cell. BMC SYSTEMS BIOLOGY 2018; 12:119. [PMID: 30547775 PMCID: PMC6293494 DOI: 10.1186/s12918-018-0633-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Accumulation of amyloid β-peptide (Aβ) is implicated in the pathogenesis and development of Alzheimer’s disease (AD). Neuron-enriched miRNA was aberrantly regulated and may be associated with the pathogenesis of AD. However, regarding whether miRNA is involved in the accumulation of Aβ in AD, the underlying molecule mechanism remains unclear. Therefore, we conduct a systematic identification of the promising role of miRNAs in Aβ deposition, and shed light on the molecular mechanism of target miRNAs underlying SH-SY5Y cells treated with Aβ-induced cytotoxicity. Results Statistical analyses of microarray data revealed that 155 significantly upregulated and 50 significantly downregulated miRNAs were found on the basis of log2 | Fold Change | ≥ 0.585 and P < 0.05 filter condition through 2588 kinds of mature miRNA probe examined. PCR results show that the expression change trend of the selected six miRNAs (miR-6845-3p, miR-4487, miR-4534, miR-3622-3p, miR-1233-3p, miR-6760-5p) was consistent with the results of the gene chip. Notably, Aβ25–35 downregulated hsa-miR-4487 and upregulated hsa-miR-6845-3p in SH-SY5Y cell lines associated with Aβ-mediated pathophysiology. Increase of hsa-miR-4487 could inhibit cells apoptosis, and diminution of hsa-miR-6845-3p could attenuate axon damage mediated by Aβ25–35 in SH-SY5Y. Conclusions Together, these findings suggest that dysregulation of hsa-miR-4487 and hsa-miR-6845-3p contributed to the pathogenesis of AD associated with Aβ25–35 mediated by triggering cell apoptosis and synaptic dysfunction. It might be beneficial to understand the pathogenesis and development of clinical diagnosis and treatment of AD. Further, our well-designed validation studies will test the miRNAs signature as a prognostication tool associated with clinical outcomes in AD.
Collapse
Affiliation(s)
- Ling Hu
- Department of Anesthesiology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430064, China.,Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei Province, China
| | - Rong Zhang
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei Province, China
| | - Qiong Yuan
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei Province, China
| | - Yinping Gao
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Mary Q Yang
- MidSouth Bioinformatics Center, Department of Information Science, George Washington Donaghey College of Engineering and Information Technology and Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR, 72204, USA
| | - Chunxiang Zhang
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei Province, China.,Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama, Birmingham, 35201, USA
| | - Jiankun Huang
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Yufei Sun
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - William Yang
- MidSouth Bioinformatics Center, Department of Information Science, George Washington Donaghey College of Engineering and Information Technology and Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR, 72204, USA
| | - Jack Y Yang
- MidSouth Bioinformatics Center, Department of Information Science, George Washington Donaghey College of Engineering and Information Technology and Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR, 72204, USA
| | - Zhen-Li Min
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei Province, China
| | - Jing Cheng
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei Province, China
| | - Youping Deng
- Bioinformatics Core, Department of Complementary & Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA.
| | - Xiamin Hu
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
48
|
Colecchia D, Dapporto F, Tronnolone S, Salvini L, Chiariello M. MAPK15 is part of the ULK complex and controls its activity to regulate early phases of the autophagic process. J Biol Chem 2018; 293:15962-15976. [PMID: 30131341 DOI: 10.1074/jbc.ra118.002527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Autophagy, a pathway for bulk protein degradation and removal of damaged organelles, represents one of the major responses of cells to stress, thereby exerting a strict control on their correct functioning. Consequently, this process has been involved in the pathogenesis and therapeutic responses of several human diseases. Mitogen-activated protein (MAP) kinase 15 (MAPK15) is an atypical member of the MAP kinase family that recently emerged as a key modulator of autophagy and, through this, of cell transformation. Still, no information is available about signaling pathways mediating the effect of MAPK15 on this process, nor is it known which phase of autophagosome biogenesis is affected by this MAP kinase. Here, we demonstrate that MAPK15 stimulated 5'-AMP-activated protein kinase-dependent activity of UNC-51-like kinase 1 (ULK1), the only protein kinase among the ATG-related proteins, toward downstream substrates and signaling intermediates. Importantly, MAPK15 directly interacted with the ULK1 complex and mediated ULK1 activation induced by starvation, a classical stimulus for the autophagic process. In turn, ULK1 and its highly homologous protein ULK2 are able to transduce MAPK15 signals stimulating early phases of autophagosomal biogenesis in a multikinase cascade that offers numerous potential targets for future therapeutic intervention in cancer and other autophagy-related human diseases.
Collapse
Affiliation(s)
- David Colecchia
- From the Consiglio Nazionale delle Ricerche (CNR), Istituto di Fisiologia Clinica (IFC), Siena 53100.,the Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Core Research Laboratory (CRL), Unit "Signal Transduction," Siena 53100
| | - Francesca Dapporto
- the Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Core Research Laboratory (CRL), Unit "Signal Transduction," Siena 53100.,the Università degli Studi di Siena, Siena 53100, and
| | - Serena Tronnolone
- the Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Core Research Laboratory (CRL), Unit "Signal Transduction," Siena 53100
| | - Laura Salvini
- the Toscana Life Sciences Foundation, Siena 53100, Italy
| | - Mario Chiariello
- From the Consiglio Nazionale delle Ricerche (CNR), Istituto di Fisiologia Clinica (IFC), Siena 53100, .,the Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Core Research Laboratory (CRL), Unit "Signal Transduction," Siena 53100
| |
Collapse
|
49
|
Fan K, Lin L, Ai Q, Wan J, Dai J, Liu G, Tang L, Yang Y, Ge P, Jiang R, Zhang L. Lipopolysaccharide-Induced Dephosphorylation of AMPK-Activated Protein Kinase Potentiates Inflammatory Injury via Repression of ULK1-Dependent Autophagy. Front Immunol 2018; 9:1464. [PMID: 29988556 PMCID: PMC6026648 DOI: 10.3389/fimmu.2018.01464] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/12/2018] [Indexed: 12/14/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a crucial metabolic regulator with profound modulatory activities on inflammation. Although the anti-inflammatory benefits of AMPK activators were well documented in experimental studies, the pathological significance of endogenous AMPK in inflammatory disorders largely remains unknown. This study investigated the phosphorylation status of endogenous AMPK and the potential roles of AMPK in mice with lipopolysaccharide (LPS)-induced lethal inflammation. The results indicated that LPS dose-dependently decreased the phosphorylation level of AMPK and its target protein acetyl-CoA carboxylase (ACC). Reactivation of AMPK with the AMPK activator A-769662 suppressed LPS-induced elevation of interleukin 6, alleviated histological abnormalities in lung and improved the survival of LPS-challenged mice. Treatment with A-769662 restored LPS-induced suppression of autophagy, inhibition of autophagy by 3-MA reversed the beneficial effects of A-769662. Treatment with A-769662 suppressed LPS-induced activation of mammalian target of rapamycin (mTOR), co-administration of mTOR activator abolished the beneficial effects of A-769662, and the suppressive effects of A-769662 on uncoordinated-51-like kinase 1 (ULK1) phosphorylation. Inhibition of ULK1 removed the beneficial effects of A-769662. These data indicated that LPS-induced dephosphorylation of AMPK could result in weakened inhibition of mTOR and repression of ULK1-dependent autophagy, which might potentiate the development of LPS-induced inflammatory injury. These data suggest that pharmacological restoration of AMPK activation might be a beneficial approach for the intervention of inflammatory disorders.
Collapse
Affiliation(s)
- Kerui Fan
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Ling Lin
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Qing Ai
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jie Dai
- Hospital of Chongqing University of Arts and Sciences, Chongqing, China
| | - Gang Liu
- Department of Emergency, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Pu Ge
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
50
|
Low Autophagy (ATG) Gene Expression Is Associated with an Immature AML Blast Cell Phenotype and Can Be Restored during AML Differentiation Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1482795. [PMID: 29743969 PMCID: PMC5878891 DOI: 10.1155/2018/1482795] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/21/2017] [Accepted: 12/31/2017] [Indexed: 02/06/2023]
Abstract
Autophagy is an intracellular degradation system that ensures a dynamic recycling of a variety of building blocks required for self-renewal, homeostasis, and cell survival under stress. We used primary acute myeloid leukemia (AML) samples and human AML cell lines to investigate the regulatory mechanisms of autophagy and its role in AML differentiation. We found a significantly lower expression of key autophagy- (ATG-) related genes in primary AML as compared to healthy granulocytes, an increased autophagic activity during all-trans retinoic acid- (ATRA-) induced neutrophil differentiation, and an impaired AML differentiation upon inhibition of ATG3, ATG4D, and ATG5. Supporting the notion of noncanonical autophagy, we found that ATRA-induced autophagy was Beclin1-independent compared to starvation- or arsenic trioxide- (ATO-) induced autophagy. Furthermore, we identified PU.1 as positive transcriptional regulator of ATG3, ATG4D, and ATG5. Low PU.1 expression in AML may account for low ATG gene expression in this disease. Low expression of the autophagy initiator ULK1 in AML can partially be attributed to high expression of the ULK1-targeting microRNA-106a. Our data clearly suggest that granulocytic AML differentiation relies on noncanonical autophagy pathways and that restoring autophagic activity might be beneficial in differentiation therapies.
Collapse
|