1
|
Choi H, Park SW, Jang DJ, Lee JA. Development of selective deconjugases for membrane-anchored LC3A/B in post-mitotic neurons. Mol Brain 2025; 18:11. [PMID: 39940005 PMCID: PMC11823225 DOI: 10.1186/s13041-025-01184-z] [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: 11/29/2024] [Accepted: 02/06/2025] [Indexed: 02/14/2025] Open
Abstract
Neuronal autophagy is essential for maintaining protein and organelle turnover, thereby safeguarding neuronal health. LC3, a central autophagy protein, exists in lipidated (LC3-II) and non-lipidated (LC3-I) forms, both critical for neurons due to their sensitivity to metabolic and proteostatic stress. To elucidate the specific roles of membrane-anchored LC3A/B in post-mitotic neurons, we engineered deconjugases with enhanced selectivity for lipidated LC3. By modifying LC3-interacting regions (LIRs) at the deconjugase termini, we significantly improved targeting specificity toward LC3A/B. Deconjugases with N-terminal LIR modifications reduced LC3A/B-associated autophagosomes, highlighting the importance of LIR positioning for specificity. Sequential N-terminal LIR arrangements further refined LC3A/B targeting without affecting GABARAP-associated autophagosomes. Moreover, reducing the hydrophobicity of the α3 helix to limit membrane residence time further improved selectivity. These targeted modifications demonstrate the potential of customized deconjugases to dissect and modulate specific autophagic pathways in neurons, paving the way for novel therapeutic strategies against neurodegenerative diseases associated with autophagy dysregulation.
Collapse
Affiliation(s)
- Haneul Choi
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Korea
| | - Sang-Won Park
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju, Korea
| | - Deok-Jin Jang
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju, Korea.
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju, Korea.
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Korea.
| |
Collapse
|
2
|
Mukherjee R, Bhattacharya A, Mello-Vieira J, Kuncha SK, Hoffmann M, Gonzalez A, Rathore R, Chadha A, Shin D, Colby T, Matic I, Mukherjee S, Misra M, Dikic I. Serine ubiquitination of SQSTM1 regulates NFE2L2-dependent redox homeostasis. Autophagy 2025; 21:407-423. [PMID: 39291751 PMCID: PMC11759625 DOI: 10.1080/15548627.2024.2404375] [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: 04/03/2023] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024] Open
Abstract
The KEAP1-NFE2L2 axis is essential for the cellular response against metabolic and oxidative stress. KEAP1 is an adaptor protein of CUL3 (cullin 3) ubiquitin ligase that controls the cellular levels of NFE2L2, a critical transcription factor of several cytoprotective genes. Oxidative stress, defective autophagy and pathogenic infections activate NFE2L2 signaling through phosphorylation of the autophagy receptor protein SQSTM1, which competes with NFE2L2 for binding to KEAP1. Here we show that phosphoribosyl-linked serine ubiquitination of SQSTM1 catalyzed by SidE effectors of Legionella pneumophila controls NFE2L2 signaling and cell metabolism upon Legionella infection. Serine ubiquitination of SQSTM1 sterically blocks its binding to KEAP1, resulting in NFE2L2 ubiquitination and degradation. This reduces NFE2L2-dependent antioxidant synthesis in the early phase of infection. Levels of serine ubiquitinated SQSTM1 diminish in the later stage of infection allowing the expression of NFE2L2-target genes; causing a differential regulation of the host metabolome and proteome in a NFE2L2-dependent manner.Abbreviation: ARE: antioxidant response element; Dup: deubiquitinase specific for phosphoribosyl-linked serine ubiquitination; ER: endoplasmic reticulum; h.p.i: hours post infection; HIF1A/HIF-1α: hypoxia inducible factor 1 subunit alpha; KEAP1: kelch like ECH associated protein 1; KIR: KEAP1-interacting region; LIR: LC3-interacting region; NES: nuclear export signal; NFKB/NF-κB: nuclear factor kappa B; NLS: nuclear localization signal; NFE2L2/Nrf2: NFE2 like bZIP transcription factor 2; PB1 domain: Phox1 and Bem1 domain; PR-Ub: phosphoribosyl-linked serine ubiquitination; ROS: reactive oxygen species; SQSTM1/p62: sequestosome 1; tBHQ: tertiary butylhydroquinone; TUBE2: tandem ubiquitiin binding entity 2; UBA domain: ubiquitin-associated domain.
Collapse
Affiliation(s)
- Rukmini Mukherjee
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Molecular Signaling, Goethe University, Frankfurt, Germany
- Biophysics, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Anshu Bhattacharya
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Molecular Signaling, Goethe University, Frankfurt, Germany
| | - João Mello-Vieira
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Molecular Signaling, Goethe University, Frankfurt, Germany
| | - Santosh Kumar Kuncha
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Molecular Signaling, Goethe University, Frankfurt, Germany
| | - Marina Hoffmann
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Alexis Gonzalez
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Rajeshwari Rathore
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Attinder Chadha
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
- The George William Hooper Foundation, University of California, San Francisco, USA
| | - Donghyuk Shin
- Molecular Signaling, Goethe University, Frankfurt, Germany
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Thomas Colby
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Ivan Matic
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Shaeri Mukherjee
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
- The George William Hooper Foundation, University of California, San Francisco, USA
- Chan Zuckerberg Biohub, San Francisco, USA
| | - Mohit Misra
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Molecular Signaling, Goethe University, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Molecular Signaling, Goethe University, Frankfurt, Germany
- Biophysics, Max Planck Institute of Biophysics, Frankfurt, Germany
- Translational Medicine and Pharmacology, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
| |
Collapse
|
3
|
Yang Y, Zhao Z, Li X, Chen Y, Liu L, Zhang SL, Yang A. Design and Evaluation of Peptide Inhibitors Targeting the Dimerization of SARS-CoV-2 Main Protease. Chembiochem 2025; 26:e202400688. [PMID: 39417610 DOI: 10.1002/cbic.202400688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024]
Abstract
The severe acute respiratory syndrome virus 2 (SARS-CoV-2) seriously impacted public health. The evolutionarily conserved viral chymotrypsin-like main protease (Mpro) is an important target for anti-SARS-CoV-2 drug development. Previous studies have shown that the eight N-terminal amino acids (N8) of SARS-CoV Mpro are essential for its dimerization, and are used to design inhibitors against SARS-CoV Mpro dimerization. Here, we established a simple readout assay using SDS-PAGE and Coomassie blue staining to measure inhibitory activity of N8 peptide derived from SARS-CoV-2 Mpro. To optimize its inhibitory effect, we then modified the side-chain length, charge, and hydrophilicity of the N8 peptide, and introduced a mutated Mpro recognition sequence. As a result, we obtained a series of potent peptide inhibitors against SARS-CoV-2 Mpro, with N8-A24 being the most efficient with an IC50 value of 1.44 mM. We observed that N8-A24 reduced Mpro dimerization with an IC50 value of 0.86 mM. Molecular docking revealed that N8-A24 formed hydrogen bond interactions with critical dimeric interface residues, thus inhibiting its dimerization and activity. In conclusion, our study not only discovers a series of peptide inhibitors targeting the SARS-CoV-2 Mpro dimerization, but also provides a promising strategy for the rational design of new inhibitors against COVID-19.
Collapse
Affiliation(s)
- Yi Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Zhiyi Zhao
- School of Life Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Xiaoying Li
- School of Life Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Yian Chen
- School of Life Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Lu Liu
- School of Life Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, P. R. China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, P. R. China
| |
Collapse
|
4
|
Park SW, Park JH, Choi H, Jeon P, Lee SH, Shin WD, Kim HJ, Lee JA, Jang DJ. Differential roles of N- and C-terminal LIR motifs in the catalytic activity and membrane targeting of RavZ and ATG4B proteins. BMB Rep 2024; 57:497-502. [PMID: 39384175 PMCID: PMC11608851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/08/2024] [Accepted: 09/02/2024] [Indexed: 10/11/2024] Open
Abstract
Mammalian ATG8 proteins (mATG8s) are essential for selective autophagy because they recruit various proteins with LC3- interacting region (LIR) motifs to autophagic membranes. The RavZ protein, secreted by Legionella pneumophila, and mammalian ATG4B possess functional LIR motifs that participate in lipidated mATG8 deconjugation on autophagic membranes. RavZ comprises three functional LIR motifs at the N- and Cterminal sides of its catalytic domain (CAD). This study demonstrated that LIR motifs at the N-terminal side of the CAD of RavZ are involved in autophagic membrane targeting and substrate recognition, while LIR motif at the C-terminal side facilitate autophagic membrane targeting. Our results also revealed that the C-terminal LIR motif in human ATG4B is pivotal in delipidating LC3B-phosphatidylethanolamine (PE), but it plays a minor role in pro-LC3B priming in the cytosol. Therefore, introducing a functional LIR motif to the N-terminal of ATG4B does not affect LC3B-PE delipidation. This study clearly described the position-dependent roles of LIR motifs in RavZ and ATG4B in cellular contexts. [BMB Reports 2024; 57(11): 497-502].
Collapse
Affiliation(s)
- Sang-Won Park
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju 37224, Korea
| | - Ju-Hui Park
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Haneul Choi
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Pureum Jeon
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Seung-Hwan Lee
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Won-Dong Shin
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju 37224, Korea
| | - Hun-Joo Kim
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Deok-Jin Jang
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju 37224, Korea
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| |
Collapse
|
5
|
Albani M, Fassi EMA, Moretti RM, Garofalo M, Montagnani Marelli M, Roda G, Sgrignani J, Cavalli A, Grazioso G. Computational Design of Novel Cyclic Peptides Endowed with Autophagy-Inhibiting Activity on Cancer Cell Lines. Int J Mol Sci 2024; 25:4622. [PMID: 38731842 PMCID: PMC11083565 DOI: 10.3390/ijms25094622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/17/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
(1) Autophagy plays a significant role in development and cell proliferation. This process is mainly accomplished by the LC3 protein, which, after maturation, builds the nascent autophagosomes. The inhibition of LC3 maturation results in the interference of autophagy activation. (2) In this study, starting from the structure of a known LC3B binder (LIR2-RavZ peptide), we identified new LC3B ligands by applying an in silico drug design strategy. The most promising peptides were synthesized, biophysically assayed, and biologically evaluated to ascertain their potential antiproliferative activity on five humans cell lines. (3) A cyclic peptide (named Pep6), endowed with high conformational stability (due to the presence of a disulfide bridge), displayed a Kd value on LC3B in the nanomolar range. Assays accomplished on PC3, MCF-7, and A549 cancer cell lines proved that Pep6 exhibited cytotoxic effects comparable to those of the peptide LIR2-RavZ, a reference LC3B ligand. Furthermore, it was ineffective on both normal prostatic epithelium PNT2 and autophagy-defective prostate cancer DU145 cells. (4) Pep6 can be considered a new autophagy inhibitor that can be employed as a pharmacological tool or even as a template for the rational design of new small molecules endowed with autophagy inhibitory activity.
Collapse
Affiliation(s)
- Marco Albani
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milano, Italy; (M.A.); (G.R.)
| | - Enrico Mario Alessandro Fassi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milano, Italy; (M.A.); (G.R.)
| | - Roberta Manuela Moretti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (R.M.M.); (M.M.M.)
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, Università di Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Marina Montagnani Marelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (R.M.M.); (M.M.M.)
| | - Gabriella Roda
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milano, Italy; (M.A.); (G.R.)
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine (IRB), Via Chiesa 5, 6500 Bellinzona, Switzerland; (J.S.); (A.C.)
| | - Andrea Cavalli
- Institute for Research in Biomedicine (IRB), Via Chiesa 5, 6500 Bellinzona, Switzerland; (J.S.); (A.C.)
- Swiss Institute of Bioinformatics (SIB), University of Lausanne, Quartier UNIL-Sorge, Bâtiment Amphipôle, 1015 Lausanne, Switzerland
| | - Giovanni Grazioso
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milano, Italy; (M.A.); (G.R.)
| |
Collapse
|
6
|
Graham CI, MacMartin TL, de Kievit TR, Brassinga AKC. Molecular regulation of virulence in Legionella pneumophila. Mol Microbiol 2024; 121:167-195. [PMID: 37908155 DOI: 10.1111/mmi.15172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/15/2023] [Accepted: 09/17/2023] [Indexed: 11/02/2023]
Abstract
Legionella pneumophila is a gram-negative bacteria found in natural and anthropogenic aquatic environments such as evaporative cooling towers, where it reproduces as an intracellular parasite of cohabiting protozoa. If L. pneumophila is aerosolized and inhaled by a susceptible person, bacteria may colonize their alveolar macrophages causing the opportunistic pneumonia Legionnaires' disease. L. pneumophila utilizes an elaborate regulatory network to control virulence processes such as the Dot/Icm Type IV secretion system and effector repertoire, responding to changing nutritional cues as their host becomes depleted. The bacteria subsequently differentiate to a transmissive state that can survive in the environment until a replacement host is encountered and colonized. In this review, we discuss the lifecycle of L. pneumophila and the molecular regulatory network that senses nutritional depletion via the stringent response, a link to stationary phase-like metabolic changes via alternative sigma factors, and two-component systems that are homologous to stress sensors in other pathogens, to regulate differentiation between the intracellular replicative phase and more transmissible states. Together, we highlight how this prototypic intracellular pathogen offers enormous potential in understanding how molecular mechanisms enable intracellular parasitism and pathogenicity.
Collapse
Affiliation(s)
- Christopher I Graham
- Department of Microbiology, Faculty of Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Teassa L MacMartin
- Department of Microbiology, Faculty of Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Teresa R de Kievit
- Department of Microbiology, Faculty of Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ann Karen C Brassinga
- Department of Microbiology, Faculty of Science, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
7
|
Yang Y, Mei L, Chen J, Chen X, Wang Z, Liu L, Yang A. Legionella pneumophila-mediated host posttranslational modifications. J Mol Cell Biol 2023; 15:mjad032. [PMID: 37156500 PMCID: PMC10720952 DOI: 10.1093/jmcb/mjad032] [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: 09/16/2022] [Revised: 01/17/2023] [Accepted: 05/06/2023] [Indexed: 05/10/2023] Open
Abstract
Legionella pneumophila is a Gram-negative bacterium ubiquitously present in freshwater environments and causes a serious type of pneumonia called Legionnaires' disease. During infections, L. pneumophila releases over 300 effector proteins into host cells through an Icm/Dot type IV secretion system to manipulate the host defense system for survival within the host. Notably, certain effector proteins mediate posttranslational modifications (PTMs), serving as useful approaches exploited by L. pneumophila to modify host proteins. Some effectors catalyze the addition of host protein PTMs, while others mediate the removal of PTMs from host proteins. In this review, we summarize L. pneumophila effector-mediated PTMs of host proteins, including phosphorylation, ubiquitination, glycosylation, AMPylation, phosphocholination, methylation, and ADP-ribosylation, as well as dephosphorylation, deubiquitination, deAMPylation, deADP-ribosylation, dephosphocholination, and delipidation. We describe their molecular mechanisms and biological functions in the regulation of bacterial growth and Legionella-containing vacuole biosynthesis and in the disruption of host immune and defense machinery.
Collapse
Affiliation(s)
- Yi Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Ligang Mei
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Jing Chen
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaorong Chen
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Zhuolin Wang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Lu Liu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
8
|
Tang Z, Jiang W, Li S, Huang X, Yang Y, Chen X, Qiu J, Xiao C, Xie Y, Zhang X, Li J, Verma CS, He Y, Yang A. Design and evaluation of tadpole-like conformational antimicrobial peptides. Commun Biol 2023; 6:1177. [PMID: 37980400 PMCID: PMC10657444 DOI: 10.1038/s42003-023-05560-0] [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: 07/03/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023] Open
Abstract
Antimicrobial peptides are promising alternatives to conventional antibiotics. Herein, we report a class of "tadpole-like" peptides consisting of an amphipathic α-helical head and an aromatic tail. A structure-activity relationship (SAR) study of "tadpole-like" temporin-SHf and its analogs revealed that increasing the number of aromatic residues in the tail, introducing Arg to the α-helical head and rearranging the peptide topology dramatically increased antimicrobial activity. Through progressive structural optimization, we obtained two peptides, HT2 and RI-HT2, which exhibited potent antimicrobial activity, no hemolytic activity and cytotoxicity, and no propensity to induce resistance. NMR and molecular dynamics simulations revealed that both peptides indeed adopted "tadpole-like" conformations. Fluorescence experiments and electron microscopy confirmed the membrane targeting mechanisms of the peptides. Our studies not only lead to the discovery of a series of ultrashort peptides with potent broad-spectrum antimicrobial activities, but also provide a new strategy for rational design of novel "tadpole-like" antimicrobial peptides.
Collapse
Affiliation(s)
- Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Wuqiao Jiang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Shuangli Li
- National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xue Huang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Yi Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaorong Chen
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Jingyi Qiu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Chuyu Xiao
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Ying Xie
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Xu Zhang
- National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jianguo Li
- Bioinformatics Institute, A∗STAR, 30 Biopolis Street, Matrix, Singapore, 138671, Singapore
- Singapore Eye Research Institute, Singapore, 169856, Singapore
| | - Chandra Shekhar Verma
- Bioinformatics Institute, A∗STAR, 30 Biopolis Street, Matrix, Singapore, 138671, Singapore
- Department of Biological Sciences, National University of, Singapore, 117543, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Yun He
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China.
- BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China.
| |
Collapse
|
9
|
Liu L, Gray JL, Tate EW, Yang A. Bacterial enzymes: powerful tools for protein labeling, cell signaling, and therapeutic discovery. Trends Biotechnol 2023; 41:1385-1399. [PMID: 37328400 DOI: 10.1016/j.tibtech.2023.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 06/18/2023]
Abstract
Bacteria have evolved a diverse set of enzymes that enable them to subvert host defense mechanisms as well as to form part of the prokaryotic immune system. Due to their unique and varied biochemical activities, these bacterial enzymes have emerged as key tools for understanding and investigating biological systems. In this review, we summarize and discuss some of the most prominent bacterial enzymes used for the site-specific modification of proteins, in vivo protein labeling, proximity labeling, interactome mapping, signaling pathway manipulation, and therapeutic discovery. Finally, we provide a perspective on the complementary advantages and limitations of using bacterial enzymes compared with chemical probes for exploring biological systems.
Collapse
Affiliation(s)
- Lu Liu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Janine L Gray
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK.
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
10
|
Nagdev PK, Agnivesh PK, Roy A, Sau S, Kalia NP. Exploring and exploiting the host cell autophagy during Mycobacterium tuberculosis infection. Eur J Clin Microbiol Infect Dis 2023; 42:1297-1315. [PMID: 37740791 DOI: 10.1007/s10096-023-04663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/06/2023] [Indexed: 09/25/2023]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis, is a fatal infectious disease that prevails to be the second leading cause of death from a single infectious agent despite the availability of multiple drugs for treatment. The current treatment regimen involves the combination of several drugs for 6 months that remain ineffective in completely eradicating the infection because of several drawbacks, such as the long duration of treatment and the side effects of drugs causing non-adherence of patients to the treatment regimen. Autophagy is an intracellular degradative process that eliminates pathogens at the early stages of infection. Mycobacterium tuberculosis's unique autophagy-blocking capability makes it challenging to eliminate compared to usual pathogens. The present review discusses recent advances in autophagy-inhibiting factors and mechanisms that could be exploited to identify autophagy-inducing chemotherapeutics that could be used as adjunctive therapy with the existing first-line anti-TB agent to shorten the duration of therapy and enhance cure rates from multidrug-resistant tuberculosis (MDR-TB) and extreme drug-resistant tuberculosis (XDR-TB).
Collapse
Affiliation(s)
- Pavan Kumar Nagdev
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Puja Kumari Agnivesh
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Arnab Roy
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Shashikanta Sau
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Nitin Pal Kalia
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
11
|
Huang X, Yao J, Liu L, Chen J, Mei L, Huangfu J, Luo D, Wang X, Lin C, Chen X, Yang Y, Ouyang S, Wei F, Wang Z, Zhang S, Xiang T, Neculai D, Sun Q, Kong E, Tate EW, Yang A. S-acylation of p62 promotes p62 droplet recruitment into autophagosomes in mammalian autophagy. Mol Cell 2023; 83:3485-3501.e11. [PMID: 37802024 PMCID: PMC10552648 DOI: 10.1016/j.molcel.2023.09.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/22/2023] [Accepted: 09/07/2023] [Indexed: 10/08/2023]
Abstract
p62 is a well-characterized autophagy receptor that recognizes and sequesters specific cargoes into autophagosomes for degradation. p62 promotes the assembly and removal of ubiquitinated proteins by forming p62-liquid droplets. However, it remains unclear how autophagosomes efficiently sequester p62 droplets. Herein, we report that p62 undergoes reversible S-acylation in multiple human-, rat-, and mouse-derived cell lines, catalyzed by zinc-finger Asp-His-His-Cys S-acyltransferase 19 (ZDHHC19) and deacylated by acyl protein thioesterase 1 (APT1). S-acylation of p62 enhances the affinity of p62 for microtubule-associated protein 1 light chain 3 (LC3)-positive membranes and promotes autophagic membrane localization of p62 droplets, thereby leading to the production of small LC3-positive p62 droplets and efficient autophagic degradation of p62-cargo complexes. Specifically, increasing p62 acylation by upregulating ZDHHC19 or by genetic knockout of APT1 accelerates p62 degradation and p62-mediated autophagic clearance of ubiquitinated proteins. Thus, the protein S-acylation-deacylation cycle regulates p62 droplet recruitment to the autophagic membrane and selective autophagic flux, thereby contributing to the control of selective autophagic clearance of ubiquitinated proteins.
Collapse
Affiliation(s)
- Xue Huang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Jia Yao
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Lu Liu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Jing Chen
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Ligang Mei
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Jingjing Huangfu
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Dong Luo
- School of Pharmacy, Chongqing University, Chongqing 401331, China
| | - Xinyi Wang
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China; Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Changhai Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xiaorong Chen
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yi Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Sheng Ouyang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Fujing Wei
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Zhuolin Wang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Shaolin Zhang
- School of Pharmacy, Chongqing University, Chongqing 401331, China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Dante Neculai
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Qiming Sun
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China; Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Eryan Kong
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Edward W Tate
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
12
|
Roberts CG, Franklin TG, Pruneda JN. Ubiquitin-targeted bacterial effectors: rule breakers of the ubiquitin system. EMBO J 2023; 42:e114318. [PMID: 37555693 PMCID: PMC10505922 DOI: 10.15252/embj.2023114318] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023] Open
Abstract
Regulation through post-translational ubiquitin signaling underlies a large portion of eukaryotic biology. This has not gone unnoticed by invading pathogens, many of which have evolved mechanisms to manipulate or subvert the host ubiquitin system. Bacteria are particularly adept at this and rely heavily upon ubiquitin-targeted virulence factors for invasion and replication. Despite lacking a conventional ubiquitin system of their own, many bacterial ubiquitin regulators loosely follow the structural and mechanistic rules established by eukaryotic ubiquitin machinery. Others completely break these rules and have evolved novel structural folds, exhibit distinct mechanisms of regulation, or catalyze foreign ubiquitin modifications. Studying these interactions can not only reveal important aspects of bacterial pathogenesis but also shed light on unexplored areas of ubiquitin signaling and regulation. In this review, we discuss the methods by which bacteria manipulate host ubiquitin and highlight aspects that follow or break the rules of ubiquitination.
Collapse
Affiliation(s)
- Cameron G Roberts
- Department of Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Tyler G Franklin
- Department of Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Jonathan N Pruneda
- Department of Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| |
Collapse
|
13
|
Farnung J, Muhar M, Liang JR, Tolmachova KA, Benoit RM, Corn JE, Bode JW. Semisynthetic LC3 Probes for Autophagy Pathways Reveal a Noncanonical LC3 Interacting Region Motif Crucial for the Enzymatic Activity of Human ATG3. ACS CENTRAL SCIENCE 2023; 9:1025-1034. [PMID: 37252361 PMCID: PMC10214526 DOI: 10.1021/acscentsci.3c00009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Indexed: 05/31/2023]
Abstract
Macroautophagy is one of two major degradation systems in eukaryotic cells. Regulation and control of autophagy are often achieved through the presence of short peptide sequences called LC3 interacting regions (LIR) in autophagy-involved proteins. Using a combination of new protein-derived activity-based probes prepared from recombinant LC3 proteins, along with protein modeling and X-ray crystallography of the ATG3-LIR peptide complex, we identified a noncanonical LIR motif in the human E2 enzyme responsible for LC3 lipidation, ATG3. The LIR motif is present in the flexible region of ATG3 and adopts an uncommon β-sheet structure binding to the backside of LC3. We show that the β-sheet conformation is crucial for its interaction with LC3 and used this insight to design synthetic macrocyclic peptide-binders to ATG3. CRISPR-enabled in cellulo studies provide evidence that LIRATG3 is required for LC3 lipidation and ATG3∼LC3 thioester formation. Removal of LIRATG3 negatively impacts the rate of thioester transfer from ATG7 to ATG3.
Collapse
Affiliation(s)
- Jakob Farnung
- Laboratory
for Organic Chemistry, Department of Chemistry and Applied Biosciences ETH Zürich, CH-8093 Zürich, Switzerland
| | - Matthias Muhar
- Institute
of Molecular Health Sciences, Department of Biology ETH Zürich, CH-8093 Zürich, Switzerland
| | - Jin Rui Liang
- Institute
of Molecular Health Sciences, Department of Biology ETH Zürich, CH-8093 Zürich, Switzerland
| | - Kateryna A. Tolmachova
- Laboratory
for Organic Chemistry, Department of Chemistry and Applied Biosciences ETH Zürich, CH-8093 Zürich, Switzerland
| | - Roger M. Benoit
- Laboratory
of Nanoscale Biology, Division of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland
| | - Jacob E. Corn
- Institute
of Molecular Health Sciences, Department of Biology ETH Zürich, CH-8093 Zürich, Switzerland
| | - Jeffrey W. Bode
- Laboratory
for Organic Chemistry, Department of Chemistry and Applied Biosciences ETH Zürich, CH-8093 Zürich, Switzerland
| |
Collapse
|
14
|
Selvapandiyan A, Puri N, Kumar P, Alam A, Ehtesham NZ, Griffin G, Hasnain SE. Zooming in on common immune evasion mechanisms of pathogens in phagolysosomes: potential broad-spectrum therapeutic targets against infectious diseases. FEMS Microbiol Rev 2023; 47:6780197. [PMID: 36309472 DOI: 10.1093/femsre/fuac041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 01/19/2023] Open
Abstract
The intracellular viral, bacterial, or parasitic pathogens evade the host immune challenges to propagate and cause fatal diseases. The microbes overpower host immunity at various levels including during entry into host cells, phagosome formation, phagosome maturation, phagosome-lysosome fusion forming phagolysosomes, acidification of phagolysosomes, and at times after escape into the cytosol. Phagolysosome is the final organelle in the phagocyte with sophisticated mechanisms to degrade the pathogens. The immune evasion strategies by the pathogens include the arrest of host cell apoptosis, decrease in reactive oxygen species, the elevation of Th2 anti-inflammatory response, avoidance of autophagy and antigen cross-presentation pathways, and escape from phagolysosomal killing. Since the phagolysosome organelle in relation to infection/cure is seldom discussed in the literature, we summarize here the common host as well as pathogen targets manipulated or utilized by the pathogens established in phagosomes and phagolysosomes, to hijack the host immune system for their benefit. These common molecules or pathways can be broad-spectrum therapeutic targets for drug development for intervention against infectious diseases caused by different intracellular pathogens.
Collapse
Affiliation(s)
| | - Niti Puri
- Cellular and Molecular Immunology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pankaj Kumar
- Department of Biochemistry, Jamia Hamdard, New Delhi, 110062, India.,Centre for Tuberculosis Research, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21218, United States
| | - Anwar Alam
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India.,Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India
| | - Nasreen Zafar Ehtesham
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| | - George Griffin
- Department of Cellular and Molecular Medicine, St. George's University of London, London, SW17 0RE, United Kingdom
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India.,Department of Life Science, School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, 201310, India
| |
Collapse
|
15
|
Huppelschoten Y, van der Heden van Noort GJ. State of the art in (semi-)synthesis of Ubiquitin- and Ubiquitin-like tools. Semin Cell Dev Biol 2022; 132:74-85. [PMID: 34961664 DOI: 10.1016/j.semcdb.2021.11.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022]
Abstract
Protein ubiquitination is a key post-translational modification in regulating many fundamental cellular processes and dysregulation of these processes can give rise to a vast array of diseases. Unravelling the molecular mechanisms of ubiquitination hence is an important area in current ubiquitin research with as aim to understand this enigmatic process. The complexity of ubiquitin (Ub) signaling arises from the large variety of Ub conjugates, where Ub is attached to other Ub proteins, Ub-like proteins, and protein substrates. The chemical preparation of such Ub conjugates in high homogeneity and in adequate amounts contributes greatly to the deciphering of Ub signaling. The strength of these chemically synthesized conjugates lies in the chemo-selectivity in which they can be created that are sometimes difficult to obtain using biochemical methodology. In this review, we will discuss the progress in the chemical protein synthesis of state-of-the-art Ub and Ub-like chemical probes, their unique concepts and related discoveries in the ubiquitin field.
Collapse
Affiliation(s)
- Yara Huppelschoten
- Oncode Institute and Dept. Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands; Global Research Technologies, Novo Nordisk Research Park, Måløv, Denmark
| | | |
Collapse
|
16
|
Jia X, Knyazeva A, Zhang Y, Castro-Gonzalez S, Nakamura S, Carlson LA, Yoshimori T, Corkery DP, Wu YW. V. cholerae MakA is a cholesterol-binding pore-forming toxin that induces non-canonical autophagy. J Cell Biol 2022; 221:213518. [PMID: 36194176 PMCID: PMC9536202 DOI: 10.1083/jcb.202206040] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/16/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023] Open
Abstract
Pore-forming toxins (PFTs) are important virulence factors produced by many pathogenic bacteria. Here, we show that the Vibrio cholerae toxin MakA is a novel cholesterol-binding PFT that induces non-canonical autophagy in a pH-dependent manner. MakA specifically binds to cholesterol on the membrane at pH < 7. Cholesterol-binding leads to oligomerization of MakA on the membrane and pore formation at pH 5.5. Unlike other cholesterol-dependent cytolysins (CDCs) which bind cholesterol through a conserved cholesterol-binding motif (Thr-Leu pair), MakA contains an Ile-Ile pair that is essential for MakA-cholesterol interaction. Following internalization, endosomal acidification triggers MakA pore-assembly followed by ESCRT-mediated membrane repair and V-ATPase-dependent unconventional LC3 lipidation on the damaged endolysosomal membranes. These findings characterize a new cholesterol-binding toxin that forms pores in a pH-dependent manner and reveals the molecular mechanism of host autophagy manipulation.
Collapse
Affiliation(s)
- Xiaotong Jia
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Anastasia Knyazeva
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Yu Zhang
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Sergio Castro-Gonzalez
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Shuhei Nakamura
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Lars-Anders Carlson
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden,Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden,Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden,Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Dale P. Corkery
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden,Dale P. Corkery:
| | - Yao-Wen Wu
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden,Correspondence to Yao-Wen Wu:
| |
Collapse
|
17
|
Lactobacillusjohnsonii L531 Protects against Salmonella Infantis-Induced Intestinal Damage by Regulating the NOD Activation, Endoplasmic Reticulum Stress, and Autophagy. Int J Mol Sci 2022; 23:ijms231810395. [PMID: 36142312 PMCID: PMC9499332 DOI: 10.3390/ijms231810395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Salmonella enterica serovar Infantis (S. Infantis) is an intracellular bacterial pathogen. It is prevalent but resistant to antibiotics. Therefore, the therapeutic effect of antibiotics on Salmonella infection is limited. In this study, we used the piglet diarrhea model and the Caco2 cell model to explore the mechanism of probiotic Lactobacillus johnsonii L531 (L. johnsonii L531) against S. Infantis infection. L. johnsonii L531 attenuated S. Infantis-induced intestinal structural and cellular ultrastructural damage. The expression of NOD pathway-related proteins (NOD1/2, RIP2), autophagy-related key proteins (ATG16L1, IRGM), and endoplasmic reticulum (ER) stress markers (GRP78, IRE1) were increased after S. Infantis infection. Notably, L. johnsonii L531 pretreatment not only inhibited the activation of the above signaling pathways but also played an anti-S. Infantis infection role in accelerating autophagic degradation. However, RIP2 knockdown did not interfere with ER stress and the activation of autophagy induced by S. Infantis in Caco2 cells. Our data suggest that L. johnsonii L531 pretreatment alleviates the intestinal damage caused by S. Infantis by inhibiting NOD activation and regulating ER stress, as well as promoting autophagic degradation.
Collapse
|
18
|
Huang X, Yao J, Liu L, Luo Y, Yang A. Atg8-PE protein-based in vitro biochemical approaches to autophagy studies. Autophagy 2022; 18:2020-2035. [PMID: 35072587 PMCID: PMC9397461 DOI: 10.1080/15548627.2022.2025572] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved intracellular degradation pathway that maintains cellular homeostasis. Over the past two decades, a series of scientific breakthroughs have helped explain autophagy-related molecular mechanisms and physiological functions. This tremendous progress continues to depend largely on powerful research methods, specifically, various autophagy marker Atg8-PE protein-based methods for studying membrane dynamics and monitoring autophagic activity. Recently, several biochemical approaches have been successfully developed to produce the lipidated protein Atg8-PE or its mimics in vitro, including enzyme-mediated reconstitution systems, chemically defined reconstitution systems, cell-free lipidation systems and protein chemical synthesis. These approaches have contributed important insights into the mechanisms underlying Atg8-mediated membrane dynamics and protein-protein interactions, creating a new perspective in autophagy studies. In this review, we comprehensively summarize Atg8-PE protein-based in vitro biochemical approaches and recent advances to facilitate a better understanding of autophagy mechanisms. In addition, we highlight the advantages and disadvantages of various Atg8-PE protein-based approaches to provide general guidance for their use in studying autophagy.Abbreviations: ATG: autophagy related; ATP: adenosine triphosphate; COPII: coat protein complex II; DGS-NTA: 1,2-dioleoyl-sn-glycero-3-[(N-(5-amino-1-carboxypentyl)iminodiacetic acid)succinyl] (nickel salt); DPPE: 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine; DSPE: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine; E. coli: Escherichia coli; EPL: expressed protein ligation; ERGIC: ER-Golgi intermediate compartment; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor associated protein like 1; GABARAPL2: GABA type A receptor associated protein like 2; GFP: green fluorescent protein; GUVs: giant unilamellar vesicles; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MBP: maltose binding protein; MEFs: mouse embryonic fibroblasts; MESNa: 2-mercaptoethanesulfonic acid sodium salt; NCL: native chemical ligation; NTA: nitrilotriacetic acid; PE: phosphatidylethanolamine; PS: phosphatidylserine; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; SPPS: solid-phase peptide synthesis; TEV: tobacco etch virus; WT: wild-type.
Collapse
Affiliation(s)
- Xue Huang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jia Yao
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lu Liu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yu Luo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, China,CONTACT Aimin Yang School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
19
|
Chen J, Yao Y, Wang X, Wang Y, Li T, Du J. Chloroquine regulates the proliferation and apoptosis of palate development on mice embryo by activating P53 through blocking autophagy in vitro. In Vitro Cell Dev Biol Anim 2022; 58:558-570. [PMID: 35947289 DOI: 10.1007/s11626-022-00704-8] [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: 04/17/2022] [Accepted: 07/02/2022] [Indexed: 11/05/2022]
Abstract
Cleft lip and palate is one of the most frequent congenital developmental defects. Autophagy is a highly conserved process of cell self-degradation in eukaryotes, involving multiple biological processes in which chloroquine (CQ) is the most common inhibitor. However, whether CQ affects and how it affects palate development is unknown. Mouse embryonic palatal cells (MEPCs) were treated with CQ to observe cell viability, apoptosis, migration, osteogenic differentiation by cell proliferation assay, flow cytometric analysis, scratch assay, and alizarin red staining. PI staining was used to measure cell cycle distribution. Immunofluorescence (IF) assay and transmission electron microscopy were used to detect autophagosomes. The autophagy-related factors (LC3 and P62), apoptosis-related markers (P53, caspase-3 cleaved caspase-3, BAX, and BCL-2), and cell cycle-related proteins (P21, CDK2, CDK4, cyclin D1, and cyclin E) were all measured by western blot. CQ inhibited the proliferation of MEPCs by arresting the G0/G1 phase of the cell cycle in a concentration- and time-dependent manner with cell cycle-related proteins P21 upregulated and CDK2, CDK4, cyclin D1, and cyclin E downregulated. Then we detected CQ also induced cell apoptosis in a dose-dependent manner by decreasing the BCL-2/BAX ratio and increasing cleaved caspase-3. Next, it was investigated that migration and osteogenesis of MEPCs decreased with CQ treatment in a dose-dependent manner. Meanwhile, CQ blocked the autophagy pathway by upregulating LC3II and P62 expressions which activated the P53 pathway. CQ activates P53 which affects MEPC biological characteristics by changing the proliferation and apoptosis of MEPCs through inhibiting autophagy.
Collapse
Affiliation(s)
- Jing Chen
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yaxia Yao
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Xiaotong Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yijia Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Tianli Li
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Juan Du
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.
| |
Collapse
|
20
|
Lockwood DC, Amin H, Costa TRD, Schroeder GN. The Legionella pneumophila Dot/Icm type IV secretion system and its effectors. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35639581 DOI: 10.1099/mic.0.001187] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
To prevail in the interaction with eukaryotic hosts, many bacterial pathogens use protein secretion systems to release virulence factors at the host–pathogen interface and/or deliver them directly into host cells. An outstanding example of the complexity and sophistication of secretion systems and the diversity of their protein substrates, effectors, is the Defective in organelle trafficking/Intracellular multiplication (Dot/Icm) Type IVB secretion system (T4BSS) of
Legionella pneumophila
and related species.
Legionella
species are facultative intracellular pathogens of environmental protozoa and opportunistic human respiratory pathogens. The Dot/Icm T4BSS translocates an exceptionally large number of effectors, more than 300 per
L. pneumophila
strain, and is essential for evasion of phagolysosomal degradation and exploitation of protozoa and human macrophages as replicative niches. Recent technological advancements in the imaging of large protein complexes have provided new insight into the architecture of the T4BSS and allowed us to propose models for the transport mechanism. At the same time, significant progress has been made in assigning functions to about a third of
L. pneumophila
effectors, discovering unprecedented new enzymatic activities and concepts of host subversion. In this review, we describe the current knowledge of the workings of the Dot/Icm T4BSS machinery and provide an overview of the activities and functions of the to-date characterized effectors in the interaction of
L. pneumophila
with host cells.
Collapse
Affiliation(s)
- Daniel C Lockwood
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL, Northern Ireland, UK
| | - Himani Amin
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, SW7 2AZ, UK
| | - Tiago R D Costa
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, SW7 2AZ, UK
| | - Gunnar N Schroeder
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL, Northern Ireland, UK
| |
Collapse
|
21
|
Wei F, Yang A, Zhao Z, An H, Li Y, Duan Y. Mechanism of ER Stress-mediated ER-phagy by CdTe-QDs in Yeast Cells. Toxicol Lett 2022; 365:36-45. [DOI: 10.1016/j.toxlet.2022.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022]
|
22
|
Cao D, Wang Y, Li W, Ji J, Guo J, Zhang D, Liu J. 3,4‑Dihydroxyacetophenone attenuates oxidative stress‑induced damage to HUVECs via regulation of the Nrf2/HO‑1 pathway. Mol Med Rep 2022; 25:199. [PMID: 35475506 PMCID: PMC9073850 DOI: 10.3892/mmr.2022.12715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/04/2022] [Indexed: 11/08/2022] Open
Abstract
It has been reported that oxidative stress plays a prominent role in diabetic macrovascular diseases. 3,4-Dihydroxyacetophenone (3,4-DHAP) has been found to have a variety of biological activities. However, few studies have assessed the antioxidant capacity of 3,4-DHAP and the underlying mechanisms. Thus, the aim of the present study was to explore the effects of 3,4-DHAP on oxidative stress in human umbilical vein endothelial cells (HUVECs). HUVECs were pre-treated with 3,4-DHAP and then exposed to high glucose conditions. Cell viability and cytotoxicity were measured using an MTT assay. Reactive oxygen species (ROS) levels were measured using an inverted fluorescence microscope and a fluorescent enzyme labeling instrument. Protein expression levels of nuclear factor E2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), microtubule-associated protein 1A/1B-light chain 3 (LC3) and poly ADP-ribose polymerase-1 (PARP-1) were measured using western blotting, and mRNA expression of Nrf2 and HO-1 were measured through reverse transcription-quantitative PCR (RT-qPCR). Nrf2 nuclear translocation was evaluated using immunofluorescence analysis and autophagosomes were observed using transmission electron microscope (TEM). The results of the present study demonstrated that compared with the control group, cell viability of the high glucose group was reduced and cell cytotoxicity of the high glucose group was increased. ROS production in the high glucose group was clearly enhanced. In addition, high glucose upregulated Nrf2 and HO-1 protein and mRNA expression levels. Nuclear translocation of Nrf2 in the high glucose group was also increased. The formation of autophagosomes in the high glucose group was also higher than that in the control group. Furthermore, LC3-II/LC3-I and PARP-1 protein expression levels were increased after treatment with high glucose. However, compared to the high glucose group, 3,4-DHAP (10 µmol/l) significantly enhanced cell viability. 3,4-DHAP markedly decreased the production of ROS, increased Nrf2 and HO-1 protein and mRNA expression levels, and promoted nuclear translocation of Nrf2 in HUVECs. In addition, 3,4-DHAP promoted the formation of autophagosomes, and notably increased the protein expression levels of LC3-II/LC3-I and PARP-1. Moreover, it was determined that compared to the 3,4-DHAP group, treatment with 3,4-DHAP and ML385 enhanced cell viability, and decreased ROS production, Nrf2 and HO-1 protein and mRNA expression levels, nuclear translocation of Nrf2, and LC3-II/LC3-I and PARP-1 protein expression levels. Collectively, the results of the present study showed that 3,4-DHAP protected HUVECs against oxidative stress via regulation of the Nrf2/HO-1 pathway, by increasing autophagy and promoting DNA damage repair.
Collapse
Affiliation(s)
- Daihong Cao
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yunhan Wang
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Wentao Li
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiafen Ji
- Department of Pediatrics, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Juntang Guo
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Daijuan Zhang
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiangyue Liu
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
23
|
Zhenzhu Xiaoji Decoction Induces Autophagy and Apoptosis Cell Death in Liver Cancer Cells through AKT/mTOR and JAK2/STAT3 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4445293. [PMID: 35432564 PMCID: PMC9007645 DOI: 10.1155/2022/4445293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/20/2022] [Accepted: 03/08/2022] [Indexed: 12/24/2022]
Abstract
Background Liver cancer is one of the most common digestive tumors. The prescription Zhenzhu Xiaoji decoction (ZZXJD) has a certain effect on the growth and survival of primary liver cancer. Object: This article aimed to explore the effect and molecular mechanism of ZZXJD on liver cancer SMMC-7721 cells. Method The research groups were divided into the model group, ZZXJD group, and cisplatin group. SMMC-7721 cells were treated with different concentrations of ZZXJD-medicated serum for 24 h and 48 h. The cell viability was measured with CCK8 assay, and cell morphology was observed by fluorescence microscope and transmission electron microscope (TEM). Western blot, RT-PCR, and gene chip were used to determine the protein expression level and gene expression level of cells and tumor tissues. Results ZZXJD inhibited the proliferation activity of SMMC-7721 cells in a concentration- and time-dependent manner. The morphological changes of the cell showed apoptosis and autophagy. The gene expression of protein kinase B (AKT), mammalian target of rapamycin (mTOR), Janus kinase 2 (JAK2), and signal transducer and activator of transcription 3(STAT3) were downregulated compared with the model group(p < 0.05). The nude mice experiments confirmed that ZZXJD inhibited the growth of tumors in tumor-bearing mice, and the effect increased with the increase of concentration. Conclusion ZZXJD induced autophagy and apoptosis of liver cancer cells via inhibiting AKT/mTOR signaling pathway and JAK2/STAT3 signaling pathway, thereby affecting the growth and survival of liver cancer cells.
Collapse
|
24
|
Sims C, Withall DM, Oldham N, Stockman R, Birkett M. Computational investigation of aphid odorant receptor structure and binding function. J Biomol Struct Dyn 2022; 41:3647-3658. [PMID: 35352606 DOI: 10.1080/07391102.2022.2053743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Odorant receptors (OR) play a critical role in signal transduction and olfactory recognition in insects. Unfortunately, insect ORs are difficult to express and purify, and limited structural data are available. Computational methods were used to predict models for aphid ORs, and binding interactions with aphid pheromones and other semiochemicals were investigated. Previously functionally characterised ORs from the pea aphid, Acyrthosiphon pisum, ApisOR4 and ApisOR5, were screened against functional ligands. ApisOR5 had a defined binding site, and had predicted interactions with the aphid alarm pheromone, (E)-β-farnesene. ApisOR4 had multiple distinct binding sites and showed broad tuning to multiple odorants. Screening of six other highly conserved ORs showed some interactions and potential enantiomeric discrimination between the aphid sex pheromone components (4aS,7S,7aR)-nepetalactone and (1R,4aS,7S,7aR)-nepetalactol. These results indicate that specific binding sites may be more critical to understanding olfactory activity of ligands and ORs than kinetic data, and greater knowledge of the method of action of ORs is required.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Cassie Sims
- Biointeractions and Crop Protection Department, Rothamsted Research, Harpenden, Hertfordshire, UK.,School of Chemistry, University of Nottingham, Nottingham, UK
| | - David M Withall
- Biointeractions and Crop Protection Department, Rothamsted Research, Harpenden, Hertfordshire, UK
| | - Neil Oldham
- School of Chemistry, University of Nottingham, Nottingham, UK
| | - Robert Stockman
- School of Chemistry, University of Nottingham, Nottingham, UK
| | - Michael Birkett
- Biointeractions and Crop Protection Department, Rothamsted Research, Harpenden, Hertfordshire, UK
| |
Collapse
|
25
|
Wei F, Xie Q, Huang Z, Yang A, Duan Y. Induction of autophagy and ER-phagy caused by CdTe-QDs are protective mechanisms of yeast cell. J Appl Toxicol 2022; 42:1146-1158. [PMID: 34989008 DOI: 10.1002/jat.4282] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 11/11/2022]
Abstract
Quantum dots (QDs), with unique and tunable optical properties, have been are widely used in many fields closely related to our daily lives, such as biomedical application and electronic products. Therefore, the potential toxicity of QDs on the human health should be understood. Autophagy plays an important role in cell survival and death. Endoplasmic reticulum autophagy (ER-phagy), a selective autophagy that degrades ER, responds to the accumulation of misfolded proteins and ER stress. Although many reports have revealed that autophagy can be disturbed by CdTe-QDs and other nanomaterials, there are still lack more detail researches to illustrate the function of autophagy in CdTe-QDs treated cells. And the function of ER-phagy in CdTe-QDs-treated cells remains to be illustrated. On the basis of transcriptome analysis, we explored the effect of CdTe-QDs on Saccharomyces cerevisiae, and firstly illustrated that both of autophagy and ER-phagy were protective mechanisms in CdTe-QDs-treated cells. It was found that CdTe-QDs inhibited the proliferation of yeast cells, disrupted homeostasis of cells, membrane integrity and metabolism process. All of these can be reasons of the reduction of cell viability. The abolish of autophagy and ER-phagy reduce the cell survival, indicating both of them are cell protective mechanisms against CdTe-QDs toxicity in yeast cells. Therefore, our data are significant for the application of CdTe-QDs and provide precious information for understanding of nanomaterials-related ER-phagy.
Collapse
Affiliation(s)
- Fujing Wei
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, P. R. China.,School of Life Sciences, Chongqing University, Chongqing, China
| | - Qiyue Xie
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, P. R. China
| | - Zhijun Huang
- Research Center of Analytical Instrumentation, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, Shanxi, PR China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
26
|
Shen X, Tang Z, Bai Y, Wan M, Yu M, Chen J, Li G, Zhang R, Ge M. Astragalus Polysaccharide Protects Against Cadmium-Induced Autophagy Injury Through Reactive Oxygen Species (ROS) Pathway in Chicken Embryo Fibroblast. Biol Trace Elem Res 2022; 200:318-329. [PMID: 33704669 DOI: 10.1007/s12011-021-02628-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/04/2021] [Indexed: 01/05/2023]
Abstract
Cadmium (Cd) is a harmful heavy metal pollutant, which can cause oxidative stress in the body and induce cell damage. Reactive oxygen species (ROS) is a general term for substances that contain oxygen and are active in the body. However, excessive ROS can damage the body. Cadmium poisoning can cause a large amount of ROS in cells and autophagy. Astragalus polysaccharide (APS) is a plant polysaccharide with biological functions, such as antioxidant and anti-stress activities. In this study, chicken embryo fibroblasts (CEF) were used to determine the relationship between ROS and autophagy damage of Cd-infected cells and the mechanism of APS on cadmium-induced autophagy damage. The results showed that a 10-μL dose of 10 μmol/L cadmium chloride (CdCl2) can induce CEF autophagy and damage when CEF was added for 36 h. Cadmium induced CEF autophagy damage by increasing ROS production. APS could significantly reduce ROS production and LC3-II and Beclin-1 protein expression, increase the expression of mTOR and the level of antioxidation, and restore the viability and morphological damage of CEF exposed to Cd. Our study suggests that APS can alleviate Cd-induced CEF autophagy damage by reducing the production of ROS.
Collapse
Affiliation(s)
- Xudong Shen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Zequn Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Yu Bai
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Meishuo Wan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Miao Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Jingyi Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Guangxing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Ruili Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China.
| | - Ming Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang, for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| |
Collapse
|
27
|
Direct Interaction of ATP7B and LC3B Proteins Suggests a Cooperative Role of Copper Transportation and Autophagy. Cells 2021; 10:cells10113118. [PMID: 34831341 PMCID: PMC8625360 DOI: 10.3390/cells10113118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 11/17/2022] Open
Abstract
Macroautophagy/autophagy plays an important role in cellular copper clearance. The means by which the copper metabolism and autophagy pathways interact mechanistically is vastly unexplored. Dysfunctional ATP7B, a copper-transporting ATPase, is involved in the development of monogenic Wilson disease, a disorder characterized by disturbed copper transport. Using in silico prediction, we found that ATP7B contains a number of potential binding sites for LC3, a central protein in the autophagy pathway, the so-called LC3 interaction regions (LIRs). The conserved LIR3, located at the C-terminal end of ATP7B, was found to directly interact with LC3B in vitro. Replacing the two conserved hydrophobic residues W1452 and L1455 of LIR3 significantly reduced interaction. Furthermore, autophagy was induced in normal human hepatocellular carcinoma cells (HepG2) leading to enhanced colocalization of ATP7B and LC3B on the autophagosome membranes. By contrast, HepG2 cells deficient of ATP7B (HepG2 ATP7B-/-) showed autophagy deficiency at elevated copper condition. This phenotype was complemented by heterologous ATP7B expression. These findings suggest a cooperative role of ATP7B and LC3B in autophagy-mediated copper clearance.
Collapse
|
28
|
Viana F, Peringathara SS, Rizvi A, Schroeder GN. Host manipulation by bacterial type III and type IV secretion system effector proteases. Cell Microbiol 2021; 23:e13384. [PMID: 34392594 PMCID: PMC11475232 DOI: 10.1111/cmi.13384] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023]
Abstract
Proteases are powerful enzymes, which cleave peptide bonds, leading most of the time to irreversible fragmentation or degradation of their substrates. Therefore they control many critical cell fate decisions in eukaryotes. Bacterial pathogens exploit this power and deliver protease effectors through specialised secretion systems into host cells. Research over the past years revealed that the functions of protease effectors during infection are diverse, reflecting the lifestyles and adaptations to specific hosts; however, only a small number of peptidase families seem to have given rise to most of these protease virulence factors by the evolution of different substrate-binding specificities, intracellular activation and subcellular targeting mechanisms. Here, we review our current knowledge about the enzymology and function of protease effectors, which Gram-negative bacterial pathogens translocate via type III and IV secretion systems to irreversibly manipulate host processes. We highlight emerging concepts such as signalling by protease cleavage products and effector-triggered immunity, which host cells employ to detect and defend themselves against a protease attack. TAKE AWAY: Proteases irreversibly cleave proteins to control critical cell fate decisions. Gram-negative bacteria use type III and IV secretion systems to inject effectors. Protease effectors are integral weapons for the manipulation of host processes. Effectors evolved from few peptidase families to target diverse substrates. Effector-triggered immunity upon proteolytic attack emerges as host defence.
Collapse
Affiliation(s)
- Flávia Viana
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesQueen's University BelfastBelfast, Northern IrelandUK
| | - Shruthi Sachidanandan Peringathara
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesQueen's University BelfastBelfast, Northern IrelandUK
| | - Arshad Rizvi
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesQueen's University BelfastBelfast, Northern IrelandUK
| | - Gunnar N. Schroeder
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesQueen's University BelfastBelfast, Northern IrelandUK
| |
Collapse
|
29
|
Tóth D, Horváth GV, Juhász G. The interplay between pathogens and Atg8 family proteins: thousand-faced interactions. FEBS Open Bio 2021; 11:3237-3252. [PMID: 34670023 PMCID: PMC8634866 DOI: 10.1002/2211-5463.13318] [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: 06/29/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 01/18/2023] Open
Abstract
Autophagy is an intracellular degradation and recycling process that can also remove pathogenic intracellular bacteria and viruses from within cells (referred to as xenophagy) and activate the adaptive immune responses. But autophagy-especially Atg proteins including Atg8 family members-can also have proviral and probacterial effects. In this review, we summarize known interactions of bacterial, parasitic, and viral proteins with Atg8 family proteins and the outcome of these interactions on pathogen replication, autophagy, or mitophagy. We discuss the value of prediction software and the research methodology in the study of pathogen protein-Atg8 family protein interactions, with selected examples of potential LC3-interacting region motif-containing SARS-CoV-2 proteins.
Collapse
Affiliation(s)
- Dávid Tóth
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - Gábor V Horváth
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - Gábor Juhász
- Institute of Genetics, Biological Research Centre, Szeged, Hungary.,Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
30
|
Park JH, Lee SH, Park SW, Jun YW, Kim K, Jeon P, Kim M, Lee JA, Jang DJ. Deciphering the role of a membrane-targeting domain in assisting endosomal and autophagic membrane localization of a RavZ protein catalytic domain. BMB Rep 2021. [PMID: 33298241 PMCID: PMC7907744 DOI: 10.5483/bmbrep.2021.54.2.190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The bacterial effector protein RavZ from a pathogen can impair autophagy in the host by delipidating the mammalian autophagy- related gene 8 (mATG8)-phosphatidylethanolamine (PE) on autophagic membranes. In RavZ, the membrane-targeting (MT) domain is an essential function. However, the molecular mechanism of this domain in regulating the intracellular localization of RavZ in cells is unclear. In this study, we found that the fusion of the green fluorescent protein (GFP) to the MT domain of RavZ (GFP-MT) resulted in localization primarily to the cytosol and nucleus, whereas the GFP-fused duplicated-MT domain (GFP-2xMT) localized to Rab5- or Rab7-positive endosomes. Similarly, GFP fusion to the catalytic domain (CA) of RavZ (GFP-CA) resulted in localization primarily to the cytosol and nucleus, even in autophagy-induced cells. However, by adding the MT domain to GFP-CA (GFP-CA-MT), the cooperation of MT and CA led to localization on the Rab5-positive endosomal membranes in a wortmannin-sensitive manner under nutrient-rich conditions, and to autophagic membranes in autophagy-induced cells. In autophagic membranes, GFP-CA-MT delipidated overexpressed or endogenous mATG8-PE. Furthermore, GFP-CAΔα3-MT, an α3 helix deletion within the CA domain, failed to localize to the endosomal or autophagic membranes and could not delipidate overexpressed mATG8-PE. Thus, the CA or MT domain alone is insufficient for stable membrane localization in cells, but the cooperation of MT and CA leads to localization to the endosomal and autophagic membranes. In autophagic membranes, the CA domain can delipidate mATG8-PE without requiring substrate recognition mediated by LC3-interacting region (LIR) motifs. [BMB Reports 2021; 54(2): 118-123].
Collapse
Affiliation(s)
- Jui-Hee Park
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Seung-Hwan Lee
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Sang-Won Park
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Yong-Woo Jun
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Kunhyung Kim
- Laboratory of Neurobiochemistry, Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, Daegu 41062, Korea
| | - Pureum Jeon
- Department of Biological Science and Biotechnology, College of Life Science and Nano Technology, Hannam University, Daejeon 34054, Korea
| | - Myungjin Kim
- Laboratory of Neurobiochemistry, Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, Daegu 41062, Korea
| | - Jin-A Lee
- Department of Biological Science and Biotechnology, College of Life Science and Nano Technology, Hannam University, Daejeon 34054, Korea
| | - Deok-Jin Jang
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| |
Collapse
|
31
|
Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, Bravo‐San Pedro JM, Cadwell K, Cecconi F, Choi AMK, Choi ME, Chu CT, Codogno P, Colombo M, Cuervo AM, Deretic V, Dikic I, Elazar Z, Eskelinen E, Fimia GM, Gewirtz DA, Green DR, Hansen M, Jäättelä M, Johansen T, Juhász G, Karantza V, Kraft C, Kroemer G, Ktistakis NT, Kumar S, Lopez‐Otin C, Macleod KF, Madeo F, Martinez J, Meléndez A, Mizushima N, Münz C, Penninger JM, Perera R, Piacentini M, Reggiori F, Rubinsztein DC, Ryan K, Sadoshima J, Santambrogio L, Scorrano L, Simon H, Simon AK, Simonsen A, Stolz A, Tavernarakis N, Tooze SA, Yoshimori T, Yuan J, Yue Z, Zhong Q, Galluzzi L, Pietrocola F. Autophagy in major human diseases. EMBO J 2021; 40:e108863. [PMID: 34459017 PMCID: PMC8488577 DOI: 10.15252/embj.2021108863] [Citation(s) in RCA: 943] [Impact Index Per Article: 235.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a core molecular pathway for the preservation of cellular and organismal homeostasis. Pharmacological and genetic interventions impairing autophagy responses promote or aggravate disease in a plethora of experimental models. Consistently, mutations in autophagy-related processes cause severe human pathologies. Here, we review and discuss preclinical data linking autophagy dysfunction to the pathogenesis of major human disorders including cancer as well as cardiovascular, neurodegenerative, metabolic, pulmonary, renal, infectious, musculoskeletal, and ocular disorders.
Collapse
Affiliation(s)
| | - Giulia Petroni
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
| | - Ravi K Amaravadi
- Department of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Abramson Cancer CenterUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Andrea Ballabio
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesSection of PediatricsFederico II UniversityNaplesItaly
- Department of Molecular and Human GeneticsBaylor College of Medicine, and Jan and Dan Duncan Neurological Research InstituteTexas Children HospitalHoustonTXUSA
| | - Patricia Boya
- Margarita Salas Center for Biological ResearchSpanish National Research CouncilMadridSpain
| | - José Manuel Bravo‐San Pedro
- Faculty of MedicineDepartment Section of PhysiologyComplutense University of MadridMadridSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball InstituteNew York University Grossman School of MedicineNew YorkNYUSA
- Department of MicrobiologyNew York University Grossman School of MedicineNew YorkNYUSA
- Division of Gastroenterology and HepatologyDepartment of MedicineNew York University Langone HealthNew YorkNYUSA
| | - Francesco Cecconi
- Cell Stress and Survival UnitCenter for Autophagy, Recycling and Disease (CARD)Danish Cancer Society Research CenterCopenhagenDenmark
- Department of Pediatric Onco‐Hematology and Cell and Gene TherapyIRCCS Bambino Gesù Children's HospitalRomeItaly
- Department of BiologyUniversity of Rome ‘Tor Vergata’RomeItaly
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care MedicineJoan and Sanford I. Weill Department of MedicineWeill Cornell MedicineNew YorkNYUSA
- New York‐Presbyterian HospitalWeill Cornell MedicineNew YorkNYUSA
| | - Mary E Choi
- New York‐Presbyterian HospitalWeill Cornell MedicineNew YorkNYUSA
- Division of Nephrology and HypertensionJoan and Sanford I. Weill Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Charleen T Chu
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Patrice Codogno
- Institut Necker‐Enfants MaladesINSERM U1151‐CNRS UMR 8253ParisFrance
- Université de ParisParisFrance
| | - Maria Isabel Colombo
- Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia‐Instituto de Histología y Embriología (IHEM)‐Universidad Nacional de CuyoCONICET‐ Facultad de Ciencias MédicasMendozaArgentina
| | - Ana Maria Cuervo
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineBronxNYUSA
| | - Vojo Deretic
- Autophagy Inflammation and Metabolism (AIMCenter of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Ivan Dikic
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt, Frankfurt am MainGermany
- Buchmann Institute for Molecular Life SciencesGoethe UniversityFrankfurt, Frankfurt am MainGermany
| | - Zvulun Elazar
- Department of Biomolecular SciencesThe Weizmann Institute of ScienceRehovotIsrael
| | | | - Gian Maria Fimia
- Department of Molecular MedicineSapienza University of RomeRomeItaly
- Department of EpidemiologyPreclinical Research, and Advanced DiagnosticsNational Institute for Infectious Diseases ‘L. Spallanzani’ IRCCSRomeItaly
| | - David A Gewirtz
- Department of Pharmacology and ToxicologySchool of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Douglas R Green
- Department of ImmunologySt. Jude Children's Research HospitalMemphisTNUSA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery InstituteProgram of DevelopmentAging, and RegenerationLa JollaCAUSA
| | - Marja Jäättelä
- Cell Death and MetabolismCenter for Autophagy, Recycling & DiseaseDanish Cancer Society Research CenterCopenhagenDenmark
- Department of Cellular and Molecular MedicineFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Terje Johansen
- Department of Medical BiologyMolecular Cancer Research GroupUniversity of Tromsø—The Arctic University of NorwayTromsøNorway
| | - Gábor Juhász
- Institute of GeneticsBiological Research CenterSzegedHungary
- Department of Anatomy, Cell and Developmental BiologyEötvös Loránd UniversityBudapestHungary
| | | | - Claudine Kraft
- Institute of Biochemistry and Molecular BiologyZBMZFaculty of MedicineUniversity of FreiburgFreiburgGermany
- CIBSS ‐ Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
| | - Guido Kroemer
- Centre de Recherche des CordeliersEquipe Labellisée par la Ligue Contre le CancerUniversité de ParisSorbonne UniversitéInserm U1138Institut Universitaire de FranceParisFrance
- Metabolomics and Cell Biology PlatformsInstitut Gustave RoussyVillejuifFrance
- Pôle de BiologieHôpital Européen Georges PompidouAP‐HPParisFrance
- Suzhou Institute for Systems MedicineChinese Academy of Medical SciencesSuzhouChina
- Karolinska InstituteDepartment of Women's and Children's HealthKarolinska University HospitalStockholmSweden
| | | | - Sharad Kumar
- Centre for Cancer BiologyUniversity of South AustraliaAdelaideSAAustralia
- Faculty of Health and Medical SciencesUniversity of AdelaideAdelaideSAAustralia
| | - Carlos Lopez‐Otin
- Departamento de Bioquímica y Biología MolecularFacultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA)Universidad de OviedoOviedoSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Kay F Macleod
- The Ben May Department for Cancer ResearchThe Gordon Center for Integrative SciencesW‐338The University of ChicagoChicagoILUSA
- The University of ChicagoChicagoILUSA
| | - Frank Madeo
- Institute of Molecular BiosciencesNAWI GrazUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
- Field of Excellence BioHealth – University of GrazGrazAustria
| | - Jennifer Martinez
- Immunity, Inflammation and Disease LaboratoryNational Institute of Environmental Health SciencesNIHResearch Triangle ParkNCUSA
| | - Alicia Meléndez
- Biology Department, Queens CollegeCity University of New YorkFlushingNYUSA
- The Graduate Center Biology and Biochemistry PhD Programs of the City University of New YorkNew YorkNYUSA
| | - Noboru Mizushima
- Department of Biochemistry and Molecular BiologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Christian Münz
- Viral ImmunobiologyInstitute of Experimental ImmunologyUniversity of ZurichZurichSwitzerland
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA)Vienna BioCenter (VBC)ViennaAustria
- Department of Medical GeneticsLife Sciences InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Rushika M Perera
- Department of AnatomyUniversity of California, San FranciscoSan FranciscoCAUSA
- Department of PathologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Mauro Piacentini
- Department of BiologyUniversity of Rome “Tor Vergata”RomeItaly
- Laboratory of Molecular MedicineInstitute of Cytology Russian Academy of ScienceSaint PetersburgRussia
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells & SystemsMolecular Cell Biology SectionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - David C Rubinsztein
- Department of Medical GeneticsCambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeUK
| | - Kevin M Ryan
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular MedicineCardiovascular Research InstituteRutgers New Jersey Medical SchoolNewarkNJUSA
| | - Laura Santambrogio
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
- Sandra and Edward Meyer Cancer CenterNew YorkNYUSA
- Caryl and Israel Englander Institute for Precision MedicineNew YorkNYUSA
| | - Luca Scorrano
- Istituto Veneto di Medicina MolecolarePadovaItaly
- Department of BiologyUniversity of PadovaPadovaItaly
| | - Hans‐Uwe Simon
- Institute of PharmacologyUniversity of BernBernSwitzerland
- Department of Clinical Immunology and AllergologySechenov UniversityMoscowRussia
- Laboratory of Molecular ImmunologyInstitute of Fundamental Medicine and BiologyKazan Federal UniversityKazanRussia
| | | | - Anne Simonsen
- Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Centre for Cancer Cell ReprogrammingInstitute of Clinical MedicineUniversity of OsloOsloNorway
- Department of Molecular Cell BiologyInstitute for Cancer ResearchOslo University Hospital MontebelloOsloNorway
| | - Alexandra Stolz
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt, Frankfurt am MainGermany
- Buchmann Institute for Molecular Life SciencesGoethe UniversityFrankfurt, Frankfurt am MainGermany
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and BiotechnologyFoundation for Research and Technology‐HellasHeraklion, CreteGreece
- Department of Basic SciencesSchool of MedicineUniversity of CreteHeraklion, CreteGreece
| | - Sharon A Tooze
- Molecular Cell Biology of AutophagyThe Francis Crick InstituteLondonUK
| | - Tamotsu Yoshimori
- Department of GeneticsGraduate School of MedicineOsaka UniversitySuitaJapan
- Department of Intracellular Membrane DynamicsGraduate School of Frontier BiosciencesOsaka UniversitySuitaJapan
- Integrated Frontier Research for Medical Science DivisionInstitute for Open and Transdisciplinary Research Initiatives (OTRI)Osaka UniversitySuitaJapan
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
- Department of Cell BiologyHarvard Medical SchoolBostonMAUSA
| | - Zhenyu Yue
- Department of NeurologyFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationDepartment of PathophysiologyShanghai Jiao Tong University School of Medicine (SJTU‐SM)ShanghaiChina
| | - Lorenzo Galluzzi
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
- Sandra and Edward Meyer Cancer CenterNew YorkNYUSA
- Caryl and Israel Englander Institute for Precision MedicineNew YorkNYUSA
- Department of DermatologyYale School of MedicineNew HavenCTUSA
- Université de ParisParisFrance
| | | |
Collapse
|
32
|
Yu M, Li R, Wan M, Chen J, Shen X, Li G, Ge M, Zhang R. MDA5 attenuate autophagy in chicken embryo fibroblasts infected with IBDV. Br Poult Sci 2021; 63:154-163. [PMID: 34406094 DOI: 10.1080/00071668.2021.1969643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
1. The role of melanoma differentiation-associated protein 5 (MDA5) in infectious bursal disease virus (IBDV)-induced autophagy was studied in chicken embryos.2. Chicken embryo fibroblasts (CEF) were used as the research model and small interfering RNA (siRNA), western blot, indirect enzyme-linked immunosorbent assay (ELISA), real-time fluorescence quantitative polymerase chain reaction (PCR) and transmission electron microscopy were used to detect autophagy, IBDV replication, CEF damage, and activation of both MDA5 and its signalling pathway.3. The results showed that CEF infected with IBDV activated the intracellular MDA5 signalling pathway and caused autophagy via inactivation of the AKT/mTOR pathway. While autophagy promotes IBDV proliferation, MDA5 weakens IBDV-induced CEF autophagy thus inhibiting IBDV replication and protecting CEF cells.4. The results indicated that chMDA5 can be activated by IBDV and attenuate CEF autophagy caused by IBDV infection, thereby inhibiting IBDV replication. This study provided a foundation for further exploring the relationship between viruses, autophagy and the pathogenic mechanism of the MDA5 pathway involved in IBDV.
Collapse
Affiliation(s)
- M Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| | - R Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| | - M Wan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| | - J Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| | - X Shen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| | - G Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| | - M Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| | - R Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, People's Republic of China
| |
Collapse
|
33
|
Schneidewind T, Brause A, Schölermann B, Sievers S, Pahl A, Sankar MG, Winzker M, Janning P, Kumar K, Ziegler S, Waldmann H. Combined morphological and proteome profiling reveals target-independent impairment of cholesterol homeostasis. Cell Chem Biol 2021; 28:1780-1794.e5. [PMID: 34214450 DOI: 10.1016/j.chembiol.2021.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/11/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022]
Abstract
Unbiased profiling approaches are powerful tools for small-molecule target or mode-of-action deconvolution as they generate a holistic view of the bioactivity space. This is particularly important for non-protein targets that are difficult to identify with commonly applied target identification methods. Thereby, unbiased profiling can enable identification of novel bioactivity even for annotated compounds. We report the identification of a large bioactivity cluster comprised of numerous well-characterized drugs with different primary targets using a combination of the morphological Cell Painting Assay and proteome profiling. Cluster members alter cholesterol homeostasis and localization due to their physicochemical properties that lead to protonation and accumulation in lysosomes, an increase in lysosomal pH, and a disturbed cholesterol homeostasis. The identified cluster enables identification of modulators of cholesterol homeostasis and links regulation of genes or proteins involved in cholesterol synthesis or trafficking to physicochemical properties rather than to nominal targets.
Collapse
Affiliation(s)
- Tabea Schneidewind
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany; Technical University Dortmund, Faculty of Chemistry and Chemical Biology, Otto-Hahn-Strasse 6, Dortmund 44227, Germany
| | - Alexandra Brause
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Beate Schölermann
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Sonja Sievers
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Axel Pahl
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Muthukumar G Sankar
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Michael Winzker
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Petra Janning
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Kamal Kumar
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Slava Ziegler
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Herbert Waldmann
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany; Technical University Dortmund, Faculty of Chemistry and Chemical Biology, Otto-Hahn-Strasse 6, Dortmund 44227, Germany.
| |
Collapse
|
34
|
Wirth M, Mouilleron S, Zhang W, Sjøttem E, Princely Abudu Y, Jain A, Lauritz Olsvik H, Bruun JA, Razi M, Jefferies HB, Lee R, Joshi D, O'Reilly N, Johansen T, Tooze SA. Phosphorylation of the LIR Domain of SCOC Modulates ATG8 Binding Affinity and Specificity. J Mol Biol 2021; 433:166987. [DOI: https:/doi.org/10.1016/j.jmb.2021.166987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
|
35
|
Durgan J, Lystad AH, Sloan K, Carlsson SR, Wilson MI, Marcassa E, Ulferts R, Webster J, Lopez-Clavijo AF, Wakelam MJ, Beale R, Simonsen A, Oxley D, Florey O. Non-canonical autophagy drives alternative ATG8 conjugation to phosphatidylserine. Mol Cell 2021; 81:2031-2040.e8. [PMID: 33909989 PMCID: PMC8122138 DOI: 10.1016/j.molcel.2021.03.020] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 01/15/2021] [Accepted: 03/16/2021] [Indexed: 01/22/2023]
Abstract
Autophagy is a fundamental catabolic process that uses a unique post-translational modification, the conjugation of ATG8 protein to phosphatidylethanolamine (PE). ATG8 lipidation also occurs during non-canonical autophagy, a parallel pathway involving conjugation of ATG8 to single membranes (CASM) at endolysosomal compartments, with key functions in immunity, vision, and neurobiology. It is widely assumed that CASM involves the same conjugation of ATG8 to PE, but this has not been formally tested. Here, we discover that all ATG8s can also undergo alternative lipidation to phosphatidylserine (PS) during CASM, induced pharmacologically, by LC3-associated phagocytosis or influenza A virus infection, in mammalian cells. Importantly, ATG8-PS and ATG8-PE adducts are differentially delipidated by the ATG4 family and bear different cellular dynamics, indicating significant molecular distinctions. These results provide important insights into autophagy signaling, revealing an alternative form of the hallmark ATG8 lipidation event. Furthermore, ATG8-PS provides a specific "molecular signature" for the non-canonical autophagy pathway.
Collapse
Affiliation(s)
- Joanne Durgan
- Signalling Programme, Babraham Institute, Cambridge, UK
| | - Alf H Lystad
- Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | | | - Sven R Carlsson
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | | | | | | | - Judith Webster
- Mass Spectrometry Facility, Babraham Institute, Cambridge, UK
| | | | - Michael J Wakelam
- Signalling Programme, Babraham Institute, Cambridge, UK; Lipidomics Facility, Babraham Institute, Cambridge, UK
| | | | - Anne Simonsen
- Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - David Oxley
- Mass Spectrometry Facility, Babraham Institute, Cambridge, UK
| | - Oliver Florey
- Signalling Programme, Babraham Institute, Cambridge, UK.
| |
Collapse
|
36
|
Phosphorylation of the LIR Domain of SCOC Modulates ATG8 Binding Affinity and Specificity. J Mol Biol 2021; 433:166987. [PMID: 33845085 PMCID: PMC8202330 DOI: 10.1016/j.jmb.2021.166987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/27/2021] [Accepted: 04/04/2021] [Indexed: 12/15/2022]
Abstract
Autophagy is a highly conserved degradative pathway, essential for cellular homeostasis and implicated in diseases including cancer and neurodegeneration. Autophagy-related 8 (ATG8) proteins play a central role in autophagosome formation and selective delivery of cytoplasmic cargo to lysosomes by recruiting autophagy adaptors and receptors. The LC3-interacting region (LIR) docking site (LDS) of ATG8 proteins binds to LIR motifs present in autophagy adaptors and receptors. LIR-ATG8 interactions can be highly selective for specific mammalian ATG8 family members (LC3A-C, GABARAP, and GABARAPL1-2) and how this specificity is generated and regulated is incompletely understood. We have identified a LIR motif in the Golgi protein SCOC (short coiled-coil protein) exhibiting strong binding to GABARAP, GABARAPL1, LC3A and LC3C. The residues within and surrounding the core LIR motif of the SCOC LIR domain were phosphorylated by autophagy-related kinases (ULK1-3, TBK1) increasing specifically LC3 family binding. More distant flanking residues also contributed to ATG8 binding. Loss of these residues was compensated by phosphorylation of serine residues immediately adjacent to the core LIR motif, indicating that the interactions of the flanking LIR regions with the LDS are important and highly dynamic. Our comprehensive structural, biophysical and biochemical analyses support and provide novel mechanistic insights into how phosphorylation of LIR domain residues regulates the affinity and binding specificity of ATG8 proteins towards autophagy adaptors and receptors.
Collapse
|
37
|
Vozandychova V, Stojkova P, Hercik K, Rehulka P, Stulik J. The Ubiquitination System within Bacterial Host-Pathogen Interactions. Microorganisms 2021; 9:638. [PMID: 33808578 PMCID: PMC8003559 DOI: 10.3390/microorganisms9030638] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022] Open
Abstract
Ubiquitination of proteins, like phosphorylation and acetylation, is an important regulatory aspect influencing numerous and various cell processes, such as immune response signaling and autophagy. The study of ubiquitination has become essential to learning about host-pathogen interactions, and a better understanding of the detailed mechanisms through which pathogens affect ubiquitination processes in host cell will contribute to vaccine development and effective treatment of diseases. Pathogenic bacteria (e.g., Salmonella enterica, Legionella pneumophila and Shigella flexneri) encode many effector proteins, such as deubiquitinating enzymes (DUBs), targeting the host ubiquitin machinery and thus disrupting pertinent ubiquitin-dependent anti-bacterial response. We focus here upon the host ubiquitination system as an integral unit, its interconnection with the regulation of inflammation and autophagy, and primarily while examining pathogens manipulating the host ubiquitination system. Many bacterial effector proteins have already been described as being translocated into the host cell, where they directly regulate host defense processes. Due to their importance in pathogenic bacteria progression within the host, they are regarded as virulence factors essential for bacterial evasion. However, in some cases (e.g., Francisella tularensis) the host ubiquitination system is influenced by bacterial infection, although the responsible bacterial effectors are still unknown.
Collapse
Affiliation(s)
- Vera Vozandychova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| | - Pavla Stojkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| | - Kamil Hercik
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000 Prague, Czech Republic
| | - Pavel Rehulka
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| | - Jiri Stulik
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| |
Collapse
|
38
|
Grishin A, Voth K, Gagarinova A, Cygler M. Structural biology of the invasion arsenal of Gram-negative bacterial pathogens. FEBS J 2021; 289:1385-1427. [PMID: 33650300 DOI: 10.1111/febs.15794] [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: 11/08/2020] [Revised: 02/11/2021] [Accepted: 02/26/2021] [Indexed: 12/20/2022]
Abstract
In the last several years, there has been a tremendous progress in the understanding of host-pathogen interactions and the mechanisms by which bacterial pathogens modulate behavior of the host cell. Pathogens use secretion systems to inject a set of proteins, called effectors, into the cytosol of the host cell. These effectors are secreted in a highly regulated, temporal manner and interact with host proteins to modify a multitude of cellular processes. The number of effectors varies between pathogens from ~ 30 to as many as ~ 350. The functional redundancy of effectors encoded by each pathogen makes it difficult to determine the cellular effects or function of individual effectors, since their individual knockouts frequently produce no easily detectable phenotypes. Structural biology of effector proteins and their interactions with host proteins, in conjunction with cell biology approaches, has provided invaluable information about the cellular function of effectors and underlying molecular mechanisms of their modes of action. Many bacterial effectors are functionally equivalent to host proteins while being structurally divergent from them. Other effector proteins display new, previously unobserved functionalities. Here, we summarize the contribution of the structural characterization of effectors and effector-host protein complexes to our understanding of host subversion mechanisms used by the most commonly investigated Gram-negative bacterial pathogens. We describe in some detail the enzymatic activities discovered among effector proteins and how they affect various cellular processes.
Collapse
Affiliation(s)
- Andrey Grishin
- Department of Biochemistry, Microbiology, & Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Kevin Voth
- Department of Biochemistry, Microbiology, & Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Alla Gagarinova
- Department of Biochemistry, Microbiology, & Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Miroslaw Cygler
- Department of Biochemistry, Microbiology, & Immunology, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
39
|
Autophagy-A Story of Bacteria Interfering with the Host Cell Degradation Machinery. Pathogens 2021; 10:pathogens10020110. [PMID: 33499114 PMCID: PMC7911818 DOI: 10.3390/pathogens10020110] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a highly conserved and fundamental cellular process to maintain cellular homeostasis through recycling of defective organelles or proteins. In a response to intracellular pathogens, autophagy further acts as an innate immune response mechanism to eliminate pathogens. This review will discuss recent findings on autophagy as a reaction to intracellular pathogens, such as Salmonella typhimurium, Listeria monocytogenes, Mycobacterium tuberculosis, Staphylococcus aureus, and pathogenic Escherichia coli. Interestingly, while some of these bacteria have developed methods to use autophagy for their own benefit within the cell, others have developed fascinating mechanisms to evade recognition, to subvert the autophagic pathway, or to escape from autophagy.
Collapse
|
40
|
Thomas DR, Newton P, Lau N, Newton HJ. Interfering with Autophagy: The Opposing Strategies Deployed by Legionella pneumophila and Coxiella burnetii Effector Proteins. Front Cell Infect Microbiol 2020; 10:599762. [PMID: 33251162 PMCID: PMC7676224 DOI: 10.3389/fcimb.2020.599762] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Autophagy is a fundamental and highly conserved eukaryotic process, responsible for maintaining cellular homeostasis and releasing nutrients during times of starvation. An increasingly important function of autophagy is its role in the cell autonomous immune response; a process known as xenophagy. Intracellular pathogens are engulfed by autophagosomes and targeted to lysosomes to eliminate the threat to the host cell. To counteract this, many intracellular bacterial pathogens have developed unique approaches to overcome, evade, or co-opt host autophagy to facilitate a successful infection. The intracellular bacteria Legionella pneumophila and Coxiella burnetii are able to avoid destruction by the cell, causing Legionnaires' disease and Q fever, respectively. Despite being related and employing homologous Dot/Icm type 4 secretion systems (T4SS) to translocate effector proteins into the host cell, these pathogens have developed their own unique intracellular niches. L. pneumophila evades the host endocytic pathway and instead forms an ER-derived vacuole, while C. burnetii requires delivery to mature, acidified endosomes which it remodels into a large, replicative vacuole. Throughout infection, L. pneumophila effectors act at multiple points to inhibit recognition by xenophagy receptors and disrupt host autophagy, ensuring it avoids fusion with destructive lysosomes. In contrast, C. burnetii employs its effector cohort to control autophagy, hypothesized to facilitate the delivery of nutrients and membrane to support the growing vacuole and replicating bacteria. In this review we explore the effector proteins that these two organisms utilize to modulate the host autophagy pathway in order to survive and replicate. By better understanding how these pathogens manipulate this highly conserved pathway, we can not only develop better treatments for these important human diseases, but also better understand and control autophagy in the context of human health and disease.
Collapse
Affiliation(s)
| | | | | | - Hayley J. Newton
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Mei L, Qiu X, Jiang C, Yang A. Host Delipidation Mediated by Bacterial Effectors. Trends Microbiol 2020; 29:238-250. [PMID: 33092951 DOI: 10.1016/j.tim.2020.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
Protein lipidation, the covalent attachment of a lipid moiety to a target protein, plays a critical role in many cellular processes in eukaryotic cells. Bacterial pathogens secrete various effectors to subvert the host signaling pathway as a mechanism of microbial pathogenesis. An increasing number of effectors from diverse bacterial pathogens function as cysteine proteases to cause irreversible delipidation of host lipidated proteins. This in turn results in disruption of crucial lipidation-mediated host signal transduction, thereby enabling pathogen survival and replication. In this review, we discuss the role of the bacterial effectors in interactions with the host and highlight our knowledge of irreversible host delipidation, with a focus on the common concerted biochemical mechanisms of the bacterial effectors.
Collapse
Affiliation(s)
- Ligang Mei
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaofeng Qiu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Chen Jiang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China; Ultrafast Transient Materials Science Center, Institute of Advanced Interdisciplinary Studies, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
42
|
Swart AL, Gomez-Valero L, Buchrieser C, Hilbi H. Evolution and function of bacterial RCC1 repeat effectors. Cell Microbiol 2020; 22:e13246. [PMID: 32720355 DOI: 10.1111/cmi.13246] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 01/02/2023]
Abstract
Intracellular bacterial pathogens harbour genes, the closest homologues of which are found in eukaryotes. Regulator of chromosome condensation 1 (RCC1) repeat proteins are phylogenetically widespread and implicated in protein-protein interactions, such as the activation of the small GTPase Ran by its cognate guanine nucleotide exchange factor, RCC1. Legionella pneumophila and Coxiella burnetii, the causative agents of Legionnaires' disease and Q fever, respectively, harbour RCC1 repeat coding genes. Legionella pneumophila secretes the RCC1 repeat 'effector' proteins LegG1, PpgA and PieG into eukaryotic host cells, where they promote the activation of the pleiotropic small GTPase Ran, microtubule stabilisation, pathogen vacuole motility and intracellular bacterial growth as well as host cell migration. The RCC1 repeat effectors localise to the pathogen vacuole or the host plasma membrane and target distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself. Coxiella burnetii translocates the RCC1 repeat effector NopA into host cells, where the protein localises to nucleoli. NopA binds to Ran GTPase and promotes the nuclear accumulation of Ran(GTP), thus pertubing the import of the transcription factor NF-κB and innate immune signalling. Hence, divergent evolution of bacterial RCC1 repeat effectors defines the range of Ran GTPase cycle targets and likely allows fine-tuning of Ran GTPase activation by the pathogens at different cellular sites.
Collapse
Affiliation(s)
- Anna Leoni Swart
- Institute of Medical Microbiology, Faculty of Medicine, University of Zurich, Zürich, Switzerland
| | - Laura Gomez-Valero
- Institut Pasteur, Unité de Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Carmen Buchrieser
- Institut Pasteur, Unité de Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Hubert Hilbi
- Institute of Medical Microbiology, Faculty of Medicine, University of Zurich, Zürich, Switzerland
| |
Collapse
|
43
|
Yang A, Pantoom S, Wu YW. Distinct Mechanisms for Processing Autophagy Protein LC3-PE by RavZ and ATG4B. Chembiochem 2020; 21:3377-3382. [PMID: 32686895 PMCID: PMC7754449 DOI: 10.1002/cbic.202000359] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/10/2020] [Indexed: 11/06/2022]
Abstract
Autophagy is a conserved catabolic process involved in the elimination of proteins, organelles and pathogens in eukaryotic cells. Lipidated LC3 proteins that are conjugated to phosphatidylethanolamine (PE) play a key role in autophagosome biogenesis. Endogenous ATG4‐mediated deconjugation of LC3‐PE is required for LC3 recycling. However, the Legionella effector RavZ irreversibly deconjugates LC3‐PE to inhibit autophagy. It is not clear how ATG4 and RavZ process LC3‐PE with distinct modes. Herein, a series of semisynthetic LC3‐PE proteins containing C‐terminal mutations or insertions were used to investigate the relationship of the C‐terminal structure of LC3‐PE with ATG4/RavZ‐mediated deconjugation. Using a combination of molecular docking and biochemical assays, we found that Gln116, Phe119 and Gly120 of LC3‐PE are required for cleavage by both RavZ and ATG4B, whereas Glu117(LC3) is specific to cleavage by RavZ. The molecular ruler mechanism exists in the active site of ATG4B, but not in RavZ. Met63 and Gln64 at the active site of RavZ are involved in accommodating LC3 C‐terminal motif. Our findings show that the distinct binding modes of the LC3 C‐terminal motif (116–120) with ATG4 and RavZ might determine the specificity of cleavage site.
Collapse
Affiliation(s)
- Aimin Yang
- School of Life Sciences, Chongqing University, 401331, Chongqing, P. R. China
| | - Supansa Pantoom
- Translational Neurodegeneration Section "Albrecht-Kossel" Department of Neurology, University Medical Center Rostock, 18147, Rostock, Germany
| | - Yao-Wen Wu
- Department of Chemistry Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden.,Chemical Genomics Centre of the Max Planck Society, Otto-Hahn-Strasse 15, 44227, Dortmund, Germany.,Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| |
Collapse
|
44
|
Cong Y, Dinesh Kumar N, Mauthe M, Verlhac P, Reggiori F. Manipulation of selective macroautophagy by pathogens at a glance. J Cell Sci 2020; 133:133/10/jcs240440. [DOI: 10.1242/jcs.240440] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
ABSTRACT
Macroautophagy (hereafter autophagy) is a highly conserved catabolic pathway, which mediates the delivery of unwanted cytoplasmic structures and organelles to lysosomes for degradation. In numerous situations, autophagy is highly selective and exclusively targets specific intracellular components. Selective types of autophagy are a central element of our cell-autonomous innate immunity as they can mediate the turnover of viruses or bacteria, that gain access to the cytoplasm of the cell. Selective autophagy also modulates other aspects of our immunity by turning over specific immunoregulators. Throughout evolution, however, the continuous interaction between this fundamental cellular pathway and pathogens has led several pathogens to develop exquisite mechanisms to inhibit or subvert selective types of autophagy, to promote their intracellular multiplication. This Cell Science at a Glance article and the accompanying poster provides an overview of the selective autophagy of both pathogens, known as xenophagy, and of immunoregulators, and highlights a few archetypal examples that illustrate molecular strategies developed by viruses and bacteria to manipulate selective autophagy for their own benefit.
Collapse
Affiliation(s)
- Yingying Cong
- Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, The Netherlands
| | - Nilima Dinesh Kumar
- Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, The Netherlands
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, The Netherlands
| | - Mario Mauthe
- Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, The Netherlands
| | - Pauline Verlhac
- Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, The Netherlands
| |
Collapse
|
45
|
Park SW, Jun YW, Jeon P, Lee YK, Park JH, Lee SH, Lee JA, Jang DJ. LIR motifs and the membrane-targeting domain are complementary in the function of RavZ. BMB Rep 2020. [PMID: 31722778 PMCID: PMC6941762 DOI: 10.5483/bmbrep.2019.52.12.211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The bacterial effector protein RavZ is secreted by the intracellular pathogen Legionella pneumophila and inhibits host autophagy through an irreversible deconjugation of mammalian ATG8 (mATG8) proteins from autophagosome membranes. However, the roles of the LC3 interacting region (LIR) motifs in RavZ function remain unclear. In this study, we show that a membrane-targeting (MT) domain or the LIR motifs of RavZ play major or minor roles in RavZ function. A RavZ mutant that does not bind to mATG8 delipidated all forms of mATG8-phosphatidylethanolamine (PE) as efficiently as did wild-type RavZ. However, a RavZ mutant with a deletion of the MT domain selectively delipidated mATG8-PE less efficiently than did wild-type RavZ. Taken together, our results suggest that the effects of LIR motifs and the MT domain on RavZ activity are complementary and work through independent pathways.
Collapse
Affiliation(s)
- Sang-Won Park
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Yong-Woo Jun
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Pureum Jeon
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - You-Kyung Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Ju-Hui Park
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Seung-Hwan Lee
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Deok-Jin Jang
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| |
Collapse
|
46
|
Luo Y, Jiang C, Yu L, Yang A. Chemical Biology of Autophagy-Related Proteins With Posttranslational Modifications: From Chemical Synthesis to Biological Applications. Front Chem 2020; 8:233. [PMID: 32309274 PMCID: PMC7145982 DOI: 10.3389/fchem.2020.00233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/11/2020] [Indexed: 02/03/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) is an evolutionarily conserved lysosomal degradation pathway in all eukaryotic cells, which is critical for maintaining cell homeostasis. A series of autophagy-related (ATG) proteins are involved in the regulation of autophagy. The activities of ATG proteins are mainly modulated by posttranslational modifications (PTMs), such as phosphorylation, lipidation, acetylation, ubiquitination, and sumoylation. To tackle molecular mechanisms of autophagy, more and more researches are focusing on the roles of PTMs in regulation of the activity of ATG proteins and autophagy process. The protein ligation techniques have emerged as powerful tools for the chemical engineering of proteins with PTMs, and provided effective methods to elucidate the molecular mechanism and physiological significance of PTMs. Recently, several ATG proteins with PTM were prepared by protein ligation techniques such as native chemical ligation (NCL), expressed protein ligation (EPL), peptide hydrazide-based NCL, and Sortase A-mediated ligation (SML). More importantly, the synthesized ATG proteins are successfully used to probe the mechanism of autophagy. In this review, we summarize protein ligation techniques for the preparation of ATG proteins with PTMs. In addition, we highlight the biological applications of synthetic ATG proteins to probe the autophagy mechanism.
Collapse
Affiliation(s)
- Yu Luo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Chen Jiang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lihua Yu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
47
|
"Make way": Pathogen exploitation of membrane traffic. Curr Opin Cell Biol 2020; 65:78-85. [PMID: 32234681 DOI: 10.1016/j.ceb.2020.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/20/2020] [Indexed: 01/18/2023]
Abstract
Intracellular pathogens have evolved numerous strategies to manipulate their host cells to survive and replicate in a hostile environment. They often exploit membrane trafficking pathways to enter the cell, establish a replicative niche, avoid degradation and immune response, acquire nutrients and lastly, egress. Recent studies on membrane trafficking exploitation by intracellular pathogens have led to the discovery of novel and fascinating cell biology, including a noncanonical mechanism of ubiquitination and a novel mitophagy receptor. Thus, studying how pathogens target host cell membrane trafficking pathways is not only important for the development of new therapeutics, but also helps understanding fundamental mechanisms of cell biology.
Collapse
|
48
|
Abstract
Protein semisynthesis-defined herein as the assembly of a protein from a combination of synthetic and recombinant fragments-is a burgeoning field of chemical biology that has impacted many areas in the life sciences. In this review, we provide a comprehensive survey of this area. We begin by discussing the various chemical and enzymatic methods now available for the manufacture of custom proteins containing noncoded elements. This section begins with a discussion of methods that are more chemical in origin and ends with those that employ biocatalysts. We also illustrate the commonalities that exist between these seemingly disparate methods and show how this is allowing for the development of integrated chemoenzymatic methods. This methodology discussion provides the technical foundation for the second part of the review where we cover the great many biological problems that have now been addressed using these tools. Finally, we end the piece with a short discussion on the frontiers of the field and the opportunities available for the future.
Collapse
Affiliation(s)
| | - Tom W. Muir
- Department of Chemistry, Princeton University, Frick Laboratory, Princeton, New Jersey 08544, United States
| |
Collapse
|
49
|
Abstract
Autophagy is a conserved and fundamental cellular process mainly to recycle or eliminate dysfunctional cellular organelles or proteins. As a response to cellular stress, autophagy is used as a defense mechanism to combat the infection with pathogenic bacteria. However, many intracellular bacteria have developed diverse mechanisms to evade recognition, to manipulate the autophagic pathway, and to hijack the autophagosomal compartment for replication. In this review, we discuss recent understandings on how bacteria interact with host autophagy.
Collapse
Affiliation(s)
- Yao-Wen Wu
- a Department of Chemistry, Umeå Centre for Microbial Research , Umeå University , Umeå , Sweden.,b Chemical Genomics Centre of the Max Planck Society , Dortmund , Germany.,c Max Planck Institute of Molecular Physiology , Dortmund , Germany
| | - Fu Li
- a Department of Chemistry, Umeå Centre for Microbial Research , Umeå University , Umeå , Sweden.,b Chemical Genomics Centre of the Max Planck Society , Dortmund , Germany.,c Max Planck Institute of Molecular Physiology , Dortmund , Germany
| |
Collapse
|
50
|
Swart AL, Hilbi H. Phosphoinositides and the Fate of Legionella in Phagocytes. Front Immunol 2020; 11:25. [PMID: 32117224 PMCID: PMC7025538 DOI: 10.3389/fimmu.2020.00025] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/08/2020] [Indexed: 01/28/2023] Open
Abstract
Legionella pneumophila is the causative agent of a severe pneumonia called Legionnaires' disease. The environmental bacterium replicates in free-living amoebae as well as in lung macrophages in a distinct compartment, the Legionella-containing vacuole (LCV). The LCV communicates with a number of cellular vesicle trafficking pathways and is formed by a plethora of secreted bacterial effector proteins, which target host cell proteins and lipids. Phosphoinositide (PI) lipids are pivotal determinants of organelle identity, membrane dynamics and vesicle trafficking. Accordingly, eukaryotic cells tightly regulate the production, turnover, interconversion, and localization of PI lipids. L. pneumophila modulates the PI pattern in infected cells for its own benefit by (i) recruiting PI-decorated vesicles, (ii) producing effectors acting as PI interactors, phosphatases, kinases or phospholipases, and (iii) subverting host PI metabolizing enzymes. The PI conversion from PtdIns(3)P to PtdIns(4)P represents a decisive step during LCV maturation. In this review, we summarize recent progress on elucidating the strategies, by which L. pneumophila subverts host PI lipids to promote LCV formation and intracellular replication.
Collapse
Affiliation(s)
- A Leoni Swart
- Faculty of Medicine, Institute of Medical Microbiology, University of Zürich, Zurich, Switzerland
| | - Hubert Hilbi
- Faculty of Medicine, Institute of Medical Microbiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|