1
|
Yao J, Sun X, Chen Y, Xu X, Feng J, Zhang M, Liu X, Shi X. Histone deacetylase 6 inhibition attenuates pathological cardiac hypertrophy by promoting autophagy through MAP1LC3B ubiquitination. J Pathol 2025; 266:217-229. [PMID: 40212005 DOI: 10.1002/path.6419] [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: 11/08/2024] [Revised: 01/17/2025] [Accepted: 02/24/2025] [Indexed: 05/08/2025]
Abstract
Cardiac hypertrophy is an adaptive response of the heart to pathological stimuli that may lead to cardiac dysfunction and heart failure. Histone deacetylase 6 (HDAC6) participates in the progression of multiple cardiovascular diseases, including chronic hypertension, ischemic stroke, and acute cardiac injury. A delicate balance of autophagy regulates heart homeostasis, whereas dysregulated autophagy is involved in myocardial hypertrophy. However, whether HDAC6 participates in pathological cardiac hypertrophy by regulating autophagy remains unclear. In this paper, we report for the first time that HDAC6 is involved in isoproterenol (ISO)-induced pathological cardiac hypertrophy by interacting with and ubiquitinating MAP1LC3B. First, the expression level of HDAC6 was found to be increased in cardiac hypertrophy models induced by ISO. HDAC6 overexpression promoted the expression of hypertrophic genes and enhanced cell surface area. Conversely, HDAC6 inhibition attenuated ISO-induced hypertrophic responses. Mechanistically, HDAC6 promoted hypertrophic responses by negatively regulating autophagy. Furthermore, HDAC6 interacted with MAP1LC3B and mediated its monoubiquitination, thereby contributing to reduced MAP1LC3B levels and impaired autophagy. Inhibition of HDAC6 activity in mice abrogated the hypertrophic effects of ISO by restoring MAP1LC3B expression. In summary, our data demonstrate that HDAC6 participates in ISO-induced cardiac hypertrophy by limiting the availability of MAP1LC3B and suppressing autophagy. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Yousheng Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xuan Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Junxiao Feng
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Mingming Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xiangdong Liu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| |
Collapse
|
2
|
Cheng X, Kang L, Liu J, Wang Q, Zhang Z, Zhang L, Xie Y, Chang L, Zeng D, Tian L, Zhang L, Xu P, Li Y. Proteomics and phosphoproteomics revealed dysregulated kinases and potential therapy for liver fibrosis. Mol Cell Proteomics 2025:100991. [PMID: 40368138 DOI: 10.1016/j.mcpro.2025.100991] [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: 08/23/2024] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025] Open
Abstract
Liver fibrosis is the initial stage of most liver diseases, and it is also a pathological process involving the liver in the late stages of many metabolic diseases. Therefore, it is important to systematically understand the pathological mechanism of liver fibrosis and seek therapeutic approaches for intervention and treatment of liver fibrosis. Disordered proteins and their post-translational modifications, such as phosphorylation, play vital roles in the occurrence and development of liver fibrosis. However, the regulatory mechanisms that govern this process remain poorly understood. In this study, we analyzed and quantified the liver proteome and phosphoproteome of CCl4-induced early liver fibrosis model in mice. Proteomic analysis revealed that the pathways involved in extracellular matrix (ECM) recombination, collagen formation, metabolism and other related disorders, and protein phosphorylation modification pathways were also significantly enriched. In addition, western blotting and phosphoproteomics demonstrated that phosphorylation levels were elevated in the context of liver fibrosis. A total of 13,152 phosphosites were identified, with 952 sites increased while only 156 ones decreased. Furthermore, the upregulated phosphorylation sites, which exhibited no change at the proteome level mainly shared a common [xxxSPxxx] motif. Consequently, the kinases-substrates analysis ascertained the overactive kinases of these up-regulated substrates, which ultimately led to the identification of 13 significantly altered kinases within this dataset. These kinases were mainly catalogued into the STE, CMGC, and CAMK kinase families. Among them, STK4, GSK3α and CDK11B were subsequently validated though cellular and animal experiments, and the results demonstrated that their inhibitors could effectively reduce the activation of hepatic stellate cells and ECM production. These kinases may represent potential therapeutic targets for liver fibrosis, and their inhibitors may serve as promising anti- hepatic fibrosis drugs.
Collapse
Affiliation(s)
- Xinyu Cheng
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Li Kang
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Jinfang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, P. R. China
| | - Qingye Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Zhenpeng Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Li Zhang
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Yuping Xie
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Lei Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Daobing Zeng
- General Surgery Department, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lantian Tian
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, P. R. China
| | - Lingqiang Zhang
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China.
| | - Ping Xu
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China; College of Life Sciences, Hebei University, 071002 Baoding, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, P. R. China.
| | - Yanchang Li
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; College of Life Sciences, Hebei University, 071002 Baoding, China.
| |
Collapse
|
3
|
Xu L, Lyu X, Wang Y, Ni L, Li P, Zeng P, Wang Q, Chang Y, Pan C, Hu Q, Huang S, Dang N. Neddylation modification stabilizes LC3B by antagonizing its ubiquitin-mediated degradation and promoting autophagy in skin. Proc Natl Acad Sci U S A 2025; 122:e2411429122. [PMID: 40208944 PMCID: PMC12012473 DOI: 10.1073/pnas.2411429122] [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: 06/07/2024] [Accepted: 02/26/2025] [Indexed: 04/12/2025] Open
Abstract
The Atg8-family proteins, including LC3B (microtubule-associated protein 1 light chain 3 beta), are pivotal for key steps in the autophagy process. Proper regulation of LC3B homeostasis is essential for its function. Although LC3B is modulated by various posttranslational modifications (PTMs), the impact of these modifications on LC3B protein homeostasis remains unclear. Neddylation, a recently identified ubiquitin-like modification, plays diverse biological roles. Here, we identify LC3B as a specific target for neddylation. This modification weakens LC3B's interaction with the ubiquitin E3 ligases VHL and BIRC6, thereby reducing LC3B ubiquitination. Depletion of ubiquitin-conjugating enzyme E2M (UBE2M), the primary E2 enzyme in the neddylation pathway, destabilizes LC3B and suppresses autophagy activity. Heterozygous Ube2m knockout (Ube2m+/-) mice exhibit pronounced aging-like phenotypes, with reduced LC3B expression and impaired autophagy in skin tissues. Our findings demonstrate that LC3B neddylation is vital for maintaining its stability and regulating autophagy flux, offering a potential therapeutic avenue to mitigate aging-related processes.
Collapse
Affiliation(s)
- Linlin Xu
- Department of Dermatology, Shandong Provincial Hospital, Shandong University, Jinan250021, Shandong, China
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan250021, Shandong, China
| | - Xinxing Lyu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan250022, Shandong, China
- School of Clinical and Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan250117, Shandong, China
| | - Yibo Wang
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan250021, Shandong, China
| | - Li Ni
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan250021, Shandong, China
| | - Pin Li
- Department of Dermatology, Shandong Provincial Hospital, Shandong University, Jinan250021, Shandong, China
| | - Piao Zeng
- School of Clinical and Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan250117, Shandong, China
| | - Qixia Wang
- School of Clinical and Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan250117, Shandong, China
| | - Yunhao Chang
- School of Clinical and Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan250117, Shandong, China
- Department of Geriatrics, Jiangsu Province Hospital, The First Clinical Medical College of Nanjing Medical University, Nanjing210029, Jiangsu, China
| | - Chenglong Pan
- School of Clinical and Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan250117, Shandong, China
| | - Qingxia Hu
- School of Clinical and Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan250117, Shandong, China
| | - Shuhong Huang
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan250021, Shandong, China
- School of Clinical and Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan250117, Shandong, China
| | - Ningning Dang
- Department of Dermatology, Shandong Provincial Hospital, Shandong University, Jinan250021, Shandong, China
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan250021, Shandong, China
| |
Collapse
|
4
|
Liao Y, Li X, Ma W, Lin X, Kuang J, Zheng X, Li Z, Qiao F, Liu C, Zhou J, Li F, Li R, Kang BH, Li H, Gao C. The plant retromer components SNXs bind to ATG8 and CLASP to mediate autophagosome movement along microtubules. MOLECULAR PLANT 2025; 18:416-436. [PMID: 39718933 DOI: 10.1016/j.molp.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/08/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
In eukaryotic cells, autophagosomes are double-membrane vesicles that are highly mobile and traffic along cytoskeletal tracks. While core autophagy-related proteins (ATGs) and other regulators involved in autophagosome biogenesis in plants have been extensively studied, the specific components regulating plant autophagosome motility remain elusive. In this study, using TurboID-based proximity labeling, we identify the retromer subcomplex comprising sorting nexin 1 (SNX1), SNX2a, and SNX2b as interacting partners of ATG8. Remarkably, SNX proteins decorate ATG8-labeled autophagosomes and facilitate their coordinated movement along microtubules. Depletion of SNX proteins restricts the motility of autophagosomes in the cytoplasm, resulting in decreased autophagic flux. Furthermore, we show that the microtubule-associated protein CLASP is a bridge, connecting the SNX-ATG8-decorated autophagosomes to the microtubules. Genetically, the clasp-1 mutant phenotype resembles that of plants with disrupted SNXs or microtubule networks, displaying diminished autophagosome motility and reduced autophagic flux. Collectively, our study unveils a hitherto unanticipated role of the SNXs subcomplex in connecting autophagosomes with microtubules to promote autophagosome mobility in Arabidopsis.
Collapse
Affiliation(s)
- Yanglan Liao
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xibao Li
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Wenlong Ma
- Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Xinyi Lin
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Jiayi Kuang
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xuanang Zheng
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Zien Li
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Fanfan Qiao
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Chuanliang Liu
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Jun Zhou
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Faqiang Li
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Ruixi Li
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Byung-Ho Kang
- Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Hongbo Li
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Caiji Gao
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
5
|
Kondo N, Mimori-Kiyosue Y, Tokuhiro K, Pezzotti G, Kinashi T. The autophagy component LC3 regulates lymphocyte adhesion via LFA1 transport in response to outside-in signaling. Nat Commun 2025; 16:1343. [PMID: 39905041 PMCID: PMC11794545 DOI: 10.1038/s41467-025-56631-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
The leukocyte integrin LFA1 is indispensable for immune responses, orchestrating lymphocyte trafficking and adhesion. While LFA1 activation induces LFA1 clustering at the cell contact surface via outside-in signaling, the regulatory mechanisms remain unclear. Here, we uncovered a previously hidden function of the autophagosome component LC3 beyond its role in autophagy by bridging two seemingly unrelated pathways: LFA1 transport and autophagosome transport. LFA1 clusters co-trafficked with LC3, facilitating LFA1 accumulation at the contact surface. LC3b knockout decreased lymphocyte adhesiveness. LFA1 activation did not induce autophagy, whereas it increased mTOR and AMPK activity. LFA1-dependent AMPK activation enhances LFA1 and LC3 clustering and adhesion. Inhibiting Mst1 kinase-mediated LC3 phosphorylation promoted LC3-mediated LFA1 recruitment to the contact surface through direct interaction with RAPL, uncovering an unprecedented integrin recruitment route. These findings uncover a function of LC3 and expand our understanding of lymphocyte regulation via LFA1.
Collapse
Affiliation(s)
- Naoyuki Kondo
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan.
| | - Yuko Mimori-Kiyosue
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Keizo Tokuhiro
- Department of Genome Editing, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Giuseppe Pezzotti
- Biomedical Engineering Center, Kansai Medical University, Osaka, Japan
| | - Tatsuo Kinashi
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| |
Collapse
|
6
|
Zhang Z, Li M, Hou Y, Huang T, Zhang B, Lin Q, Shao G. SETD7 promotes LC3B methylation and degradation in ovarian cancer. J Biol Chem 2025; 301:108134. [PMID: 39725038 PMCID: PMC11791264 DOI: 10.1016/j.jbc.2024.108134] [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: 09/03/2024] [Revised: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Microtubule-associated protein 1 light chain 3 (LC3) is a key autophagy-related protein involved in regulating autophagosome formation and autophagy activity. Post-translational modifications of LC3 are necessary to modulate its function. However, LC3 protein methylation and its physiological significance have not yet been elucidated. Here, we show that SET domain containing lysine methyltransferase 7 (SETD7) interacts with LC3B, a common isoform of LC3, and methylates LC3B at lysine 51 (K51). SETD7-mediated methylation of LC3B promotes ubiquitination and degradation of LC3B, resulting in reduced autophagosome formation. Furthermore, SETD7 exerts a tumor-promotive function in ovarian cancer (OC) cells in a K51 methylation-dependent manner. Collectively, our data define a novel modification of LC3B and highlight the oncogenic effect of SETD7 via mediating LC3B methylation and degradation.
Collapse
Affiliation(s)
- Ziwei Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mingyang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yanan Hou
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ting Huang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bowen Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiong Lin
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Genbao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
7
|
Chen Y, Yi H, Liao S, He J, Zhou Y, Lei Y. LC3B: A microtubule-associated protein influences disease progression and prognosis. Cytokine Growth Factor Rev 2025; 81:16-26. [PMID: 39701849 DOI: 10.1016/j.cytogfr.2024.11.006] [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: 10/10/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024]
Abstract
Microtubule-associated protein 1 light chain 3B (MAP1LC3B, also known as LC3B) is a mammalian homolog of the autophagy-related protein 8 (ATG8) family. It plays a crucial role in cellular autophagy and is involved in several vital biological processes, including apoptosis and differentiation. Additionally, LC3B regulates immune responses. Due to its close association with malignant tumors and neurodegenerative diseases, and its potential as a prognostic indicator and therapeutic target, LC3B has become a significant research focus. This article aims to provide a comprehensive and systematic understanding of LC3B's role and mechanisms in autophagy, its impact on apoptosis and the underlying mechanisms, its regulation of cellular differentiation and transdifferentiation, its modulation of immune and inflammatory responses, the influence of upstream regulatory factors on LC3B's function, and its relevance to disease diagnosis, treatment, and prognosis. The goal is to establish a solid foundation for understanding LC3B's role in cellular processes and its regulatory mechanisms.
Collapse
Affiliation(s)
- Yan Chen
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Hong Yi
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Shan Liao
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Junyu He
- Department of Clinical Laboratory, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan 410007, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China.
| | - Yan Lei
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, Hunan 410013, China.
| |
Collapse
|
8
|
Kück U, Pöggeler S. STRIPAK, a fundamental signaling hub of eukaryotic development. Microbiol Mol Biol Rev 2024; 88:e0020523. [PMID: 39526753 DOI: 10.1128/mmbr.00205-23] [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] [Indexed: 11/16/2024] Open
Abstract
SUMMARYThe striatin-interacting phosphatase and kinase (STRIPAK) complex is involved in the regulation of many developmental processes in eukaryotic microorganisms and all animals, including humans. STRIPAK is a component of protein phosphatase 2A (PP2A), a highly conserved serine-threonine phosphatase composed of catalytic subunits (PP2Ac), a scaffolding subunit (PP2AA) and various substrate-directing B regulatory subunits. In particular, the B''' regulatory subunit called striatin has evoked major interest over the last 20 years. Studies in fungal systems have contributed substantially to our current knowledge about STRIPAK composition, assembly, and cellular localization, as well as its regulatory role in autophagy and the morphology of fungal development. STRIPAK represents a signaling hub with many kinases and thus integrates upstream and downstream information from many conserved eukaryotic signaling pathways. A profound understanding of STRIPAK's regulatory role in fungi opens the gateway to understanding the multifarious functions carried out by STRIPAK in higher eukaryotes, including its contribution to malignant cell growth.
Collapse
Affiliation(s)
- Ulrich Kück
- Allgemeine & Molekulare Botanik, Ruhr-University, Bochum, Germany
| | - Stefanie Pöggeler
- Department of Genetics of Eukaryotic Microorganisms, Institute of Microbiology and Genetics, Georg-August-University, Göttingen, Germany
- Göttingen Center for Molecular Biosciences (GZMB), Georg-August-University, Göttingen, Germany
| |
Collapse
|
9
|
Zhang L, Gao M, Wu Y, Liu H, Zhuang X, Zhou Y, Song Q, Bi S, Zhang W, Cui Y. MST1 interactomes profiling across cell death in esophageal squamous cell carcinoma. MEDICAL REVIEW (2021) 2024; 4:531-543. [PMID: 39664081 PMCID: PMC11629308 DOI: 10.1515/mr-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/27/2024] [Indexed: 12/13/2024]
Abstract
Objectives Resistance to apoptosis in esophageal squamous cell carcinoma (ESCC) constitutes a significant impediment to treatment efficacy. Exploring alternative cell death pathways and their regulatory factors beyond apoptosis is crucial for overcoming drug resistance and enhancing therapeutic outcomes in ESCC. Methods Mammalian Ste 20-like kinase 1 (MST1) is implicated in regulating various cell deaths, including apoptosis, autophagy, and pyroptosis. Employing enhanced ascorbate peroxidase 2 (APEX2) proximity labeling coupled with immunoprecipitation-mass spectrometry (IP-MS), we elucidated the interactomes of MST1 across these three cell death paradigms. Results Proteomic profiling unveiled the functional roles and subcellular localization of MST1 and its interacting proteins during normal proliferation and various cell death processes. Notably, MST1 exhibited an expanded interactome during cell death compared to normal proliferation and chromosome remodeling functions consistently. In apoptosis, there was a notable increase of mitosis-associated proteins such as INCENP, ANLN, KIF23, SHCBP1 and SUPT16H, which interacted with MST1, alongside decreased expression of the pre-apoptotic protein STK3. During autophagy, the bindings of DNA repair-related proteins CBX8 and m6A reader YTHDC1 to MST1 were enhanced. In pyroptosis, LRRFIP2 and FLII which can inhibit pyroptosis increasingly binding to MST1. Conclusions Our findings delineate potential mechanisms through which MST1 and its interactomes regulate cell death, paving the way for further investigation to validate and consolidate these observations.
Collapse
Affiliation(s)
- Li Zhang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Mingwei Gao
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yueguang Wu
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Huijuan Liu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Xuehan Zhuang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yan Zhou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Qiqin Song
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Shanshan Bi
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Weimin Zhang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yongping Cui
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Rajesh R U, Sangeetha D. Therapeutic potentials and targeting strategies of quercetin on cancer cells: Challenges and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155902. [PMID: 39059266 DOI: 10.1016/j.phymed.2024.155902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Every cell in the human body is vital because it maintains equilibrium and carries out a variety of tasks, including growth and development. These activities are carried out by a set of instructions carried by many different genes and organized into DNA. It is well recognized that some lifestyle decisions, like using tobacco, alcohol, UV, or multiple sexual partners, might increase one's risk of developing cancer. The advantages of natural products for any health issue are well known, and researchers are making attempts to separate flavonoid-containing substances from plants. Various parts of plants contain a phenolic compound called flavonoid. Quercetin, which belongs to the class of compounds known as flavones with chromone skeletal structure, has anti-cancer activity. PURPOSE The study was aimed at investigating the therapeutic action of the flavonoid quercetin on various cancer cells. METHODS The phrases quercetin, anti-cancer, nanoparticles, and cell line were used to search the data using online resources such as PubMed, and Google Scholar. Several critical previous studies have been included. RESULTS Quercetin inhibits various dysregulated signaling pathways that cause cancer cells to undergo apoptosis to exercise its anticancer effects. Numerous signaling pathways are impacted by quercetin, such as the Hedgehog system, Akt, NF-κB pathway, downregulated mutant p53, JAK/STAT, G1 phase arrest, Wnt/β-Catenin, and MAPK. There are downsides to quercetin, like hydrophobicity, first-pass effect, instability in the gastrointestinal tract, etc., because of which it is not well-established in the pharmaceutical industry. The solution to these drawbacks in the future is using bio-nanomaterials like chitosan, PLGA, liposomes, and silk fibroin as carriers, which can enhance the target specificity of quercetin. The first section of this review covers the specifics of flavonoids and quercetin; the second section covers the anti-cancer activity of quercetin; and the third section explains the drawbacks and conjugation of quercetin with nanoparticles for drug delivery by overcoming quercetin's drawback. CONCLUSIONS Overall, this review presented details about quercetin, which is a plant derivative with a promising molecular mechanism of action. They inhibit cancer by various mechanisms with little or no side effects. It is anticipated that plant-based materials will become increasingly relevant in the treatment of cancer.
Collapse
Affiliation(s)
- Udaya Rajesh R
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India
| | - Dhanaraj Sangeetha
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India.
| |
Collapse
|
11
|
Lim SHY, Hansen M, Kumsta C. Molecular Mechanisms of Autophagy Decline during Aging. Cells 2024; 13:1364. [PMID: 39195254 PMCID: PMC11352966 DOI: 10.3390/cells13161364] [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: 07/06/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Macroautophagy (hereafter autophagy) is a cellular recycling process that degrades cytoplasmic components, such as protein aggregates and mitochondria, and is associated with longevity and health in multiple organisms. While mounting evidence supports that autophagy declines with age, the underlying molecular mechanisms remain unclear. Since autophagy is a complex, multistep process, orchestrated by more than 40 autophagy-related proteins with tissue-specific expression patterns and context-dependent regulation, it is challenging to determine how autophagy fails with age. In this review, we describe the individual steps of the autophagy process and summarize the age-dependent molecular changes reported to occur in specific steps of the pathway that could impact autophagy. Moreover, we describe how genetic manipulations of autophagy-related genes can affect lifespan and healthspan through studies in model organisms and age-related disease models. Understanding the age-related changes in each step of the autophagy process may prove useful in developing approaches to prevent autophagy decline and help combat a number of age-related diseases with dysregulated autophagy.
Collapse
Affiliation(s)
- Shaun H. Y. Lim
- Graduate School of Biological Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA;
| | - Malene Hansen
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA;
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Caroline Kumsta
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA;
| |
Collapse
|
12
|
Zhao Z, Chu Y, Feng A, Zhang S, Wu H, Li Z, Sun M, Zhang L, Chen T, Xu M. STK3 kinase activation inhibits tumor proliferation through FOXO1-TP53INP1/P21 pathway in esophageal squamous cell carcinoma. Cell Oncol (Dordr) 2024; 47:1295-1314. [PMID: 38436783 PMCID: PMC11322239 DOI: 10.1007/s13402-024-00928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
PURPOSE Esophageal squamous cell carcinoma (ESCC) is an aggressive disease with a poor prognosis, caused by the inactivation of critical cell growth regulators that lead to uncontrolled proliferation and increased malignancy. Although Serine/Threonine Kinase 3 (STK3), also known as Mammalian STE20-like protein kinase 2 (MST2), is a highly conserved kinase of the Hippo pathway, plays a critical role in immunomodulation, organ development, cellular differentiation, and cancer suppression, its phenotype and function in ESCC require further investigation. In this study, we report for the first time on the role of STK3 kinase and its activation condition in ESCC, as well as the mechanism and mediators of kinase activation. METHODS In this study, we investigated the expression and clinical significance of STK3 in ESCC. We first used bioinformatics databases and immunohistochemistry to analyze STK3 expression in the ESCC patient cohort and conducted survival analysis. In vivo, we conducted a tumorigenicity assay using nude mouse models to demonstrate the phenotypes of STK3 kinase. In vitro, we conducted Western blot analysis, qPCR analysis, CO-IP, and immunofluorescence (IF) staining analysis to detect molecule expression, interaction, and distribution. We measured proliferation, migration, and apoptosis abilities in ESCC cells in the experimental groups using CCK-8 and transwell assays, flow cytometry, and EdU staining. We used RNA-seq to identify genes that were differentially expressed in ESCC cells with silenced STK3 or FOXO1. We demonstrated the regulatory relationship of the TP53INP1/P21 gene medicated by the STK3-FOXO1 axis using Western blotting and ChIP in vitro. RESULTS We demonstrate high STK3 expression in ESCC tissue and cell lines compared to esophageal epithelium. Cellular ROS induces STK3 autophosphorylation in ESCC cells, resulting in upregulated p-STK3/4. STK3 activation inhibits ESCC cell proliferation and migration by triggering apoptosis and suppressing the cell cycle. STK3 kinase activation phosphorylates FOXO1Ser212, promoting nuclear translocation, enhancing transcriptional activity, and upregulating TP53INP1 and P21. We also investigated TP53INP1 and P21's phenotypic effects in ESCC, finding that their knockdown significantly increases tumor proliferation, highlighting their crucial role in ESCC tumorigenesis. CONCLUSION STK3 kinase has a high expression level in ESCC and can be activated by cellular ROS, inhibiting cell proliferation and migration. Additionally, STK3 activation-mediated FOXO1 regulates ESCC cell apoptosis and cell cycle arrest by targeting TP53INP1/P21. Our research underscores the anti-tumor function of STK3 in ESCC and elucidates the mechanism underlying its anti-tumor effect on ESCC.
Collapse
Affiliation(s)
- Ziying Zhao
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuan Chu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Anqi Feng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shihan Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hao Wu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhaoxing Li
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Mingchuang Sun
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Li Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Meidong Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
13
|
Samuel VP, Moglad E, Afzal M, Kazmi I, Alzarea SI, Ali H, Almujri SS, Abida, Imran M, Gupta G, Chinni SV, Tiwari A. Exploring Ubiquitin-specific proteases as therapeutic targets in Glioblastoma. Pathol Res Pract 2024; 260:155443. [PMID: 38981348 DOI: 10.1016/j.prp.2024.155443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Glioblastoma (GB) remains a formidable challenge and requires new treatment strategies. The vital part of the Ubiquitin-proteasome system (UPS) in cellular regulation has positioned it as a potentially crucial target in GB treatment, given its dysregulation oncolines. The Ubiquitin-specific proteases (USPs) in the UPS system were considered due to the garden role in the cellular processes associated with oncolines and their vital function in the apoptotic process, cell cycle regulation, and autophagy. The article provides a comprehensive summary of the evidence base for targeting USPs as potential factors for neoplasm treatment. The review considers the participation of the UPS system in the development, resulting in the importance of p53, Rb, and NF-κB, and evaluates specific goals for therapeutic administration using midnight proteasomal inhibitors and small molecule antagonists of E1 and E2 enzymes. Despite the slowed rate of drug creation, recent therapeutic discoveries based on USP system dynamics hold promise for specialized therapies. The review concludes with an analysis of future wanderers and the feasible effects of targeting USPs on personalized GB therapies, which can improve patient hydration in this current and unattractive therapeutic landscape. The manuscript emphasizes the possibility of USP oncogene therapy as a promising alternative treatment line for GB. It stresses the direct creation of research on the medical effectiveness of the approach.
Collapse
Affiliation(s)
- Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, the United Arab Emirates
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Al-Jouf, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Aseer 61421, Saudi Arabia
| | - Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, Jenjarom, Selangor 42610, Malaysia
| | - Abhishek Tiwari
- Department of Pharmacy, Pharmacy Academy, IFTM University, Lodhipur-Rajpur, Moradabad 244102, India.
| |
Collapse
|
14
|
St. Louis BM, Quagliato SM, Su YT, Dyson G, Lee PC. The Hippo kinases control inflammatory Hippo signaling and restrict bacterial infection in phagocytes. mBio 2024; 15:e0342923. [PMID: 38624208 PMCID: PMC11078001 DOI: 10.1128/mbio.03429-23] [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: 12/20/2023] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
The Hippo kinases MST1 and MST2 initiate a highly conserved signaling cascade called the Hippo pathway that limits organ size and tumor formation in animals. Intriguingly, pathogens hijack this host pathway during infection, but the role of MST1/2 in innate immune cells against pathogens is unclear. In this report, we generated Mst1/2 knockout macrophages to investigate the regulatory activities of the Hippo kinases in immunity. Transcriptomic analyses identified differentially expressed genes (DEGs) regulated by MST1/2 that are enriched in biological pathways, such as systemic lupus erythematosus, tuberculosis, and apoptosis. Surprisingly, pharmacological inhibition of the downstream components LATS1/2 in the canonical Hippo pathway did not affect the expression of a set of immune DEGs, suggesting that MST1/2 control these genes via alternative inflammatory Hippo signaling. Moreover, MST1/2 may affect immune communication by influencing the release of cytokines, including TNFα, CXCL10, and IL-1ra. Comparative analyses of the single- and double-knockout macrophages revealed that MST1 and MST2 differentially regulate TNFα release and expression of the immune transcription factor MAF, indicating that the two homologous Hippo kinases individually play a unique role in innate immunity. Notably, both MST1 and MST2 can promote apoptotic cell death in macrophages upon stimulation. Lastly, we demonstrate that the Hippo kinases are critical factors in mammalian macrophages and single-cell amoebae to restrict infection by Legionella pneumophila, Escherichia coli, and Pseudomonas aeruginosa. Together, these results uncover non-canonical inflammatory Hippo signaling in macrophages and the evolutionarily conserved role of the Hippo kinases in the anti-microbial defense of eukaryotic hosts. IMPORTANCE Identifying host factors involved in susceptibility to infection is fundamental for understanding host-pathogen interactions. Clinically, individuals with mutations in the MST1 gene which encodes one of the Hippo kinases experience recurrent infection. However, the impact of the Hippo kinases on innate immunity remains largely undetermined. This study uses mammalian macrophages and free-living amoebae with single- and double-knockout in the Hippo kinase genes and reveals that the Hippo kinases are the evolutionarily conserved determinants of host defense against microbes. In macrophages, the Hippo kinases MST1 and MST2 control immune activities at multiple levels, including gene expression, immune cell communication, and programmed cell death. Importantly, these activities controlled by MST1 and MST2 in macrophages are independent of the canonical Hippo cascade that is known to limit tissue growth and tumor formation. Together, these findings unveil a unique inflammatory Hippo signaling pathway that plays an essential role in innate immunity.
Collapse
Affiliation(s)
- Brendyn M. St. Louis
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| | - Sydney M. Quagliato
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| | - Yu-Ting Su
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| | - Gregory Dyson
- Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Pei-Chung Lee
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
15
|
Su MSW, Cheng YL, Lin YS, Wu JJ. Interplay between group A Streptococcus and host innate immune responses. Microbiol Mol Biol Rev 2024; 88:e0005222. [PMID: 38451081 PMCID: PMC10966951 DOI: 10.1128/mmbr.00052-22] [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] [Indexed: 03/08/2024] Open
Abstract
SUMMARYGroup A Streptococcus (GAS), also known as Streptococcus pyogenes, is a clinically well-adapted human pathogen that harbors rich virulence determinants contributing to a broad spectrum of diseases. GAS is capable of invading epithelial, endothelial, and professional phagocytic cells while evading host innate immune responses, including phagocytosis, selective autophagy, light chain 3-associated phagocytosis, and inflammation. However, without a more complete understanding of the different ways invasive GAS infections develop, it is difficult to appreciate how GAS survives and multiplies in host cells that have interactive immune networks. This review article attempts to provide an overview of the behaviors and mechanisms that allow pathogenic GAS to invade cells, along with the strategies that host cells practice to constrain GAS infection. We highlight the counteractions taken by GAS to apply virulence factors such as streptolysin O, nicotinamide-adenine dinucleotidase, and streptococcal pyrogenic exotoxin B as a hindrance to host innate immune responses.
Collapse
Affiliation(s)
- Marcia Shu-Wei Su
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Lin Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yee-Shin Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jiunn-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
16
|
Rak M, Menge A, Tesch R, Berger LM, Balourdas DI, Shevchenko E, Krämer A, Elson L, Berger BT, Abdi I, Wahl LM, Poso A, Kaiser A, Hanke T, Kronenberger T, Joerger AC, Müller S, Knapp S. Development of Selective Pyrido[2,3- d]pyrimidin-7(8 H)-one-Based Mammalian STE20-Like (MST3/4) Kinase Inhibitors. J Med Chem 2024; 67:3813-3842. [PMID: 38422480 DOI: 10.1021/acs.jmedchem.3c02217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Mammalian STE20-like (MST) kinases 1-4 play key roles in regulating the Hippo and autophagy pathways, and their dysregulation has been implicated in cancer development. In contrast to the well-studied MST1/2, the roles of MST3/4 are less clear, in part due to the lack of potent and selective inhibitors. Here, we re-evaluated literature compounds, and used structure-guided design to optimize the p21-activated kinase (PAK) inhibitor G-5555 (8) to selectively target MST3/4. These efforts resulted in the development of MR24 (24) and MR30 (27) with good kinome-wide selectivity and high cellular potency. The distinct cellular functions of closely related MST kinases can now be elucidated with subfamily-selective chemical tool compounds using a combination of the MST1/2 inhibitor PF-06447475 (2) and the two MST3/4 inhibitors developed. We found that MST3/4-selective inhibition caused a cell-cycle arrest in the G1 phase, whereas MST1/2 inhibition resulted in accumulation of cells in the G2/M phase.
Collapse
Affiliation(s)
- Marcel Rak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Amelie Menge
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Roberta Tesch
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Lena M Berger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Dimitrios-Ilias Balourdas
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Ekaterina Shevchenko
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
- German Translational Cancer Network (DKTK) and Frankfurt Cancer Institute (FCI), 60438 Frankfurt am Main, Germany
| | - Lewis Elson
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Benedict-Tilman Berger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Ismahan Abdi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Laurenz M Wahl
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Antti Poso
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Astrid Kaiser
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Thomas Hanke
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
- German Translational Cancer Network (DKTK) and Frankfurt Cancer Institute (FCI), 60438 Frankfurt am Main, Germany
| |
Collapse
|
17
|
Jeong DJ, Um JH, Kim YY, Shin DJ, Im S, Lee KM, Lee YH, Lim DS, Kim D, Yun J. The Mst1/2-BNIP3 axis is required for mitophagy induction and neuronal viability under mitochondrial stress. Exp Mol Med 2024; 56:674-685. [PMID: 38443598 PMCID: PMC10984967 DOI: 10.1038/s12276-024-01198-y] [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: 09/12/2023] [Revised: 12/21/2023] [Accepted: 01/01/2024] [Indexed: 03/07/2024] Open
Abstract
Mitophagy induction upon mitochondrial stress is critical for maintaining mitochondrial homeostasis and cellular function. Here, we found that Mst1/2 (Stk3/4), key regulators of the Hippo pathway, are required for the induction of mitophagy under various mitochondrial stress conditions. Knockdown of Mst1/2 or pharmacological inhibition by XMU-MP-1 treatment led to impaired mitophagy induction upon CCCP and DFP treatment. Mechanistically, Mst1/2 induces mitophagy independently of the PINK1-Parkin pathway and the canonical Hippo pathway. Moreover, our results suggest the essential involvement of BNIP3 in Mst1/2-mediated mitophagy induction upon mitochondrial stress. Notably, Mst1/2 knockdown diminishes mitophagy induction, exacerbates mitochondrial dysfunction, and reduces cellular survival upon neurotoxic stress in both SH-SY5Y cells and Drosophila models. Conversely, Mst1 and Mst2 expression enhances mitophagy induction and cell survival. In addition, AAV-mediated Mst1 expression reduced the loss of TH-positive neurons, ameliorated behavioral deficits, and improved mitochondrial function in an MPTP-induced Parkinson's disease mouse model. Our findings reveal the Mst1/2-BNIP3 regulatory axis as a novel mediator of mitophagy induction under conditions of mitochondrial stress and suggest that Mst1/2 play a pivotal role in maintaining mitochondrial function and neuronal viability in response to neurotoxic treatment.
Collapse
Affiliation(s)
- Dae Jin Jeong
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Jee-Hyun Um
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Young Yeon Kim
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Dong Jin Shin
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Sangwoo Im
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Kang-Min Lee
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Dae-Sik Lim
- Department of Biological Sciences, National Creative Research Center for Cell Plasticity, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Donghoon Kim
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
- Department of Pharmacology, College of Medicine, Dong-A University, Busan, Korea
| | - Jeanho Yun
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea.
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea.
| |
Collapse
|
18
|
Yang Y, Arnold ML, Lange CM, Sun LH, Broussalian M, Doroodian S, Ebata H, Choy EH, Poon K, Moreno TM, Singh A, Driscoll M, Kumsta C, Hansen M. Autophagy protein ATG-16.2 and its WD40 domain mediate the beneficial effects of inhibiting early-acting autophagy genes in C. elegans neurons. NATURE AGING 2024; 4:198-212. [PMID: 38177330 PMCID: PMC11022750 DOI: 10.1038/s43587-023-00548-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024]
Abstract
While autophagy genes are required for lifespan of long-lived animals, their tissue-specific roles in aging remain unclear. Here, we inhibited autophagy genes in Caenorhabditis elegans neurons, and found that knockdown of early-acting autophagy genes, except atg-16.2, increased lifespan, and decreased neuronal PolyQ aggregates, independently of autophagosomal degradation. Neurons can secrete protein aggregates via vesicles called exophers. Inhibiting neuronal early-acting autophagy genes, except atg-16.2, increased exopher formation and exopher events extended lifespan, suggesting exophers promote organismal fitness. Lifespan extension, reduction in PolyQ aggregates and increase in exophers were absent in atg-16.2 null mutants, and restored by full-length ATG-16.2 expression in neurons, but not by ATG-16.2 lacking its WD40 domain, which mediates noncanonical functions in mammalian systems. We discovered a neuronal role for C. elegans ATG-16.2 and its WD40 domain in lifespan, proteostasis and exopher biogenesis. Our findings suggest noncanonical functions for select autophagy genes in both exopher formation and in aging.
Collapse
Affiliation(s)
- Yongzhi Yang
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Scripps Research Institute, La Jolla, CA, USA
| | - Meghan Lee Arnold
- Rutgers, The State University of New Jersey, Nelson Biological Labs, Piscataway, NJ, USA
| | - Caitlin M Lange
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ling-Hsuan Sun
- Buck Institute for Aging Research, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Elizabeth H Choy
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Karie Poon
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Tatiana M Moreno
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Anupama Singh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Monica Driscoll
- Rutgers, The State University of New Jersey, Nelson Biological Labs, Piscataway, NJ, USA
| | - Caroline Kumsta
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
- Buck Institute for Aging Research, Novato, CA, USA.
| |
Collapse
|
19
|
Csabai L, Bohár B, Türei D, Prabhu S, Földvári-Nagy L, Madgwick M, Fazekas D, Módos D, Ölbei M, Halka T, Poletti M, Kornilova P, Kadlecsik T, Demeter A, Szalay-Bekő M, Kapuy O, Lenti K, Vellai T, Gul L, Korcsmáros T. AutophagyNet: high-resolution data source for the analysis of autophagy and its regulation. Autophagy 2024; 20:188-201. [PMID: 37589496 PMCID: PMC10761021 DOI: 10.1080/15548627.2023.2247737] [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: 03/29/2023] [Revised: 07/31/2023] [Accepted: 08/06/2023] [Indexed: 08/18/2023] Open
Abstract
Macroautophagy/autophagy is a highly-conserved catabolic procss eliminating dysfunctional cellular components and invading pathogens. Autophagy malfunction contributes to disorders such as cancer, neurodegenerative and inflammatory diseases. Understanding autophagy regulation in health and disease has been the focus of the last decades. We previously provided an integrated database for autophagy research, the Autophagy Regulatory Network (ARN). For the last eight years, this resource has been used by thousands of users. Here, we present a new and upgraded resource, AutophagyNet. It builds on the previous database but contains major improvements to address user feedback and novel needs due to the advancement in omics data availability. AutophagyNet contains updated interaction curation and integration of over 280,000 experimentally verified interactions between core autophagy proteins and their protein, transcriptional and post-transcriptional regulators as well as their potential upstream pathway connections. AutophagyNet provides annotations for each core protein about their role: 1) in different types of autophagy (mitophagy, xenophagy, etc.); 2) in distinct stages of autophagy (initiation, expansion, termination, etc.); 3) with subcellular and tissue-specific localization. These annotations can be used to filter the dataset, providing customizable download options tailored to the user's needs. The resource is available in various file formats (e.g. CSV, BioPAX and PSI-MI), and data can be analyzed and visualized directly in Cytoscape. The multi-layered regulation of autophagy can be analyzed by combining AutophagyNet with tissue- or cell type-specific (multi-)omics datasets (e.g. transcriptomic or proteomic data). The resource is publicly accessible at http://autophagynet.org.Abbreviations: ARN: Autophagy Regulatory Network; ATG: autophagy related; BCR: B cell receptor pathway; BECN1: beclin 1; GABARAP: GABA type A receptor-associated protein; IIP: innate immune pathway; LIR: LC3-interacting region; lncRNA: long non-coding RNA; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; miRNA: microRNA; NHR: nuclear hormone receptor; PTM: post-translational modification; RTK: receptor tyrosine kinase; TCR: T cell receptor; TLR: toll like receptor.
Collapse
Affiliation(s)
- Luca Csabai
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Balázs Bohár
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dénes Türei
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | | | - László Földvári-Nagy
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Matthew Madgwick
- Earlham Institute, Norwich, UK
- Quadram Institute, Norwich Research Park, Norwich, UK
| | - Dávid Fazekas
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dezső Módos
- Earlham Institute, Norwich, UK
- Quadram Institute, Norwich Research Park, Norwich, UK
| | - Márton Ölbei
- Earlham Institute, Norwich, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Themis Halka
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Martina Poletti
- Earlham Institute, Norwich, UK
- Quadram Institute, Norwich Research Park, Norwich, UK
| | | | - Tamás Kadlecsik
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | | | | | - Orsolya Kapuy
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Katalin Lenti
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Tibor Vellai
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
- ELKH/MTA-ELTE Genetics Research Group, Budapest, Hungary
| | - Lejla Gul
- Earlham Institute, Norwich, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tamás Korcsmáros
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
- Quadram Institute, Norwich Research Park, Norwich, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
20
|
Rogov VV, Nezis IP, Tsapras P, Zhang H, Dagdas Y, Noda NN, Nakatogawa H, Wirth M, Mouilleron S, McEwan DG, Behrends C, Deretic V, Elazar Z, Tooze SA, Dikic I, Lamark T, Johansen T. Atg8 family proteins, LIR/AIM motifs and other interaction modes. AUTOPHAGY REPORTS 2023; 2:27694127.2023.2188523. [PMID: 38214012 PMCID: PMC7615515 DOI: 10.1080/27694127.2023.2188523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
The Atg8 family of ubiquitin-like proteins play pivotal roles in autophagy and other processes involving vesicle fusion and transport where the lysosome/vacuole is the end station. Nuclear roles of Atg8 proteins are also emerging. Here, we review the structural and functional features of Atg8 family proteins and their protein-protein interaction modes in model organisms such as yeast, Arabidopsis, C. elegans and Drosophila to humans. Although varying in number of homologs, from one in yeast to seven in humans, and more than ten in some plants, there is a strong evolutionary conservation of structural features and interaction modes. The most prominent interaction mode is between the LC3 interacting region (LIR), also called Atg8 interacting motif (AIM), binding to the LIR docking site (LDS) in Atg8 homologs. There are variants of these motifs like "half-LIRs" and helical LIRs. We discuss details of the binding modes and how selectivity is achieved as well as the role of multivalent LIR-LDS interactions in selective autophagy. A number of LIR-LDS interactions are known to be regulated by phosphorylation. New methods to predict LIR motifs in proteins have emerged that will aid in discovery and analyses. There are also other interaction surfaces than the LDS becoming known where we presently lack detailed structural information, like the N-terminal arm region and the UIM-docking site (UDS). More interaction modes are likely to be discovered in future studies.
Collapse
Affiliation(s)
- Vladimir V. Rogov
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University, 60438 Frankfurt, am Main, and Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University, 60438 Frankfurt am Main, Germany
| | - Ioannis P. Nezis
- School of Life Sciences, University of Warwick, CV4 7AL Coventry, UK
| | | | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China and College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yasin Dagdas
- Gregor Mendel Institute, Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Nobuo N. Noda
- Institute for Genetic Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo 060-0815, Japan
| | - Hitoshi Nakatogawa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Martina Wirth
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK
| | - Stephane Mouilleron
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | | | - Christian Behrends
- Munich Cluster of Systems Neurology, Ludwig-Maximilians-Universität München, München, Germany
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM and Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Zvulun Elazar
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Sharon A. Tooze
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK
| | - Ivan Dikic
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main, and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Trond Lamark
- Autophagy Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Terje Johansen
- Autophagy Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
21
|
Seo G, Yu C, Han H, Xing L, Kattan RE, An J, Kizhedathu A, Yang B, Luo A, Buckle AL, Tifrea D, Edwards R, Huang L, Ju HQ, Wang W. The Hippo pathway noncanonically drives autophagy and cell survival in response to energy stress. Mol Cell 2023; 83:3155-3170.e8. [PMID: 37595580 PMCID: PMC10568779 DOI: 10.1016/j.molcel.2023.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 06/22/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023]
Abstract
The Hippo pathway is known for its crucial involvement in development, regeneration, organ size control, and cancer. While energy stress is known to activate the Hippo pathway and inhibit its effector YAP, the precise role of the Hippo pathway in energy stress response remains unclear. Here, we report a YAP-independent function of the Hippo pathway in facilitating autophagy and cell survival in response to energy stress, a process mediated by its upstream components MAP4K2 and STRIPAK. Mechanistically, energy stress disrupts the MAP4K2-STRIPAK association, leading to the activation of MAP4K2. Subsequently, MAP4K2 phosphorylates ATG8-family member LC3, thereby facilitating autophagic flux. MAP4K2 is highly expressed in head and neck cancer, and its mediated autophagy is required for head and neck tumor growth in mice. Altogether, our study unveils a noncanonical role of the Hippo pathway in energy stress response, shedding light on this key growth-related pathway in tissue homeostasis and cancer.
Collapse
Affiliation(s)
- Gayoung Seo
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Clinton Yu
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Han Han
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Li Xing
- Irvine Materials Research Institute, University of California, Irvine, Irvine, CA 92697, USA
| | - Rebecca Elizabeth Kattan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Jeongmin An
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Amrutha Kizhedathu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Bing Yang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Annabella Luo
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Abigail L Buckle
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Delia Tifrea
- Department of Pathology, University of California, Irvine, Irvine, CA 92697, USA
| | - Robert Edwards
- Department of Pathology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lan Huang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
22
|
Nieto-Torres JL, Zaretski S, Liu T, Adams PD, Hansen M. Post-translational modifications of ATG8 proteins - an emerging mechanism of autophagy control. J Cell Sci 2023; 136:jcs259725. [PMID: 37589340 PMCID: PMC10445744 DOI: 10.1242/jcs.259725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Abstract
Autophagy is a recycling mechanism involved in cellular homeostasis with key implications for health and disease. The conjugation of the ATG8 family proteins, which includes LC3B (also known as MAP1LC3B), to autophagosome membranes, constitutes a hallmark of the canonical autophagy process. After ATG8 proteins are conjugated to the autophagosome membranes via lipidation, they orchestrate a plethora of protein-protein interactions that support key steps of the autophagy process. These include binding to cargo receptors to allow cargo recruitment, association with proteins implicated in autophagosome transport and autophagosome-lysosome fusion. How these diverse and critical protein-protein interactions are regulated is still not well understood. Recent reports have highlighted crucial roles for post-translational modifications of ATG8 proteins in the regulation of ATG8 functions and the autophagy process. This Review summarizes the main post-translational regulatory events discovered to date to influence the autophagy process, mostly described in mammalian cells, including ubiquitylation, acetylation, lipidation and phosphorylation, as well as their known contributions to the autophagy process, physiology and disease.
Collapse
Affiliation(s)
- Jose L. Nieto-Torres
- Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging, and Regeneration, La Jolla, CA 92037, USA
- Department of Biomedical Sciences, School of Health Sciences and Veterinary, Universidad Cardenal Herrera-CEU, CEU Universities, 46113 Moncada, Spain
| | - Sviatlana Zaretski
- Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging, and Regeneration, La Jolla, CA 92037, USA
| | - Tianhui Liu
- Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging, and Regeneration, La Jolla, CA 92037, USA
| | - Peter D. Adams
- Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging, and Regeneration, La Jolla, CA 92037, USA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging, and Regeneration, La Jolla, CA 92037, USA
- The Buck Institute for Aging Research, Novato, CA 94945, USA
| |
Collapse
|
23
|
Funes S, Gadd DH, Mosqueda M, Zhong J, Jung J, Shankaracharya, Unger M, Cameron D, Dawes P, Keagle PJ, McDonough JA, Boopathy S, Sena-Esteves M, Lutz C, Skarnes WC, Lim ET, Schafer DP, Massi F, Landers JE, Bosco DA. Expression of ALS-PFN1 impairs vesicular degradation in iPSC-derived microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.541136. [PMID: 37398081 PMCID: PMC10312575 DOI: 10.1101/2023.06.01.541136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Microglia play a pivotal role in neurodegenerative disease pathogenesis, but the mechanisms underlying microglia dysfunction and toxicity remain to be fully elucidated. To investigate the effect of neurodegenerative disease-linked genes on the intrinsic properties of microglia, we studied microglia-like cells derived from human induced pluripotent stem cells (iPSCs), termed iMGs, harboring mutations in profilin-1 (PFN1) that are causative for amyotrophic lateral sclerosis (ALS). ALS-PFN1 iMGs exhibited lipid dysmetabolism and deficits in phagocytosis, a critical microglia function. Our cumulative data implicate an effect of ALS-linked PFN1 on the autophagy pathway, including enhanced binding of mutant PFN1 to the autophagy signaling molecule PI3P, as an underlying cause of defective phagocytosis in ALS-PFN1 iMGs. Indeed, phagocytic processing was restored in ALS-PFN1 iMGs with Rapamycin, an inducer of autophagic flux. These outcomes demonstrate the utility of iMGs for neurodegenerative disease research and highlight microglia vesicular degradation pathways as potential therapeutic targets for these disorders.
Collapse
|
24
|
Xu X, Lin J, Li X, Shao Q, Cui X, Zhu G, Lou S, Zhong W, Liu L, Pan Y. Genetic Variants in Mammalian STE20-like Protein Kinase 2 were associated with risk of NSCL/P. Gene 2023; 873:147459. [PMID: 37141954 DOI: 10.1016/j.gene.2023.147459] [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: 01/05/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
AIM Mammalian STE20-like protein kinase 2 (MST2) plays an important role in apoptosis and the development of many disorders. Here, we aim to explore if genetic variants in MST2 are associated with the risk of non-syndromic cleft lip with or without palate (NSCL/P). MATERIALS AND METHODS The association study was performed in a two-stage study of 1,069 cases and 1,724 controls to evaluate the association between genetic variants in the MST2 and NSCL/P risk. The potential function of the candidate single nucleotide polymorphism (SNP) was predicted using HaploReg, RegulomeDB, and public craniofacial histone chromatin immunoprecipitation sequencing (ChIP-seq) data. Haploview was used to perform the haplotype of risk alleles. The expression quantitative trait loci (eQTL) effect was assessed using the Genotype-Tissue Expression (GTEx) project. Gene expression in mouse embryo tissue was performed using data downloaded from GSE67985. The potential role of candidate gene in the development of NSCL/P was assessed by correlation and enrichment analysis. RESULTS Among SNPs in MST2, rs2922070 C allele (Pmeta = 2.93E-04) and rs6988087 T allele (Pmeta = 1.57E-03) were linked with significantly increased risk of NSCL/P. Rs2922070, rs6988087 and their high linkage disequilibrium (LD) SNPs constituted a risk haplotype of NSCL/P. Individuals carrying 3-4 risk alleles had an elevated risk of NSCL/P compared to those who carried less risk alleles (P = 2.00E-04). The eQTL analysis revealed a significant association between these two variants and MST2 in muscle tissue of the body. The MST2 expressed during mouse craniofacial development and over-expressed in the human orbicularis oris muscle (OOM) of NSCL/P patients compared to controls. MST2 was involved in the development of NSCL/P by regulating the mRNA surveillance pathway, the MAPK signaling pathway, the neurotrophin signaling pathway, the FoxO signaling pathway and the VEGF signaling pathway. CONCLUSION MST2 was associated with the development of NSCL/P.
Collapse
Affiliation(s)
- Xinze Xu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Junyan Lin
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Xiaofeng Li
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Qinghua Shao
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Xing Cui
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Guirong Zhu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Shu Lou
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Weijie Zhong
- Department of Stomatology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China Suzhou, 215127, China; Department of Stomatology, Medical Center of Soochow University, Suzhou, 215127, China.
| | - Luwei Liu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China.
| | - Yongchu Pan
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
25
|
Kiriyama Y, Nochi H. Role of Microbiota-Modified Bile Acids in the Regulation of Intracellular Organelles and Neurodegenerative Diseases. Genes (Basel) 2023; 14:825. [PMID: 37107583 PMCID: PMC10137455 DOI: 10.3390/genes14040825] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Bile acids (BAs) are amphiphilic steroidal molecules generated from cholesterol in the liver and facilitate the digestion and absorption of fat-soluble substances in the gut. Some BAs in the intestine are modified by the gut microbiota. Because BAs are modified in a variety of ways by different types of bacteria present in the gut microbiota, changes in the gut microbiota can affect the metabolism of BAs in the host. Although most BAs absorbed from the gut are transferred to the liver, some are transferred to the systemic circulation. Furthermore, BAs have also been detected in the brain and are thought to migrate into the brain through the systemic circulation. Although BAs are known to affect a variety of physiological functions by acting as ligands for various nuclear and cell-surface receptors, BAs have also been found to act on mitochondria and autophagy in the cell. This review focuses on the BAs modified by the gut microbiota and their roles in intracellular organelles and neurodegenerative diseases.
Collapse
Affiliation(s)
- Yoshimitsu Kiriyama
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
- Institute of Neuroscience, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Hiromi Nochi
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| |
Collapse
|
26
|
Feng H, Liu X, Zhou C, Gu Q, Li Y, Chen J, Teng J, Zheng P. CCDC115 inhibits autophagy-mediated degradation of YAP to promote cell proliferation. FEBS Lett 2023; 597:618-630. [PMID: 36650560 DOI: 10.1002/1873-3468.14575] [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: 10/18/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 01/19/2023]
Abstract
Autophagy and Hippo signalling pathways both play important roles in cell homeostasis and are often involved in tumourigenesis. However, the crosstalk between these two signal pathways in response to stress conditions, such as nutrient deficiency, is incompletely understood. Here, we show that vesicular localised coiled-coil domain containing 115 (CCDC115) inhibits autophagy as well as Hippo signalling pathway under starvation. Moreover, we show that CCDC115 interacts with the HOPS complex. This interaction competes with STX17, thus inhibiting the fusion of autophagosomes with lysosomes. Hence, CCDC115 inhibits the autophagic degradation of yes-associated protein (YAP), thereby promoting cell proliferation in nutrient-restricted situation.
Collapse
Affiliation(s)
- Hui Feng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Department of Biotechnology, Beijing Polytechnic, China
| | - Xiao Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Chenqian Zhou
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Qiuchen Gu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- School of Life Sciences, Beijing Normal University, China
| | - Ye Li
- Department of Biotechnology, Beijing Polytechnic, China
| | - Jianguo Chen
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Junlin Teng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Pengli Zheng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
27
|
Maejima Y, Zablocki D, Nah J, Sadoshima J. The role of the Hippo pathway in autophagy in the heart. Cardiovasc Res 2023; 118:3320-3330. [PMID: 35150237 DOI: 10.1093/cvr/cvac014] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/07/2022] [Indexed: 01/25/2023] Open
Abstract
The Hippo pathway, an evolutionarily conserved signalling mechanism, controls organ size and tumourigenesis. Increasing lines of evidence suggest that autophagy, an important mechanism of lysosome-mediated cellular degradation, is regulated by the Hippo pathway, which thereby profoundly affects cell growth and death responses in various cell types. In the heart, Mst1, an upstream component of the Hippo pathway, not only induces apoptosis but also inhibits autophagy through phosphorylation of Beclin 1. YAP/TAZ, transcription factor co-factors and the terminal effectors of the Hippo pathway, affect autophagy through transcriptional activation of TFEB, a master regulator of autophagy and lysosomal biogenesis. The cellular abundance of YAP is negatively regulated by autophagy and suppression of autophagy induces accumulation of YAP, which, in turn, acts as a feedback mechanism to induce autophagosome formation. Thus, the Hippo pathway and autophagy regulate each other, thereby profoundly affecting cardiomyocyte survival and death. This review discusses the interaction between the Hippo pathway and autophagy and its functional significance during stress conditions in the heart and the cardiomyocytes therein.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA.,Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA
| | - Jihoon Nah
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA
| |
Collapse
|
28
|
Xiang YC, Peng P, Liu XW, Jin X, Shen J, Zhang T, Zhang L, Wan F, Ren YL, Yu QQ, Zhao HZ, Si Y, Liu Y. Paris saponin VII, a Hippo pathway activator, induces autophagy and exhibits therapeutic potential against human breast cancer cells. Acta Pharmacol Sin 2022; 43:1568-1580. [PMID: 34522004 PMCID: PMC9159991 DOI: 10.1038/s41401-021-00755-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023]
Abstract
Dysregulation of the Hippo signaling pathway seen in many types of cancer is usually associated with a poor prognosis. Paris saponin VII (PSVII) is a steroid saponin isolated from traditional Chinese herbs with therapeutic action against various human cancers. In this study we investigated the effects of PSVII on human breast cancer (BC) cells and its anticancer mechanisms. We showed that PSVII concentration-dependently inhibited the proliferation of MDA-MB-231, MDA-MB-436 and MCF-7 BC cell lines with IC50 values of 3.16, 3.45, and 2.86 μM, respectively, and suppressed their colony formation. PSVII (1.2-1.8 μM) induced caspase-dependent apoptosis in the BC cell lines. PSVII treatment also induced autophagy and promoted autophagic flux in the BC cell lines. PSVII treatment decreased the expression and nuclear translocation of Yes-associated protein (YAP), a downstream transcriptional effector in the Hippo signaling pathway; overexpression of YAP markedly attenuated PSVII-induced autophagy. PSVII-induced, YAP-mediated autophagy was associated with increased active form of LATS1, an upstream effector of YAP. The activation of LATS1 was involved the participation of multiple proteins (including MST2, MOB1, and LATS1 itself) in an MST2-dependent sequential activation cascade. We further revealed that PSVII promoted the binding of LATS1 with MST2 and MOB1, and activated LATS1 in the BC cell lines. Molecular docking showed that PSVII directly bound to the MST2-MOB1-LATS1 ternary complex. Microscale thermophoresis analysis and drug affinity responsive targeting stability assay confirmed the high affinity between PSVII and the MST2-MOB1-LATS1 ternary complex. In mice bearing MDA-MB-231 cell xenograft, administration of PSVII (1.5 mg/kg, ip, 4 times/week, for 4 weeks) significantly suppressed the tumor growth with increased pLATS1, LC3-II and Beclin 1 levels and decreased YAP, p62 and Ki67 levels in the tumor tissue. Overall, this study demonstrates that PSVII is a novel and direct Hippo activator that has great potential in the treatment of BC.
Collapse
Affiliation(s)
- Yu-Chen Xiang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Peng Peng
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Xue-Wen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China
| | - Xin Jin
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Jie Shen
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Te Zhang
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China
| | - Liang Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Fang Wan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Yu-Liang Ren
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Qing-Qing Yu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China
| | - Hu-Zi Zhao
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China.
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China.
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China.
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
29
|
Zheng L, Wei F, Li G. The crosstalk between bacteria and host autophagy: host defense or bacteria offense. J Microbiol 2022; 60:451-460. [DOI: 10.1007/s12275-022-2009-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/18/2022] [Accepted: 03/29/2022] [Indexed: 12/26/2022]
|
30
|
Roy PK, Biswas A, Deepak K, Mandal M. An insight into the ubiquitin-proteasomal axis and related therapeutic approaches towards central nervous system malignancies. Biochim Biophys Acta Rev Cancer 2022; 1877:188734. [PMID: 35489645 DOI: 10.1016/j.bbcan.2022.188734] [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: 02/10/2022] [Revised: 04/12/2022] [Accepted: 04/22/2022] [Indexed: 10/18/2022]
Abstract
The Ubiquitin-Protease system (UPS) is a major destruction system that is responsible for the elimination of dysfunctional/misfolded proteins, thus acting as a pivotal regulator of protein homeostasis in eukaryotic cells. In this review, the UPS system and its various functions in the cell and their detailed impact such as cell cycle control, DNA damage response, apoptosis, and cellular stress regulations have been elucidated with a focus on the central nervous system. Since the Ubiquitin-Protease pathway(UPP) plays a prominent role in the sculpting of the CNS cells and their maintenance, it is naturally deeply involved in many malignancies that develop due to dysregulation of the UPS. Understanding the major disruptive players of the UPS in the development of these malignancies, for example, insoluble protein aggregates or inclusion bodies deposits due to malfunctioning of the UPS has paved the pathway for the development of new therapeutics. Here, the de-regulation of the UPS at various checkpoints in CNS malignancies has been detailed, thus facilitating an easy comprehension of the different targets that remain to be explored yet. The present therapeutic advancements in the field of CNS malignancies management through UPS targeting have also been included thus broadening the scope of drug development. Thus, this review while shedding sufficient light on the details of the UPS system and its connection to CNS malignancies, also opens new avenues for therapeutic advancements in the form of novel targetable UPP proteins and their interactions.
Collapse
Affiliation(s)
- Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Angana Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India..
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India..
| |
Collapse
|
31
|
Cancer cell's internal and external warriors: Autophagosomes and exosomes. Life Sci 2022; 300:120552. [PMID: 35452638 DOI: 10.1016/j.lfs.2022.120552] [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: 02/24/2022] [Revised: 04/09/2022] [Accepted: 04/10/2022] [Indexed: 12/24/2022]
Abstract
"That survival instinct, that will to live, that need to get back to life again, is more powerful than any consideration of taste, decency, politeness, manners, civility, anything. It's such a powerful force." This quote by famous director Danny Boyle is a perfect analogy to describe the cancer cell's inexhaustible drive to persist against all odds. In order to adapt to a hostile environment, the cancer cells rely on multiple mechanisms including immune escape, epithelial to mesenchymal transition, angiogenesis, extravasation, autophagy, exosome release among others. Cancer cells depute their internal and external warriors, autophagosomes and exosomes, to dwell in the belligerent tumor microenvironment. It is quite reasonable for a cancer cell, striving to survive, to invest in pathways that will provide the maximum advantage. Autophagy is an important cellular degradation pathway, while the exosome pathway provides an alternative cargo disposal mechanism to maintain the homeostasis and cell survival. While autophagic degradation provides the essential nutrients to rapidly dividing cells, exosomal secretion ensures that the tumor microenvironment is attuned to accommodate the swiftly expanding tumor mass. Studies have revealed that exosomes secreted by cancer cells can modulate autophagy in recipient cells, while autophagy can influence the biogenesis of exosomes. Autophagy and exosome crosstalk is extremely complex and it is only beginning to be recognized and documented. This review is focused on discussing the roles of autophagy and exosomes in the cancer cell's adaptation to the tumor microenvironment and how the two pathways are coordinately regulated to facilitate cancer cell survival.
Collapse
|
32
|
Cable J, Weber-Ban E, Clausen T, Walters KJ, Sharon M, Finley DJ, Gu Y, Hanna J, Feng Y, Martens S, Simonsen A, Hansen M, Zhang H, Goodwin JM, Reggio A, Chang C, Ge L, Schulman BA, Deshaies RJ, Dikic I, Harper JW, Wertz IE, Thomä NH, Słabicki M, Frydman J, Jakob U, David DC, Bennett EJ, Bertozzi CR, Sardana R, Eapen VV, Carra S. Targeted protein degradation: from small molecules to complex organelles-a Keystone Symposia report. Ann N Y Acad Sci 2022; 1510:79-99. [PMID: 35000205 DOI: 10.1111/nyas.14745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Targeted protein degradation is critical for proper cellular function and development. Protein degradation pathways, such as the ubiquitin proteasomes system, autophagy, and endosome-lysosome pathway, must be tightly regulated to ensure proper elimination of misfolded and aggregated proteins and regulate changing protein levels during cellular differentiation, while ensuring that normal proteins remain unscathed. Protein degradation pathways have also garnered interest as a means to selectively eliminate target proteins that may be difficult to inhibit via other mechanisms. On June 7 and 8, 2021, several experts in protein degradation pathways met virtually for the Keystone eSymposium "Targeting protein degradation: from small molecules to complex organelles." The event brought together researchers working in different protein degradation pathways in an effort to begin to develop a holistic, integrated vision of protein degradation that incorporates all the major pathways to understand how changes in them can lead to disease pathology and, alternatively, how they can be leveraged for novel therapeutics.
Collapse
Affiliation(s)
| | - Eilika Weber-Ban
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Tim Clausen
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter and Medical University of Vienna, Vienna, Austria
| | - Kylie J Walters
- Protein Processing Section, Center for Structural Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Michal Sharon
- Department of Bimolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel J Finley
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Yangnan Gu
- Department of Plant and Microbial Biology and Innovative Genomics Institute, University of California, Berkeley, California
| | - John Hanna
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Yue Feng
- Princess Margaret Cancer Centre, University Health Network and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Sascha Martens
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging, and Regeneration, La Jolla, California
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences and College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | | | - Alessio Reggio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Chunmei Chang
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, California
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Ivan Dikic
- Institute of Biochemistry II, School of Medicine and Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - J Wade Harper
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
| | - Ingrid E Wertz
- Departments of Molecular Oncology and Early Discovery Biochemistry, Genentech, Inc., South San Francisco, California
- Bristol Myers Squibb, Brisbane, California
| | - Nicolas H Thomä
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Mikołaj Słabicki
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Judith Frydman
- Biophysics Graduate Program, Department of Biology and Department of Genetics, Stanford University, Stanford, California
- Biohub, San Francisco, California
- Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Menlo Park, California
| | - Ursula Jakob
- Department of Molecular, Cellular and Developmental Biology, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan
| | - Della C David
- German Center for Neurodegenerative Diseases (DZNE), and Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Eric J Bennett
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, California
| | - Carolyn R Bertozzi
- Department of Chemistry and Stanford ChEM-H, Stanford University and Howard Hughes Medical Institute, Stanford, California
| | - Richa Sardana
- Weill Institute of Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York
| | - Vinay V Eapen
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
33
|
Ramaccini D, Pedriali G, Perrone M, Bouhamida E, Modesti L, Wieckowski MR, Giorgi C, Pinton P, Morciano G. Some Insights into the Regulation of Cardiac Physiology and Pathology by the Hippo Pathway. Biomedicines 2022; 10:biomedicines10030726. [PMID: 35327528 PMCID: PMC8945338 DOI: 10.3390/biomedicines10030726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 11/16/2022] Open
Abstract
The heart is one of the most fascinating organs in living beings. It beats up to 100,000 times a day throughout the lifespan, without resting. The heart undergoes profound anatomical, biochemical, and functional changes during life, from hypoxemic fetal stages to a completely differentiated four-chambered cardiac muscle. In the middle, many biological events occur after and intersect with each other to regulate development, organ size, and, in some cases, regeneration. Several studies have defined the essential roles of the Hippo pathway in heart physiology through the regulation of apoptosis, autophagy, cell proliferation, and differentiation. This molecular route is composed of multiple components, some of which were recently discovered, and is highly interconnected with multiple known prosurvival pathways. The Hippo cascade is evolutionarily conserved among species, and in addition to its regulatory roles, it is involved in disease by drastically changing the heart phenotype and its function when its components are mutated, absent, or constitutively activated. In this review, we report some insights into the regulation of cardiac physiology and pathology by the Hippo pathway.
Collapse
Affiliation(s)
- Daniela Ramaccini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Gaia Pedriali
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Mariasole Perrone
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| | - Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| |
Collapse
|
34
|
El-Saiy KA, Sayed RH, El-Sahar AE, Kandil EA. Modulation of histone deacetylase, the ubiquitin proteasome system, and autophagy underlies the neuroprotective effects of venlafaxine in a rotenone-induced Parkinson's disease model in rats. Chem Biol Interact 2022; 354:109841. [PMID: 35104487 DOI: 10.1016/j.cbi.2022.109841] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by motor and non-motor symptoms. Impairment of the ubiquitin proteasome system (UPS) and autophagy has been suggested to contribute to α-synuclein accumulation, which is identified as the pathological hallmark of PD. Recently, alteration in histone-3 acetylation has also been found to be correlated to PD. Interestingly, the histone deacetylase 6 (HDAC6) enzyme, which regulates the acetylation of histone-3, was shown to be involved in autophagy. Venlafaxine is an antidepressant that was proposed to inhibit HDAC expression in depressive rats' hippocampi. In this study, we aimed to examine the ability of venlafaxine to inhibit striatal HDAC6 and to enhance α-synuclein clearance through the activation of the UPS and autophagy, in addition to treating depression, which is the most debilitating non-motor symptom, in a rotenone model of PD. Venlafaxine administration was noted to decrease α-synuclein accumulation and preserve dopaminergic neurons along with restoration of striatal dopamine levels and motor recovery. Its administration augmented the UPS and autophagic markers (beclin-1, p62, and LC3) with consequent modulation of apoptotic indicators (Bax/Bcl-2 ratio, cytochrome c, and caspase-3). Additionally, venlafaxine inhibited HDAC6 with further enhancement of autophagy and restoration of histone-3 acetylation with subsequent increases in survival gene expressions (Bcl-2 and brain-derived neurotrophic factor). Chloroquine (autophagy inhibitor) was used to indicate the proposed pathway. Moreover, venlafaxine hampered depressive symptoms and improved hippocampal noradrenaline and serotonin levels. Collectively, venlafaxine is suggested to display neuroprotective effects with improvement of motor and non-motor PD symptoms.
Collapse
Affiliation(s)
- Khalid A El-Saiy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Ayman E El-Sahar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
35
|
Pei T, Luo B, Huang W, Liu D, Li Y, Xiao L, Huang X, Ouyang Y, Zhu H. Increased Expression of YAP Inhibited the Autophagy Level by Upregulating mTOR Signal in the Eutopic ESCs of Endometriosis. Front Endocrinol (Lausanne) 2022; 13:813165. [PMID: 35173685 PMCID: PMC8842667 DOI: 10.3389/fendo.2022.813165] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/03/2022] [Indexed: 01/18/2023] Open
Abstract
We first reported that the Hippo-YAP signaling pathway plays a critical role in the pathogenesis of endometriosis (EMS). Autophagy is also related to the invasion ability of endometrial cells and is involved in the pathogenesis of EMS through multi-levels. However, the precise regulatory mechanism of YAP on autophagy in the eutopic endometrial stromal cells (ESCs) is still unclear. Primary eutopic ESCs of EMS patients (n = 12) and control patients without EMS (n = 9) were isolated and cultured to investigate the expressions of YAP and mTOR, the role of YAP in autophagy, and the effect of the YAP-autophagy signal on the decidualization of the eutopic ESCs. Endometriosis-related sequencing data (GSE51981) in the GEO database were used to find the genes significantly correlated with YAP. We found 155 genes with significant differences in the interaction with YAP in EMS from the dataset, and the autophagy pathway was significantly enriched. Following on from our previous studies of YAP knockdown, overexpression of YAP resulted in an increased expression of mTOR and decreased ratio of LC3-II/LC3-I and autophagy markers, in the eutopic ESCs; transmission electron microscope observation also showed fewer autophagosomes compared with the control cells. Furthermore, ESCs of the Rapamycin-treated group showed significant decidual-like changes with significantly increased decidual prolactin level at 72 h after in vitro decidualization. These results demonstrate that the increased YAP inhibited the level of autophagy by upregulating the mTOR signal in the eutopic ESCs of endometriosis. The YAP-autophagy signal plays an important role in the pathogenesis of endometriosis-associated infertility.
Collapse
Affiliation(s)
- Tianjiao Pei
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Bin Luo
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Wei Huang
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Dong Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- Department of Reproductive Endocrinology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Yujing Li
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Li Xiao
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Xin Huang
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Yunwei Ouyang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- Department of Reproductive Endocrinology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- *Correspondence: Huili Zhu,
| |
Collapse
|
36
|
Nieto-Torres JL, Hansen M. Macroautophagy and aging: The impact of cellular recycling on health and longevity. Mol Aspects Med 2021; 82:101020. [PMID: 34507801 PMCID: PMC8671213 DOI: 10.1016/j.mam.2021.101020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/11/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023]
Abstract
Aging is associated with many deleterious changes at the cellular level, including the accumulation of potentially toxic components that can have devastating effects on health. A key protective mechanism to this end is the cellular recycling process called autophagy. During autophagy, damaged or surplus cellular components are delivered to acidic vesicles called lysosomes, that secure degradation and recycling of the components. Numerous links between autophagy and aging exist. Autophagy declines with age, and increasing evidence suggests that this reduction plays important roles in both physiological aging and the development of age-associated disorders. Studies in pharmacologically and genetically manipulated model organisms indicate that defects in autophagy promote age-related diseases, and conversely, that enhancement of autophagy has beneficial effects on both healthspan and lifespan. Here, we review our current understanding of the role of autophagy in different physiological processes and their molecular links with aging and age-related diseases. We also highlight some recent advances in the field that could accelerate the development of autophagy-based therapeutic interventions.
Collapse
Affiliation(s)
- Jose L Nieto-Torres
- Sanford Burnham Prebys Medical Discovery Institute. Program of Development, Aging, and Regeneration, La Jolla, CA, USA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute. Program of Development, Aging, and Regeneration, La Jolla, CA, USA.
| |
Collapse
|
37
|
Non-canonical roles of autophagy proteins in endocytosis and exocytosis. Biochem Soc Trans 2021; 49:2841-2851. [PMID: 34783341 DOI: 10.1042/bst20210811] [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] [Received: 09/13/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 01/18/2023]
Abstract
Autophagy, the pathways that degrade cytoplasmic constituents in lysosomes, contribute to most biological processes from aging and neurodegeneration to pathogen restriction and immunity. In recent years, it was realized that the autophagy machinery serves additional functions, primarily in endo- and exocytosis. In this review, I summarize recent advances in our understanding on how these non-canonical functions differ from canonical macroautophagy, and contribute to immune activation and viral replication. Understanding these pathways will allow us to harness them for the treatment of human diseases, as well as appreciate how cells use modules of membrane remodeling and trafficking for multiple biological functions.
Collapse
|
38
|
Chen S, Wu H, Wang Z, Jia M, Guo J, Jin J, Li X, Meng D, Lin L, He AR, Zhou P, Zhi X. Loss of SPTBN1 Suppresses Autophagy Via SETD7-mediated YAP Methylation in Hepatocellular Carcinoma Initiation and Development. Cell Mol Gastroenterol Hepatol 2021; 13:949-973.e7. [PMID: 34737104 PMCID: PMC8864474 DOI: 10.1016/j.jcmgh.2021.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Loss of Spectrin beta, non-erythrocytic 1 (SPTBN1) plays an important role in the carcinogenesis of hepatocellular carcinoma (HCC); however, the mechanisms underlying its involvement remain poorly understood. Defects in autophagy contribute to hepatic tumor formation. Hence, in this study, we explored the role and mechanism of SPTBN1 in the autophagy of hepatic stem cells (HSCs) and HCC cells. METHODS Expansion, autophagy, and malignant transformation of HSCs were detected in the injured liver of Sptbn1+/- mice induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine treatment. Hippo pathway and Yes-associated protein (YAP) stabilization were examined in isolated HSCs, Huh-7, and PLC/PRF/5 HCC cells and hepatocytes with or without loss of SPTBN1. RESULTS We found that heterozygous SPTBN1 knockout accelerated liver tumor development with 3,5-diethoxycarbonyl-1,4-dihydrocollidine induction. Rapamycin promoted autophagy in murine HSCs and reversed the increased malignant transformation induced by heterozygous SPTBN1 deletion. Loss of SPTBN1 also decreased autophagy and increased YAP stability and nuclear localization in human HCC cells and tissues, whereas YAP inhibition attenuated the effects of SPTBN1 deficiency on autophagy. Finally, we found that SPTBN1 positively regulated the expression of suppressor of variegation 3-9-enhancer of zeste-trithorax domain containing lysine methyltransferase 7 to promote YAP methylation, which may lead to YAP degradation and inactivation. CONCLUSIONS Our findings provide the first demonstration that loss of SPTBN1 impairs autophagy of HSCs to promote expansion and malignant transformation during hepatocarcinogenesis. SPTBN1 also cooperates with suppressor of variegation 3-9-enhancer of zeste-trithorax domain containing lysine methyltransferase 7 to inactive YAP, resulting in enhanced autophagy of HCC cells. These results may open new avenues targeting SPTBN1 for the prevention and treatment of HCC.
Collapse
Affiliation(s)
- Shuyi Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Huijie Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhengyang Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mengping Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jieyu Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiayu Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ling Lin
- Department of Medicine and Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Aiwu Ruth He
- Department of Medicine and Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.
| | - Ping Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
39
|
Fas BA, Maiani E, Sora V, Kumar M, Mashkoor M, Lambrughi M, Tiberti M, Papaleo E. The conformational and mutational landscape of the ubiquitin-like marker for autophagosome formation in cancer. Autophagy 2021; 17:2818-2841. [PMID: 33302793 PMCID: PMC8525936 DOI: 10.1080/15548627.2020.1847443] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
Macroautophagy/autophagy is a cellular process to recycle damaged cellular components, and its modulation can be exploited for disease treatments. A key autophagy player is the ubiquitin-like protein MAP1LC3B/LC3B. Mutations and changes in MAP1LC3B expression occur in cancer samples. However, the investigation of the effects of these mutations on MAP1LC3B protein structure is still missing. Despite many LC3B structures that have been solved, a comprehensive study, including dynamics, has not yet been undertaken. To address this knowledge gap, we assessed nine physical models for biomolecular simulations for their capabilities to describe the structural ensemble of MAP1LC3B. With the resulting MAP1LC3B structural ensembles, we characterized the impact of 26 missense mutations from pan-cancer studies with different approaches, and we experimentally validated our prediction for six variants using cellular assays. Our findings shed light on damaging or neutral mutations in MAP1LC3B, providing an atlas of its modifications in cancer. In particular, P32Q mutation was found detrimental for protein stability with a propensity to aggregation. In a broader context, our framework can be applied to assess the pathogenicity of protein mutations or to prioritize variants for experimental studies, allowing to comprehensively account for different aspects that mutational events alter in terms of protein structure and function.Abbreviations: ATG: autophagy-related; Cα: alpha carbon; CG: coarse-grained; CHARMM: Chemistry at Harvard macromolecular mechanics; CONAN: contact analysis; FUNDC1: FUN14 domain containing 1; FYCO1: FYVE and coiled-coil domain containing 1; GABARAP: GABA type A receptor-associated protein; GROMACS: Groningen machine for chemical simulations; HP: hydrophobic pocket; LIR: LC3 interacting region; MAP1LC3B/LC3B microtubule associated protein 1 light chain 3 B; MD: molecular dynamics; OPTN: optineurin; OSF: open software foundation; PE: phosphatidylethanolamine, PLEKHM1: pleckstrin homology domain-containing family M 1; PSN: protein structure network; PTM: post-translational modification; SA: structural alphabet; SLiM: short linear motif; SQSTM1/p62: sequestosome 1; WT: wild-type.
Collapse
Affiliation(s)
- Burcu Aykac Fas
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Emiliano Maiani
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Valentina Sora
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Mukesh Kumar
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maliha Mashkoor
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Lambrughi
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Tiberti
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
- Translational Disease Systems Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Sun T, Peng H, Mao W, Ma L, Liu H, Mai J, Jiao L. Autophagy-mediated negative feedback attenuates the oncogenic activity of YAP in pancreatic cancer. Int J Biol Sci 2021; 17:3634-3645. [PMID: 34512171 PMCID: PMC8416727 DOI: 10.7150/ijbs.61795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/11/2021] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal malignancy in humans, and new therapeutic targets are urgently needed. Yes-associated protein (YAP) plays a significant role in cancer progression. Autophagy is also closely associated with various human cancers. However, the interplay between YAP and autophagy in PDAC remains poorly understood. In this study, we found that YAP was upregulated and activated in PDAC. Further analysis revealed that there is a YAP-autophagy feedback loop in pancreatic cancer. Mechanistically, YAP activates autophagy by promoting Atg5 transcription via TEAD1-mediated binding, while autophagy negatively regulates YAP through autophagic degradation. The hyperactivation of YAP in PDAC unbalances the YAP-autophagy circuit and promotes cancer progression. Inhibition of autophagy enhances the oncogenic activity of YAP in PDAC. The autophagy activator rapamycin promotes the antitumor effect of verteporfin, a YAP inhibitor. Therefore, our study elucidated the interaction between YAP and autophagy in PDAC and our results suggest that targeting the YAP-autophagy circuit may be a new therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hui Peng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wenhao Mao
- Department of Clinical Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jia Mai
- Department of Laboratory Medicine, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Jiao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
41
|
Feng C, Zhou R, Liu F, Wang T, Liu S, Shao Y. STK3-ALK, a Novel ALK Rearrangement in Non-Small Cell Lung Cancer With Sensitivity to Tyrosine Kinase Inhibitors: A Case Report. Front Oncol 2021; 11:700341. [PMID: 34490099 PMCID: PMC8417525 DOI: 10.3389/fonc.2021.700341] [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: 04/26/2021] [Accepted: 07/21/2021] [Indexed: 12/24/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) rearrangement occurs in 5% to 8% of patients with non-small cell lung cancer (NSCLC). More than 90 different ALK fusion partners have been discovered in NSCLC patients, and ALK tyrosine kinase inhibitors (TKIs) such as crizotinib and alectinib have achieved tumor responses in patients with advanced ALK-positive NSCLC. Here, we report the case of a patient with an advanced NSCLC carrying a novel serine/threonine kinase 3 (STK3)-ALK rearrangement, which was identified by targeted next-generation sequencing (NGS) and was confirmed by RNA sequencing. Anti-ALK immunohistochemistry (IHC) staining also revealed the high expression of ALK. The patient benefitted from alectinib treatment after experiencing crizotinib resistance and achieved an overall response to TKI of over 14 months. At the timepoint of submission of this manuscript, this patient is still receiving alectinib treatment with a good tolerance. This study provides meaningful insights into the potential treatment option for NSCLC patients with brain metastases harboring STK3-ALK fusions and highlights the advantages of NGS in rapidly identifying novel molecular targets.
Collapse
Affiliation(s)
- Chunlai Feng
- Department of Respiratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Rong Zhou
- Department of Respiratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Feng Liu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Tingting Wang
- Department of Research and Development, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Sisi Liu
- Department of Research and Development, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Yang Shao
- Department of Research and Development, Nanjing Geneseeq Technology Inc., Nanjing, China
| |
Collapse
|
42
|
Yuan T, Annamalai K, Naik S, Lupse B, Geravandi S, Pal A, Dobrowolski A, Ghawali J, Ruhlandt M, Gorrepati KDD, Azizi Z, Lim DS, Maedler K, Ardestani A. The Hippo kinase LATS2 impairs pancreatic β-cell survival in diabetes through the mTORC1-autophagy axis. Nat Commun 2021; 12:4928. [PMID: 34389720 PMCID: PMC8363615 DOI: 10.1038/s41467-021-25145-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetes results from a decline in functional pancreatic β-cells, but the molecular mechanisms underlying the pathological β-cell failure are poorly understood. Here we report that large-tumor suppressor 2 (LATS2), a core component of the Hippo signaling pathway, is activated under diabetic conditions and induces β-cell apoptosis and impaired function. LATS2 deficiency in β-cells and primary isolated human islets as well as β-cell specific LATS2 ablation in mice improves β-cell viability, insulin secretion and β-cell mass and ameliorates diabetes development. LATS2 activates mechanistic target of rapamycin complex 1 (mTORC1), a physiological suppressor of autophagy, in β-cells and genetic and pharmacological inhibition of mTORC1 counteracts the pro-apoptotic action of activated LATS2. We further show a direct interplay between Hippo and autophagy, in which LATS2 is an autophagy substrate. On the other hand, LATS2 regulates β-cell apoptosis triggered by impaired autophagy suggesting an existence of a stress-sensitive multicomponent cellular loop coordinating β-cell compensation and survival. Our data reveal an important role for LATS2 in pancreatic β-cell turnover and suggest LATS2 as a potential therapeutic target to improve pancreatic β-cell survival and function in diabetes.
Collapse
Affiliation(s)
- Ting Yuan
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Karthika Annamalai
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Shruti Naik
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Blaz Lupse
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Shirin Geravandi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Anasua Pal
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Jaee Ghawali
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Marina Ruhlandt
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Zahra Azizi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dae-Sik Lim
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.
| | - Amin Ardestani
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
43
|
Nieto-Torres JL, Shanahan SL, Chassefeyre R, Chaiamarit T, Zaretski S, Landeras-Bueno S, Verhelle A, Encalada SE, Hansen M. LC3B phosphorylation regulates FYCO1 binding and directional transport of autophagosomes. Curr Biol 2021; 31:3440-3449.e7. [PMID: 34146484 PMCID: PMC8439105 DOI: 10.1016/j.cub.2021.05.052] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 01/07/2021] [Accepted: 05/25/2021] [Indexed: 01/22/2023]
Abstract
Macroautophagy (hereafter referred to as autophagy) is a conserved process that promotes cellular homeostasis through the degradation of cytosolic components, also known as cargo. During autophagy, cargo is sequestered into double-membrane vesicles called autophagosomes, which are predominantly transported in the retrograde direction to the perinuclear region to fuse with lysosomes, thus ensuring cargo degradation.1 The mechanisms regulating directional autophagosomal transport remain unclear. The ATG8 family of proteins associates with autophagosome membranes2 and plays key roles in autophagy, including the movement of autophagosomes. This is achieved via the association of ATG8 with adaptor proteins like FYCO1, involved in the anterograde transport of autophagosomes toward the cell periphery.1,3-5 We previously reported that phosphorylation of LC3B/ATG8 on threonine 50 (LC3B-T50) by the Hippo kinase STK4/MST1 is required for autophagy through unknown mechanisms.6 Here, we show that STK4-mediated phosphorylation of LC3B-T50 reduces the binding of FYCO1 to LC3B. In turn, impairment of LC3B-T50 phosphorylation decreases starvation-induced perinuclear positioning of autophagosomes as well as their colocalization with lysosomes. Moreover, a significantly higher number of LC3B-T50A-positive autophagosomes undergo aberrant anterograde movement to axonal tips in mammalian neurons and toward the periphery of mammalian cells. Our data support a role of a nutrient-sensitive STK4-LC3B-FYCO1 axis in the regulation of the directional transport of autophagosomes, a key step of the autophagy process, via the post-translational modification of LC3B.
Collapse
Affiliation(s)
- Jose L Nieto-Torres
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sean-Luc Shanahan
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Romain Chassefeyre
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tai Chaiamarit
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sviatlana Zaretski
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sara Landeras-Bueno
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Adriaan Verhelle
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sandra E Encalada
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Malene Hansen
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
44
|
Hepowit NL, Kolbe CC, Zelle SR, Latz E, MacGurn JA. Regulation of ubiquitin and ubiquitin-like modifiers by phosphorylation. FEBS J 2021; 289:4797-4810. [PMID: 34214249 PMCID: PMC9271371 DOI: 10.1111/febs.16101] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/28/2021] [Accepted: 07/01/2021] [Indexed: 12/31/2022]
Abstract
The regulatory influence of ubiquitin is vast, encompassing all cellular processes, by virtue of its central roles in protein degradation, membrane trafficking, and cell signaling. But how does ubiquitin, a 76 amino acid peptide, carry out such diverse, complex functions in eukaryotic cells? Part of the answer is rooted in the high degree of complexity associated with ubiquitin polymers, which can be 'read' and processed differently depending on topology and cellular context. However, recent evidence indicates that post-translational modifications on ubiquitin itself enhance the complexity of the ubiquitin code. Here, we review recent discoveries related to the regulation of the ubiquitin code by phosphorylation. We summarize what is currently known about phosphorylation of ubiquitin at Ser65, Ser57, and Thr12, and we discuss the potential for phosphoregulation of ubiquitin at other sites. We also discuss accumulating evidence that ubiquitin-like modifiers, such as SUMO, are likewise regulated by phosphorylation. A complete understanding of these regulatory codes and their complex lexicon will require dissection of mechanisms that govern phosphorylation of ubiquitin and ubiquitin-like proteins, particularly in the context of cellular stress and disease.
Collapse
Affiliation(s)
- Nathaniel L Hepowit
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Carl-Christian Kolbe
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Germany
| | - Sarah R Zelle
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA, USA.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jason A MacGurn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
45
|
Carbamylated Erythropoietin-Fc (CEPO-Fc) ameliorates Aβ25-35 induced neurotoxicity by modulating autophagy, apoptosis, and necroptosis in Alzheimer's Disease model rats. PHYSIOLOGY AND PHARMACOLOGY 2021. [DOI: 10.52547/phypha.26.3.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
46
|
Licheva M, Raman B, Kraft C, Reggiori F. Phosphoregulation of the autophagy machinery by kinases and phosphatases. Autophagy 2021; 18:104-123. [PMID: 33970777 PMCID: PMC8865292 DOI: 10.1080/15548627.2021.1909407] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Eukaryotic cells use post-translational modifications to diversify and dynamically coordinate the function and properties of protein networks within various cellular processes. For example, the process of autophagy strongly depends on the balanced action of kinases and phosphatases. Highly conserved from the budding yeast Saccharomyces cerevisiae to humans, autophagy is a tightly regulated self-degradation process that is crucial for survival, stress adaptation, maintenance of cellular and organismal homeostasis, and cell differentiation and development. Many studies have emphasized the importance of kinases and phosphatases in the regulation of autophagy and identified many of the core autophagy proteins as their direct targets. In this review, we summarize the current knowledge on kinases and phosphatases acting on the core autophagy machinery and discuss the relevance of phosphoregulation for the overall process of autophagy.
Collapse
Affiliation(s)
- Mariya Licheva
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Babu Raman
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, AV Groningen, The Netherlands
| | - Claudine Kraft
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Fulvio Reggiori
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, AV Groningen, The Netherlands
| |
Collapse
|
47
|
Sun T, Chi JT. Regulation of ferroptosis in cancer cells by YAP/TAZ and Hippo pathways: The therapeutic implications. Genes Dis 2021; 8:241-249. [PMID: 33997171 PMCID: PMC8093643 DOI: 10.1016/j.gendis.2020.05.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022] Open
Abstract
Ferroptosis is a novel form of iron-dependent cell death characterized by lipid peroxidation. While the importance and disease relevance of ferroptosis is gaining recognition, much remains unknown about various genetic and non-genetic determinants of ferroptosis. Hippo signaling pathway is an evolutionarily conserved pathway that responds to various environmental cues and controls organ size, cell proliferation, death, and self-renewal capacity. In cancer biology, Hippo pathway is a potent tumor suppressing mechanism and its dysregulation contributes to apoptosis evasion, cancer development, metastasis, and treatment resistance. Hippo dysregulation leads to aberrant activation of YAP and TAZ, the two major transcription co-activators of TEADs, that induce the expression of genes triggering tumor-promoting phenotypes, including enhanced cell proliferation, self-renewal and apoptosis inhibition. The Hippo pathway is regulated by the cell-cell contact and cellular density/confluence. Recently, ferroptosis has also been found being regulated by the cellular contact and density. The YAP/TAZ activation under low density, while confers apoptosis resistance, renders cancer cells sensitivity to ferroptosis. These findings establish YAP/TAZ and Hippo pathways as novel determinants of ferroptosis. Therefore, inducing ferroptosis may have therapeutic potential for YAP/TAZ-activated chemo-resistant and metastatic tumor cells. Reciprocally, various YAP/TAZ-targeting treatments under clinical development may confer ferroptosis resistance, limiting the therapeutic efficacy.
Collapse
Affiliation(s)
- Tianai Sun
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
48
|
Molecular Basis of Neuronal Autophagy in Ageing: Insights from Caenorhabditis elegans. Cells 2021; 10:cells10030694. [PMID: 33800981 PMCID: PMC8004021 DOI: 10.3390/cells10030694] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/19/2023] Open
Abstract
Autophagy is an evolutionarily conserved degradation process maintaining cell homeostasis. Induction of autophagy is triggered as a response to a broad range of cellular stress conditions, such as nutrient deprivation, protein aggregation, organelle damage and pathogen invasion. Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane organelle referred to as the autophagosome with subsequent degradation of its contents upon delivery to lysosomes. Autophagy plays critical roles in development, maintenance and survival of distinct cell populations including neurons. Consequently, age-dependent decline in autophagy predisposes animals for age-related diseases including neurodegeneration and compromises healthspan and longevity. In this review, we summarize recent advances in our understanding of the role of neuronal autophagy in ageing, focusing on studies in the nematode Caenorhabditis elegans.
Collapse
|
49
|
Ma YC, Dai LL, Qiu BB, Zhou Y, Zhao YQ, Ran Y, Zhang KQ, Zou CG. TOR functions as a molecular switch connecting an iron cue with host innate defense against bacterial infection. PLoS Genet 2021; 17:e1009383. [PMID: 33657091 PMCID: PMC7928448 DOI: 10.1371/journal.pgen.1009383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 01/26/2021] [Indexed: 01/22/2023] Open
Abstract
As both host and pathogen require iron for survival, iron is an important regulator of host-pathogen interactions. However, the molecular mechanism by which how the availability of iron modulates host innate immunity against bacterial infections remains largely unknown. Using the metazoan Caenorhabditis elegans as a model, we demonstrate that infection with a pathogenic bacterium Salmonella enterica serovar Typhimurium induces autophagy by inactivating the target of rapamycin (TOR). Although the transcripts of ftn-1 and ftn-2 encoding two H-ferritin subunits are upregulated upon S. Typhimurium infection, the ferritin protein is kept at a low level due to its degradation mediated by autophagy. Autophagy, but not ferritin, is required for defense against S. Typhimurium infection under normal circumstances. Increased abundance of iron suppresses autophagy by activating TOR, leading to an increase in the ferritin protein level. Iron sequestration, but not autophagy, becomes pivotal to protect the host from S. Typhimurium infection in the presence of exogenous iron. Our results show that TOR acts as a regulator linking iron availability with host defense against bacterial infection. Iron, an essential nutrient for both hosts and pathogens, can impact host-pathogen interactions. Thus, depending on availability of iron, hosts may use distinct strategies to defend against bacterial infections. Using the model organism Caenorhabditis elegans, we show that autophagy is activated and required for resistance to S. Typhimurium infection under normal conditions. In the presence of exogenous iron, autophagy is suppressed, leading to an increase in the protein level of ferritin, which in turn protects worms against S. Typhimurium infection. Finally, our data demonstrate that TOR is a molecular switch for turning autophagy and ferritin on and off. Thus, our study reveals a previously unknown role of TOR in modulating the transition between autophagy and iron sequestration in response to a bacterial infection via sensing an iron cue.
Collapse
Affiliation(s)
- Yi-Cheng Ma
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Li-Li Dai
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
- School of Agronomy and Life Sciences, Kunming University, Kunming, Yunnan, China
| | - Bei-Bei Qiu
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Ying Zhou
- College of Chemical Science and Technology, Yunnan University, Kunming, Yunnan, China
| | - Yu-Qiang Zhao
- College of Chemical Science and Technology, Yunnan University, Kunming, Yunnan, China
| | - Yu Ran
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Ke-Qin Zhang
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
- * E-mail: (K-QZ); (C-GZ)
| | - Cheng-Gang Zou
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
- * E-mail: (K-QZ); (C-GZ)
| |
Collapse
|
50
|
Cho YK, Son Y, Saha A, Kim D, Choi C, Kim M, Park JH, Im H, Han J, Kim K, Jung YS, Yun J, Bae EJ, Seong JK, Lee MO, Lee S, Granneman JG, Lee YH. STK3/STK4 signalling in adipocytes regulates mitophagy and energy expenditure. Nat Metab 2021; 3:428-441. [PMID: 33758424 DOI: 10.1038/s42255-021-00362-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/12/2021] [Indexed: 11/08/2022]
Abstract
Obesity reduces adipocyte mitochondrial function, and expanding adipocyte oxidative capacity is an emerging strategy to improve systemic metabolism. Here, we report that serine/threonine-protein kinase 3 (STK3) and STK4 are key physiological suppressors of mitochondrial capacity in brown, beige and white adipose tissues. Levels of STK3 and STK4, kinases in the Hippo signalling pathway, are greater in white than brown adipose tissues, and levels in brown adipose tissue are suppressed by cold exposure and greatly elevated by surgical denervation. Genetic inactivation of Stk3 and Stk4 increases mitochondrial mass and function, stabilizes uncoupling protein 1 in beige adipose tissue and confers resistance to metabolic dysfunction induced by high-fat diet feeding. Mechanistically, STK3 and STK4 increase adipocyte mitophagy in part by regulating the phosphorylation and dimerization status of the mitophagy receptor BNIP3. STK3 and STK4 expression levels are elevated in human obesity, and pharmacological inhibition improves metabolic profiles in a mouse model of obesity, suggesting STK3 and STK4 as potential targets for treating obesity-related diseases.
Collapse
Affiliation(s)
- Yoon Keun Cho
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yeonho Son
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Abhirup Saha
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Doeun Kim
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Cheoljun Choi
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Minsu Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hyun Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyeonyeong Im
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Juhyeong Han
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Kyungmin Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Jeanho Yun
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Eun Ju Bae
- College of Pharmacy, Chonbuk National University, Jeonju, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yun-Hee Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
- Bio-Max Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|