1
|
Kuo CF, Chen YY, Chiu CC, Chiu CW, Li TC, Chang YS, Tsao N. Comparative in vitro efficacy of AR-12 derivatives against Streptococcus pyogenes. J Antimicrob Chemother 2024:dkae462. [PMID: 39704166 DOI: 10.1093/jac/dkae462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVES Group A Streptococcus (GAS) results in invasive diseases. Our published studies show that AR-12 can directly kill GAS. However, AR-12 is toxic to the human microvascular endothelial cells (HMEC-1 cells) even at its MIC. In this study, we examined various AR-12 pyrrole derivatives, selected the most effective one and used it to combat GAS. METHODS The bacterial numbers after treatment with AR-12 derivatives were assessed using either spectrophotometry or the colony-forming unit assay. The integrity of cell envelope and the contents of proteins and nucleic acids in GAS were sequentially examined by staining with SYTOX Green, SYPRO or propidium iodide. The protein expression was assessed by western blotting. The cytotoxicity of AR-12 derivatives was evaluated using WST-1 assay, the lactate dehydrogenase release assay and Annexin V staining. RESULTS We tested AR-12 pyrrole derivatives P12, P12-3 and P12-8 on GAS growth and found that P12 and P12-8 were effective against various M-type strains. Both P12 and P12-8 disrupted the GAS envelope and reduced protein and nucleic acid content in GAS at their MICs. At sub-MIC levels, both P12 and P12-8 inhibited GAS chaperone protein and streptococcal pyrogenic exotoxin B expression. P12 and P12-8 also exhibited a synergistic effect with gentamicin against GAS. However, only P12-8 did not affect cell death at its MIC. Besides its bactericidal efficacy, P12-8 also enhanced the clearance of intracellular bacteria in GAS-infected A549 and HMEC-1 cells. CONCLUSIONS Among these three AR-12 derivatives, P12-8 had the best potential to be an alternative agent to fight against GAS.
Collapse
Affiliation(s)
- Chih-Feng Kuo
- School of Medicine, I-Shou University, Kaohsiung City, Taiwan
- Department of Nursing, I-Shou University, Kaohsiung City, Taiwan
| | - You-Yan Chen
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Ching-Chen Chiu
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Chih-Wei Chiu
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Tang-Chi Li
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Yu-Shan Chang
- Department of Laboratory Medicine, E-DA Hospital, Kaohsiung City, Taiwan
| | - Nina Tsao
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| |
Collapse
|
2
|
Pan JM, Gao J, Liu MJ, Zhu KC, Guo HY, Liu BS, Zhang N, Zhang DC. Functional Characterization of Galectin-8 from Golden Pompano Trachinotus ovatus Reveals Its Broad-Spectrum Antimicrobial Activity. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 27:21. [PMID: 39688731 DOI: 10.1007/s10126-024-10393-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024]
Abstract
Galectins exhibit a variety of biological functions through interactions with their ligands, including galactose and its derivatives. Tandem-repeat galectins, such as Galectin-8, can act as pattern recognition receptors to aggregate and neutralize bacterial pathogens. In this study, Galectin-8 was identified in Trachinotus ovatus (golden pompano). Galectin-8 consists of two carbohydrate recognition domains (CRDs) connected by a linker region. Furthermore, molecular docking analysis suggests that the C-terminal CRD can bind galactose, mannose, and N-acetylglucosamine at similar binding sites. ToGal-8 expression levels were highest in the brain and blood of healthy T. ovatus. However, following infection with Streptococcus agalactiae, expression levels in the spleen and head kidney surged at 48 h, while liver expression significantly decreased by 96 h. Cytoplasmic Galectin-8 expression was upregulated after stimulation by peptidoglycan compared with lipopolysaccharide. Recombinant ToGal-8 (rToGal-8) was produced using a prokaryotic expression system. This protein could agglutinate red blood cells from rabbits, carp, and T. ovatus independently of Ca2+. Moreover, it was also effective in aggregating and eliminating several bacterial strains, such as Staphylococcus aureus, Bacillus subtilis, Vibrio vulnificus, S. agalactiae, Pseudomonas aeruginosa, and Aeromonas hydrophila. Therefore, this study provides an in-depth analysis of the function of T. ovatus Galectin-8 for the first time, offering guidance for the healthy aquaculture of T. ovatus.
Collapse
Affiliation(s)
- Jin-Min Pan
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China
- School of Life Science, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jie Gao
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China
- School of Life Science, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Ming Jian Liu
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China
| | - Ke-Cheng Zhu
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, SouthChina Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences., Guangzhou, 510300, 231 xingang street West, Haizhu District., Guangdong, China
- Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, China
| | - Hua-Yang Guo
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China
- Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, China
| | - Bao-Suo Liu
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, SouthChina Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences., Guangzhou, 510300, 231 xingang street West, Haizhu District., Guangdong, China
- Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, China
| | - Nan Zhang
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, SouthChina Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences., Guangzhou, 510300, 231 xingang street West, Haizhu District., Guangdong, China
- Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, China
| | - Dian-Chang Zhang
- Sanya Tropical Fisheriers Research Institute, Sanya, 572108, Hainan Province, China.
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, SouthChina Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences., Guangzhou, 510300, 231 xingang street West, Haizhu District., Guangdong, China.
- Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, China.
| |
Collapse
|
3
|
Song Q, Li Q, Yang Y, Gao H, Han F. Antimicrobial Functions of Galectins from Fish, Mollusks, and Crustaceans: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24895-24907. [PMID: 39471068 DOI: 10.1021/acs.jafc.4c05412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Galectins are a member of the β-galactoside binding protein family, which play a pivotal role in the immune defense of vertebrates as a pattern recognition receptor and occupy an important position in the innate immune system of invertebrates. The study of galectins in aquatic organisms has only recently emerged. Galectins in aquatic animals exhibit agglutination activity toward bacteria, inhibit bacterial growth, and enhance phagocytosis of immune cells. Additionally, some galectins contribute to the antiviral immune defenses of aquatic animals. This review aims to review recent advancements in the antimicrobial mechanisms, molecular structures, and evolution of galectins from fish, mollusks, and crustaceans. The antimicrobial galectins, as crucial components in the innate immune defense, pave new avenues for developing innovative disease control strategies in aquaculture.
Collapse
Affiliation(s)
- Qing Song
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China
| | - Qiaoying Li
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Yao Yang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Haijun Gao
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| |
Collapse
|
4
|
Purić E, Nilsson UJ, Anderluh M. Galectin-8 inhibition and functions in immune response and tumor biology. Med Res Rev 2024; 44:2236-2265. [PMID: 38613488 DOI: 10.1002/med.22041] [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: 04/13/2023] [Revised: 03/03/2024] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Galectins are among organisms' most abundantly expressed lectins (carbohydrate-binding proteins) that specifically bind β-galactosides. They act not only outside the cell, where they bind to extracellular matrix glycans, but also inside the cell, where they have a significant impact on signaling pathways. Galectin-8 is a galectin family protein encoded by the LGALS8 gene. Its role is evident in both T- and B-cell immunity and in the innate immune response, where it acts directly on dendritic cells and induces some pro-inflammatory cytokines. Galectin-8 also plays an important role in the defense against bacterial and viral infections. It is known to promote antibacterial autophagy by recognizing and binding glycans present on the vacuolar membrane, thus acting as a danger receptor. The most important role of galectin-8 is the regulation of cancer growth, metastasis, tumor progression, and tumor cell survival. Importantly, the expression of galectins is typically higher in tumor tissues than in noncancerous tissues. In this review article, we focus on galectin-8 and its function in immune response, microbial infections, and cancer. Given all of these functions of galectin-8, we emphasize the importance of developing new and selective galectin-8 inhibitors and report the current status of their development.
Collapse
Affiliation(s)
- Edvin Purić
- Department of Pharmaceutical Chemistry, University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Ulf J Nilsson
- Department of Chemistry, Lund University, Lund, Sweden
| | - Marko Anderluh
- Department of Pharmaceutical Chemistry, University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| |
Collapse
|
5
|
Shi YA, Lu SL, Noda T, Chiu CH, Chiang-Ni C. Capsule-deficient group A Streptococcus evades autophagy-mediated killing in macrophages. mBio 2024; 15:e0077124. [PMID: 38819157 PMCID: PMC11253618 DOI: 10.1128/mbio.00771-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 06/01/2024] Open
Abstract
The hyaluronic acid capsule is crucial in protecting group A Streptococcus (GAS) against phagocytic killing. However, there have been reported outbreaks caused by capsule-deficient GAS strains, and the mechanisms underlying their evasion of immune clearance remain unclear. This study demonstrated that the capsule-deficient mutant [Cap(-)] of the emm1 strain increased survival within phagocytic cells compared to the wild-type strain [Cap(+)]. Although both Cap(+) and Cap(-) strains exhibited similar abilities to disrupt the phagosome, only the Cap(+) strain was colocalized with lysosomes and acidified compartments in phagocytic cells, indicating its susceptibility to autophagosome elimination. In contrast, the Cap(-) mutant evaded the recognition of galectin-8 and ubiquitin, impairing selective autophagy-mediated elimination. These findings suggest that a deficiency in the capsule could impair the intracellular elimination of GAS in macrophages, revealing previously unknown aspects of the host's recognition of the GAS capsule in macrophages. IMPORTANCE Group A Streptococcus (GAS) is a Gram-positive bacterium that causes diseases ranging from mild pharyngitis to severe necrotizing fasciitis. Phagocytic cells serve as the primary defense against bacterial infections, exhibiting remarkable efficiency in eliminating intracellular pathogens. The hyaluronic acid capsule is a critical virulence factor that contributes to the resistance of phagocytosis in GAS. Nevertheless, the outbreaks caused by GAS strains that lack the hyaluronic acid capsule have been reported, and the selective advantage of capsule-deficient strains during infection is not fully understood. This study showed that the autophagic adaptor proteins recognize the capsulated GAS strain but not the capsule-deficient mutant, indicating that the hyaluronic acid capsule could be the autophagic target in macrophages. These findings imply that the hyaluronic acid capsule of GAS actually enhances its elimination within phagocytic cells, subverting the understanding of the capsule in GAS pathogenesis.
Collapse
Affiliation(s)
- Yong-An Shi
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shiou-Ling Lu
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Takeshi Noda
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Cheng-Hsun Chiu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chuan Chiang-Ni
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| |
Collapse
|
6
|
Nishioka ST, Snipper J, Lee J, Schapiro J, Zhang RZ, Abe H, Till A, Okumura CYM. Group A Streptococcus induces lysosomal dysfunction in THP-1 macrophages. Infect Immun 2024; 92:e0014124. [PMID: 38722166 PMCID: PMC11237432 DOI: 10.1128/iai.00141-24] [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: 04/01/2024] [Accepted: 04/11/2024] [Indexed: 06/12/2024] Open
Abstract
The human-specific bacterial pathogen group A Streptococcus (GAS) is a significant cause of morbidity and mortality. Macrophages are important to control GAS infection, but previous data indicate that GAS can persist in macrophages. In this study, we detail the molecular mechanisms by which GAS survives in THP-1 macrophages. Our fluorescence microscopy studies demonstrate that GAS is readily phagocytosed by macrophages, but persists within phagolysosomes. These phagolysosomes are not acidified, which is in agreement with our findings that GAS cannot survive in low pH environments. We find that the secreted pore-forming toxin Streptolysin O (SLO) perforates the phagolysosomal membrane, allowing leakage of not only protons but also large proteins including the lysosomal protease cathepsin B. Additionally, GAS recruits CD63/LAMP-3, which may contribute to lysosomal permeabilization, especially in the absence of SLO. Thus, although GAS does not inhibit fusion of the lysosome with the phagosome, it has multiple mechanisms to prevent proper phagolysosome function, allowing for persistence of the bacteria within the macrophage. This has important implications for not only the initial response but also the overall functionality of the macrophages, which may lead to the resulting pathologies in GAS infection. Our data suggest that therapies aimed at improving macrophage function may positively impact patient outcomes in GAS infection.
Collapse
Affiliation(s)
- Scott T Nishioka
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Joshua Snipper
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Jimin Lee
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Joshua Schapiro
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Robert Z Zhang
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Hyewon Abe
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Andreas Till
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
- The San Diego Center for Systems Biology, University of California San Diego, La Jolla, California, USA
- University Hospital of Bonn, Bonn, Germany
| | | |
Collapse
|
7
|
Su MSW, Cheng YL, Lin YS, Wu JJ. Interplay between group A Streptococcus and host innate immune responses. Microbiol Mol Biol Rev 2024; 88:e0005222. [PMID: 38451081 PMCID: PMC10966951 DOI: 10.1128/mmbr.00052-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
SUMMARYGroup A Streptococcus (GAS), also known as Streptococcus pyogenes, is a clinically well-adapted human pathogen that harbors rich virulence determinants contributing to a broad spectrum of diseases. GAS is capable of invading epithelial, endothelial, and professional phagocytic cells while evading host innate immune responses, including phagocytosis, selective autophagy, light chain 3-associated phagocytosis, and inflammation. However, without a more complete understanding of the different ways invasive GAS infections develop, it is difficult to appreciate how GAS survives and multiplies in host cells that have interactive immune networks. This review article attempts to provide an overview of the behaviors and mechanisms that allow pathogenic GAS to invade cells, along with the strategies that host cells practice to constrain GAS infection. We highlight the counteractions taken by GAS to apply virulence factors such as streptolysin O, nicotinamide-adenine dinucleotidase, and streptococcal pyrogenic exotoxin B as a hindrance to host innate immune responses.
Collapse
Affiliation(s)
- Marcia Shu-Wei Su
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Lin Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yee-Shin Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jiunn-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
8
|
Wang J, He Y, Zhou D. The role of ubiquitination in microbial infection induced endothelial dysfunction: potential therapeutic targets for sepsis. Expert Opin Ther Targets 2023; 27:827-839. [PMID: 37688775 DOI: 10.1080/14728222.2023.2257888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/11/2023] [Accepted: 09/07/2023] [Indexed: 09/11/2023]
Abstract
INTRODUCTION The ubiquitin system is an evolutionarily conserved and universal means of protein modification that regulates many essential cellular processes. Endothelial dysfunction plays a critical role in the pathophysiology of sepsis and organ failure. However, the mechanisms underlying the ubiquitination-mediated regulation on endothelial dysfunction are not fully understood. AREAS COVERED Here we review the advances in basic and clinical research for relevant papers in PubMed database. We attempt to provide an updated overview of diverse ubiquitination events in endothelial cells, discussing the fundamental role of ubiquitination mediated regulations involving in endothelial dysfunction to provide potential therapeutic targets for sepsis. EXPERT OPINION The central event underlying sepsis syndrome is the overwhelming host inflammatory response to the pathogen infection, leading to endothelial dysfunction. As the key components of the ubiquitin system, E3 ligases are at the center stage of the battle between host and microbial pathogens. Such a variety of ubiquitination regulates a multitude of cellular regulatory processes, including signal transduction, autophagy, inflammasome activation, redox reaction and immune response and so forth. In this review, we discuss the many mechanisms of ubiquitination-mediated regulation with a focus on those that modulate endothelial function to provide potential therapeutic targets for the management of sepsis.
Collapse
Affiliation(s)
- Junshuai Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Yang He
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Daixing Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| |
Collapse
|
9
|
Weng IC, Chen HL, Lin WH, Liu FT. Sialylation of cell surface glycoconjugates modulates cytosolic galectin-mediated responses upon organelle damage : Minireview. Glycoconj J 2023; 40:295-303. [PMID: 37052731 DOI: 10.1007/s10719-023-10112-z] [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: 01/03/2023] [Revised: 02/26/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
Sialylation is an important terminal modification of glycoconjugates that mediate diverse functions in physiology and disease. In this review we focus on how altered cell surface sialylation status is sensed by cytosolic galectins when the integrity of intracellular vesicles or organelles is compromised to expose luminal glycans to the cytosolic milieu, and how this impacts galectin-mediated cellular responses. In addition, we discuss the roles of mammalian sialidases on the cell surface, in the organelle lumen and cytosol, and raise the possibility that intracellular glycan processing may be critical in controlling various galectin-mediated responses when cells encounter stress.
Collapse
Affiliation(s)
- I-Chun Weng
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Hung-Lin Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wei-Han Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
10
|
Zhang Y, Liu X, Klionsky DJ, Lu B, Zhong Q. Manipulating autophagic degradation in human diseases: from mechanisms to interventions. LIFE MEDICINE 2022; 1:120-148. [PMID: 39871921 PMCID: PMC11749641 DOI: 10.1093/lifemedi/lnac043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/08/2022] [Indexed: 01/29/2025]
Abstract
Targeted degradation, having emerged as a powerful and promising strategy in drug discovery in the past two decades, has provided a solution for many once undruggable targets involved in various diseases. While earlier targeted degradation tools, as exemplified by PROteolysis-TArgeting Chimera (PROTAC), focused on harnessing the ubiquitin-proteasome system, novel approaches that aim to utilize autophagy, a potent, lysosome-dependent degradation pathway, have also surfaced recently as promising modalities. In this review, we first introduce the mechanisms that establish selectivity in autophagy, which provides the rationales for autophagy-based targeted degradation; we also provide an overview on the panoply of cellular machinery involved in this process, an arsenal that could be potentially harnessed. On this basis, we propose four strategies for designing autophagy-based targeted degraders, including Tagging Targets, Directly Engaging Targets, Initiating Autophagy at Targets, and Phagophore-Tethering to Targets. We introduce the current frontiers in this field, including AUtophagy-TArgeting Chimera (AUTAC), Targeted Protein Autophagy (TPA), AUTOphagy-TArgeting Chimera (AUTOTAC, not to be confused with AUTAC), AuTophagosome TEthering Compound (ATTEC), and other experimental approaches as case studies for each strategy. Finally, we put forward a workflow for generating autophagy-based degraders and some important questions that may guide and inspire the process.
Collapse
Affiliation(s)
- Yiqing Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 20025, China
| | - Xiaoxia Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 20025, China
| | - Daniel J Klionsky
- Department of Molecular, Cellular, and Developmental Biology, and the Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109-2216, USA
| | - Boxun Lu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Huashan Hospital, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 20025, China
| |
Collapse
|
11
|
Borgo GM, Burke TP, Tran CJ, Lo NTN, Engström P, Welch MD. A patatin-like phospholipase mediates Rickettsia parkeri escape from host membranes. Nat Commun 2022; 13:3656. [PMID: 35760786 PMCID: PMC9237051 DOI: 10.1038/s41467-022-31351-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/15/2022] [Indexed: 12/25/2022] Open
Abstract
Rickettsia species of the spotted fever group are arthropod-borne obligate intracellular bacteria that can cause mild to severe human disease. These bacteria invade host cells, replicate in the cell cytosol, and spread from cell to cell. To access the host cytosol and avoid immune detection, they escape membrane-bound vacuoles by expressing factors that disrupt host membranes. Here, we show that a patatin-like phospholipase A2 enzyme (Pat1) facilitates Rickettsia parkeri infection by promoting escape from host membranes and cell-cell spread. Pat1 is important for infection in a mouse model and, at the cellular level, is crucial for efficiently escaping from single and double membrane-bound vacuoles into the host cytosol, and for avoiding host galectins that mark damaged membranes. Pat1 is also important for avoiding host polyubiquitin, preventing recruitment of autophagy receptor p62, and promoting actin-based motility and cell-cell spread. Pathogenic Rickettsia species are arthropod-borne, obligate intracellular bacteria that invade host cells, replicate in the cell cytosol, and spread from cell to cell. Here, Borgo et al. identify a Rickettsia phospholipase enzyme that is important for infection by helping the bacteria escape from host cell vacuoles into the host cytosol, preventing targeting by autophagy, and promoting bacterial motility and spread to other cells.
Collapse
Affiliation(s)
- Gina M Borgo
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Thomas P Burke
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.,Department of Microbiology & Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Cuong J Tran
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Nicholas T N Lo
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Patrik Engström
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.,Primordial Genetics, San Diego, CA, USA
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
12
|
Aitkulova A, Mukhtarova K, Zholdybayeva E, Medetov Y, Dzhamantayeva B, Kassymbek K, Utupov T, Akhmetollayev I, Akshulakov S, Kulmambetova G, Ramankulov Y. Activated leukocyte cell adhesion molecule/cluster of differentiation 166 rs10933819 (G>A) variant is associated with familial intracranial aneurysms. Biomed Rep 2022; 17:65. [PMID: 35815187 PMCID: PMC9260160 DOI: 10.3892/br.2022.1548] [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: 01/12/2022] [Accepted: 04/01/2022] [Indexed: 11/06/2022] Open
Abstract
Rupture of intracranial aneurysms (IAs) is the most common cause of subarachnoid hemorrhage (SAH). Currently, there is sufficient evidence to indicate that inflammatory responses contribute to aneurysm rupture. Moreover, the familial occurrence of SAH suggests that genetic factors may be involved in disease susceptibility. In the present study, a clinically proven case of IA in a patient who is a heterozygous mutation carrier of the activated leukocyte cell adhesion molecule (ALCAM)/cluster of differentiation 166 (CD166) gene, is reported. Genomic DNA was extracted from two siblings diagnosed with SAH and other available family members. A variant prioritization strategy that focused on functional prediction, frequency, predicted pathogenicity, and segregation within the family was employed. Sanger sequencing was also performed on the unaffected relatives to assess the segregation of variants within the phenotype. The verified mutations were sequenced in 145 ethnicity-matched healthy individuals. Based on whole exome sequencing data obtained from three individuals, two of whom were diagnosed with IAs, the single-nucleotide variant rs10933819 was prioritized in the family. Only one variant, rs10933819 (G>A), in ALCAM co-segregated with the phenotype, and this mutation was absent in ethnicity-matched healthy individuals. Collectively, ALCAM c1382 G>A p.Gly229Val was identified, for the first time, as a pathogenic mutation in this IA pedigree.
Collapse
Affiliation(s)
- Akbota Aitkulova
- National Center for Biotechnology, Nur Sultan 010000, Republic of Kazakhstan
| | - Kymbat Mukhtarova
- National Center for Biotechnology, Nur Sultan 010000, Republic of Kazakhstan
| | - Elena Zholdybayeva
- National Center for Biotechnology, Nur Sultan 010000, Republic of Kazakhstan
| | - Yerkin Medetov
- National Center of Neurosurgery, Nazarbayev University, Nur Sultan 010000, Republic of Kazakhstan
| | - Botagoz Dzhamantayeva
- National Center of Neurosurgery, Nazarbayev University, Nur Sultan 010000, Republic of Kazakhstan
| | - Kuat Kassymbek
- National Center for Biotechnology, Nur Sultan 010000, Republic of Kazakhstan
| | - Talgat Utupov
- National Center for Biotechnology, Nur Sultan 010000, Republic of Kazakhstan
| | - Ilyas Akhmetollayev
- National Center for Biotechnology, Nur Sultan 010000, Republic of Kazakhstan
| | - Serik Akshulakov
- National Center of Neurosurgery, Nazarbayev University, Nur Sultan 010000, Republic of Kazakhstan
| | | | - Yerlan Ramankulov
- National Center for Biotechnology, Nur Sultan 010000, Republic of Kazakhstan
| |
Collapse
|
13
|
Chen P, Yang J, Wu N, Han B, Kastelic JP, Gao J. Streptococcus lutetiensis Induces Autophagy via Oxidative Stress in Bovine Mammary Epithelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2549772. [PMID: 35178153 PMCID: PMC8843784 DOI: 10.1155/2022/2549772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/11/2022] [Indexed: 02/07/2023]
Abstract
Streptococcus lutetiensis, an emerging pathogen causing bovine mastitis, has not been well characterized. We reported that S. lutetiensis was pathogenic both in vivo and in vitro and caused inflammatory reactions in the mammary gland. However, roles of autophagy and oxidative stress in the pathogenesis of S. lutetiensis-induced mastitis are unclear. In this study, an autophagy model of S. lutetiensis-infected bovine mammary epithelial cells (bMECs) was used to assess oxidative stress and autophagy flux. Expressions of Beclin1, light chain 3II, and Sequestosome 1/p62 were elevated in bMECs after S. lutetiensis infection. In addition, autophagosome and lysosome formation confirmed autophagy occurred. Based on LysoTracker Red and acridine orange, lysosome degradation was blocked, and lower expressions of lysosomal-associated membrane protein 2, cathepsins D, and cathepsins L confirmed lysosomal damage. Concurrently, the nuclear factor erythroid 2-related factor 2 (Nrf2), kelch-like ECH-associated protein 1 (Keap1), heme oxygenase 1 (HO1), and NAD (P)H: quinone oxidoreductase 1 (NQO1), and basilic proteins associated with the Nrf2/Keap1 signaling pathway, were detected. Decreased keap1 and increased Nrf2, HO1, NQO1, and reactive oxygen species (ROS) indicated increased oxidative stress. Treatment with N-Acetyl-L-cysteine (NAC), an ROS inhibitor, decreased both oxidative stress and autophagy. Therefore, we concluded that S. lutetiensis caused intracellular oxidative stress and autophagy in bMECs. In addition, crosstalk between autophagy and oxidative stress affected the autophagic flux and blocked downstream autophagy. The Nrf2-keap1-p62 pathway participated in this process, with ROS acting upstream of these effects, interfering with normal cell functions.
Collapse
Affiliation(s)
- Peng Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jingyue Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Naiwen Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
14
|
Tripathi-Giesgen I, Behrends C, Alpi AF. The ubiquitin ligation machinery in the defense against bacterial pathogens. EMBO Rep 2021; 22:e52864. [PMID: 34515402 PMCID: PMC8567218 DOI: 10.15252/embr.202152864] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/21/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
The ubiquitin system is an important part of the host cellular defense program during bacterial infection. This is in particular evident for a number of bacteria including Salmonella Typhimurium and Mycobacterium tuberculosis which—inventively as part of their invasion strategy or accidentally upon rupture of seized host endomembranes—become exposed to the host cytosol. Ubiquitylation is involved in the detection and clearance of these bacteria as well as in the activation of innate immune and inflammatory signaling. Remarkably, all these defense responses seem to emanate from a dense layer of ubiquitin which coats the invading pathogens. In this review, we focus on the diverse group of host cell E3 ubiquitin ligases that help to tailor this ubiquitin coat. In particular, we address how the divergent ubiquitin conjugation mechanisms of these ligases contribute to the complexity of the anti‐bacterial coating and the recruitment of different ubiquitin‐binding effectors. We also discuss the activation and coordination of the different E3 ligases and which strategies bacteria evolved to evade the activities of the host ubiquitin system.
Collapse
Affiliation(s)
- Ishita Tripathi-Giesgen
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-University München, München, Germany
| | - Arno F Alpi
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
15
|
Glycans in autophagy, endocytosis and lysosomal functions. Glycoconj J 2021; 38:625-647. [PMID: 34390447 PMCID: PMC8497297 DOI: 10.1007/s10719-021-10007-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Glycans have been shown to function as versatile molecular signals in cells. This prompted us to look at their roles in endocytosis, endolysosomal system and autophagy. We start by introducing the cell biological aspects of these pathways, the concept of the sugar code, and provide an overview on the role of glycans in the targeting of lysosomal proteins and in lysosomal functions. Moreover, we review evidence on the regulation of endocytosis and autophagy by glycans. Finally, we discuss the emerging concept that cytosolic exposure of luminal glycans, and their detection by endogenous lectins, provides a mechanism for the surveillance of the integrity of the endolysosomal compartments, and serves their eventual repair or disposal.
Collapse
|
16
|
Caridi B, Doncheva D, Sivaprasad S, Turowski P. Galectins in the Pathogenesis of Common Retinal Disease. Front Pharmacol 2021; 12:687495. [PMID: 34079467 PMCID: PMC8165321 DOI: 10.3389/fphar.2021.687495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Diseases of the retina are major causes of visual impairment and blindness in developed countries and, due to an ageing population, their prevalence is continually rising. The lack of effective therapies and the limitations of those currently in use highlight the importance of continued research into the pathogenesis of these diseases. Vascular endothelial growth factor (VEGF) plays a major role in driving vascular dysfunction in retinal disease and has therefore become a key therapeutic target. Recent evidence also points to a potentially similarly important role of galectins, a family of β-galactoside-binding proteins. Indeed, they have been implicated in regulating fundamental processes, including vascular hyperpermeability, angiogenesis, neuroinflammation, and oxidative stress, all of which also play a prominent role in retinopathies. Here, we review direct evidence for pathological roles of galectins in retinal disease. In addition, we extrapolate potential roles of galectins in the retina from evidence in cancer, immune and neuro-biology. We conclude that there is value in increasing understanding of galectin function in retinal biology, in particular in the context of the retinal vasculature and microglia. With greater insight, recent clinical developments of galectin-targeting drugs could potentially also be of benefit to the clinical management of many blinding diseases.
Collapse
Affiliation(s)
- Bruna Caridi
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Dilyana Doncheva
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Sobha Sivaprasad
- UCL Institute of Ophthalmology, University College London, London, United Kingdom.,NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
17
|
Batu ED, Vezir E, Öğüş E, Özbaş Demirel Ö, Akpınar G, Demir S, Özen S. Galectin-3: a new biomarker for differentiating periodic fever, adenitis, pharyngitis, aphthous stomatitis (PFAPA) syndrome from familial Mediterranean fever? Rheumatol Int 2021; 42:71-80. [PMID: 33709179 DOI: 10.1007/s00296-021-04827-1] [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] [Received: 02/02/2021] [Accepted: 03/01/2021] [Indexed: 10/21/2022]
Abstract
Differentiating PFAPA (periodic fever, aphthosis, pharyngitis, and adenitis) syndrome from familial Mediterranean fever (FMF) could be challenging in some cases. Galectin-3 is a lectin with regulatory functions in apoptosis and inflammation. We aimed to test whether galectin-3 could be a biomarker for differentiating PFAPA syndrome from FMF. Patients with PFAPA syndrome, FMF, cryopyrin-associated periodic syndrome (CAPS), and streptococcal pharyngitis, and healthy controls were included in this study. Serum galectin-3 levels were measured using enzyme-linked immunosorbent assay. Eighty-seven patients (36 with PFAPA, 39 with FMF, 8 with CAPS, 4 with streptococcal pharyngitis), and 17 healthy controls were included. Blood samples were drawn during attacks from 20 PFAPA and 7 FMF patients and attack-free periods from 22 PFAPA, 35 FMF, and 8 CAPS patients. The median serum galectin-3 level in the PFAPA-attack group (1.025 ng/ml) was significantly lower than the levels in healthy control (2.367 ng/ml), streptococcal pharyngitis (3.021 ng/ml), FMF attack (2.402 ng/ml), and FMF-attack-free groups (2.797 ng/ml) (p = 0.006, 0.03, 0.01, and < 0.001, respectively). PFAPA-attack-free group had lower galectin-3 levels than the FMF-attack-free group (1.794 vs. 2.797 ng/ml, respectively; p = 0.01). Galectin-3 levels did not differ significantly between CAPS and attack-free PFAPA patients (1.439 ng/ml vs. 1.794 ng/ml, respectively; p = 0.63). In our study, for the first time, we defined galectin-3 as a promising biomarker that differs between PFAPA and FMF patients during both disease flares and attack-free periods. Further studies with high number of patients could validate its role as a biomarker.
Collapse
Affiliation(s)
- Ezgi D Batu
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey.
| | - Emine Vezir
- Division of Allergy and Immunology, Department of Pediatrics, Ankara Health Research and Application Center, University of Health Sciences, Ankara, Turkey
| | - Elmas Öğüş
- Department of Biochemistry, Ankara Health Research and Application Center, University of Health Sciences, Ankara, Turkey
| | - Özlem Özbaş Demirel
- Department of Biochemistry, Ankara Health Research and Application Center, University of Health Sciences, Ankara, Turkey
| | - Gizem Akpınar
- Department of Pediatrics, Ankara Health Research and Application Center, University of Health Sciences, Ankara, Turkey
| | - Selcan Demir
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey
| | - Seza Özen
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey
| |
Collapse
|
18
|
Hong MH, Weng IC, Li FY, Lin WH, Liu FT. Intracellular galectins sense cytosolically exposed glycans as danger and mediate cellular responses. J Biomed Sci 2021; 28:16. [PMID: 33663512 PMCID: PMC7931364 DOI: 10.1186/s12929-021-00713-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/07/2021] [Indexed: 12/18/2022] Open
Abstract
Galectins are animal lectins that recognize carbohydrates and play important roles in maintaining cellular homeostasis. Recent studies have indicated that under a variety of challenges, intracellular galectins bind to host glycans displayed on damaged endocytic vesicles and accumulate around these damaged organelles. Accumulated galectins then engage cellular proteins and subsequently control cellular responses, such as autophagy. In this review, we have summarized the stimuli that lead to the accumulation of galectins, the molecular mechanisms of galectin accumulation, and galectin-mediated cellular responses, and elaborate on the differential regulatory effects among galectins.
Collapse
Affiliation(s)
- Ming-Hsiang Hong
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - I-Chun Weng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Fang-Yen Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Han Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
19
|
Ma PY, Tan JE, Hee EW, Yong DWX, Heng YS, Low WX, Wu XH, Cletus C, Kumar Chellappan D, Aung K, Yong CY, Liew YK. Human Genetic Variation Influences Enteric Fever Progression. Cells 2021; 10:cells10020345. [PMID: 33562108 PMCID: PMC7915608 DOI: 10.3390/cells10020345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 01/06/2023] Open
Abstract
In the 21st century, enteric fever is still causing a significant number of mortalities, especially in high-risk regions of the world. Genetic studies involving the genome and transcriptome have revealed a broad set of candidate genetic polymorphisms associated with susceptibility to and the severity of enteric fever. This review attempted to explain and discuss the past and the most recent findings on human genetic variants affecting the progression of Salmonella typhoidal species infection, particularly toll-like receptor (TLR) 4, TLR5, interleukin (IL-) 4, natural resistance-associated macrophage protein 1 (NRAMP1), VAC14, PARK2/PACRG, cystic fibrosis transmembrane conductance regulator (CFTR), major-histocompatibility-complex (MHC) class II and class III. These polymorphisms on disease susceptibility or progression in patients could be related to multiple mechanisms in eliminating both intracellular and extracellular Salmonella typhoidal species. Here, we also highlighted the limitations in the studies reported, which led to inconclusive results in association studies. Nevertheless, the knowledge obtained through this review may shed some light on the development of risk prediction tools, novel therapies as well as strategies towards developing a personalised typhoid vaccine.
Collapse
Affiliation(s)
- Pei Yee Ma
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Jing En Tan
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.E.T.); (E.W.H.); (D.W.X.Y.); (Y.S.H.); (W.X.L.); (X.H.W.); (C.C.)
| | - Edd Wyn Hee
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.E.T.); (E.W.H.); (D.W.X.Y.); (Y.S.H.); (W.X.L.); (X.H.W.); (C.C.)
| | - Dylan Wang Xi Yong
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.E.T.); (E.W.H.); (D.W.X.Y.); (Y.S.H.); (W.X.L.); (X.H.W.); (C.C.)
| | - Yi Shuan Heng
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.E.T.); (E.W.H.); (D.W.X.Y.); (Y.S.H.); (W.X.L.); (X.H.W.); (C.C.)
| | - Wei Xiang Low
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.E.T.); (E.W.H.); (D.W.X.Y.); (Y.S.H.); (W.X.L.); (X.H.W.); (C.C.)
| | - Xun Hui Wu
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.E.T.); (E.W.H.); (D.W.X.Y.); (Y.S.H.); (W.X.L.); (X.H.W.); (C.C.)
| | - Christy Cletus
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.E.T.); (E.W.H.); (D.W.X.Y.); (Y.S.H.); (W.X.L.); (X.H.W.); (C.C.)
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Kyan Aung
- Department of Pathology, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Chean Yeah Yong
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia;
| | - Yun Khoon Liew
- Department of Life Sciences, International Medical University, Kuala Lumpur 57000, Malaysia;
- Correspondence:
| |
Collapse
|
20
|
Lin CY, Nozawa T, Minowa-Nozawa A, Toh H, Hikichi M, Iibushi J, Nakagawa I. Autophagy Receptor Tollip Facilitates Bacterial Autophagy by Recruiting Galectin-7 in Response to Group A Streptococcus Infection. Front Cell Infect Microbiol 2021; 10:583137. [PMID: 33425778 PMCID: PMC7786282 DOI: 10.3389/fcimb.2020.583137] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Bacterial autophagy—a type of macroautophagy that is also termed xenophagy—selectively targets intracellular bacteria such as group A Streptococcus (GAS), a ubiquitous pathogen that causes numerous serious diseases, including pharyngitis, skin infections, and invasive life-threatening infections. Although bacterial autophagy is known to eliminate invading bacteria via the action of autophagy receptors, the underlying mechanism remains unclear. Herein, we elucidated that Tollip functions as a bacterial-autophagy receptor in addition to participating involved in the intracellular immunity mechanism that defends against bacterial infection. Tollip was recruited to GAS-containing endosomal vacuoles prior to the escape of GAS into the cytosol; additionally, Tollip knockout disrupted the recruitment of other autophagy receptors, such as NBR1, TAX1BP1, and NDP52, to GAS-containing autophagosomes and led to prolonged intracellular survival of GAS. Furthermore, Tollip was found to be required for the recruitment of galectin-1 and -7 to GAS-containing autophagosomes, and immunoprecipitation results indicated that Tollip interacts with galectin-7. Lastly, our data also revealed that galectin-1 and -7 are involved in the restriction of GAS replication in cells. These results demonstrated that Tollip modulates bacterial autophagy by recruiting other autophagy receptors and galectins.
Collapse
Affiliation(s)
- Ching-Yu Lin
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsuko Minowa-Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirotaka Toh
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Miyako Hikichi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junpei Iibushi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
21
|
Ayona D, Fournier PE, Henrissat B, Desnues B. Utilization of Galectins by Pathogens for Infection. Front Immunol 2020; 11:1877. [PMID: 32973776 PMCID: PMC7466766 DOI: 10.3389/fimmu.2020.01877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022] Open
Abstract
Galectins are glycan-binding proteins which are expressed by many different cell types and secreted extracellularly. These molecules are well-known regulators of immune responses and involved in a broad range of cellular and pathophysiological functions. During infections, host galectins can either avoid or facilitate infections by interacting with host cells- and/or pathogen-derived glycoconjugates and less commonly, with proteins. Some pathogens also express self-produced galectins to interfere with host immune responses. This review summarizes pathogens which take advantage of host- or pathogen-produced galectins to establish the infection.
Collapse
Affiliation(s)
- Diyoly Ayona
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | | | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques, CNRS, Aix-Marseille University, Marseille, France
- USC1408 Architecture et Fonction des Macromolécules Biologiques, Institut National de la Recherche Agronomique, Marseille, France
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Benoit Desnues
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
22
|
Integrin α5β1, as a Receptor of Fibronectin, Binds the FbaA Protein of Group A Streptococcus To Initiate Autophagy during Infection. mBio 2020; 11:mBio.00771-20. [PMID: 32518187 PMCID: PMC7371361 DOI: 10.1128/mbio.00771-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Autophagy is generally considered a strategy used by the innate immune system to eliminate invasive pathogens through capturing and transferring them to lysosomes. Currently, researchers pay more attention to how virulence factors secreted by GAS regulate the autophagic process. Here, we provide the first evidence that the structural protein FbaA of M1 GAS strain SF370 is a potent inducer of autophagy in epithelial cells. Furthermore, we demonstrate that integrin α5β1 in epithelial cells in vitro and in vivo acts as a receptor to initiate the signaling for inducing autophagy by binding to FbaA of M1 GAS strain SF370 via Fn. Our study reveals the underlying mechanisms by which pathogens induce Fn-integrin α5β1 to trigger autophagy in a conserved pattern in epithelial cells. Group A Streptococcus (GAS), one of the most common extracellular pathogens, has been reported to invade epithelial and endothelial cells. Our results reveal that M1 GAS strain SF370 can be effectively eliminated by respiratory epithelial cells. Emerging evidence indicates that autophagy is an important strategy for nonphagocytes to eliminate intracellular bacteria. Upon pathogen recognition, cell surface receptors can directly trigger autophagy, which is a critical step in controlling infection. However, the mechanisms of how cells sense invading bacteria and use this information specifically to trigger autophagy remain unclear. In this study, we stimulated cells and infected mice with M and FbaA mutants of M1 GAS strain SF370 or with purified M and FbaA proteins (two critical surface structural proteins of GAS), and found that only FbaA protein was involved in autophagy induction. Furthermore, the FbaA protein induced autophagy independent of common pattern recognition receptors (such as Toll-like receptors); rather, it relies on binding to integrin α5β1 expressed on the cell surface, which is mediated by extracellular matrix protein fibronectin (Fn). The FbaA-Fn-integrin α5β1 complex activates Beclin-1 through the mTOR-ULK1–Beclin-1 pathway, which enables the Beclin-1/Vps34 complex to recruit Rab7 and, ultimately, to promote the formation of autophagosomes. By knocking down integrin α5β1, Fn, Atg5, Beclin-1, and ULK1 in Hep2 cells and deleting Atg5 or integrin α5β1 in mice, we reveal a novel role for integrin α5β1 in inducing autophagy. Our study demonstrates that integrin α5β1, through interacting with pathogen components, initiates effective host innate immunity against invading intracellular pathogens.
Collapse
|
23
|
Galectins in the brain: advances in neuroinflammation, neuroprotection and therapeutic opportunities. Curr Opin Neurol 2020; 33:381-390. [DOI: 10.1097/wco.0000000000000812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
A MicroRNA Network Controls Legionella pneumophila Replication in Human Macrophages via LGALS8 and MX1. mBio 2020; 11:mBio.03155-19. [PMID: 32209695 PMCID: PMC7157531 DOI: 10.1128/mbio.03155-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cases of Legionella pneumophila pneumonia occur worldwide, with potentially fatal outcome. When causing human disease, Legionella injects a plethora of virulence factors to reprogram macrophages to circumvent immune defense and create a replication niche. By analyzing Legionella-induced changes in miRNA expression and genomewide chromatin modifications in primary human macrophages, we identified a cell-autonomous immune network restricting Legionella growth. This network comprises three miRNAs governing expression of the cytosolic RNA receptor DDX58/RIG-I, the tumor suppressor TP53, the antibacterial effector LGALS8, and MX1, which has been described as an antiviral factor. Our findings for the first time link TP53, LGALS8, DDX58, and MX1 in one miRNA-regulated network and integrate them into a functional node in the defense against L. pneumophila. Legionella pneumophila is an important cause of pneumonia. It invades alveolar macrophages and manipulates the immune response by interfering with signaling pathways and gene transcription to support its own replication. MicroRNAs (miRNAs) are critical posttranscriptional regulators of gene expression and are involved in defense against bacterial infections. Several pathogens have been shown to exploit the host miRNA machinery to their advantage. We therefore hypothesize that macrophage miRNAs exert positive or negative control over Legionella intracellular replication. We found significant regulation of 85 miRNAs in human macrophages upon L. pneumophila infection. Chromatin immunoprecipitation and sequencing revealed concordant changes of histone acetylation at the putative promoters. Interestingly, a trio of miRNAs (miR-125b, miR-221, and miR-579) was found to significantly affect intracellular L. pneumophila replication in a cooperative manner. Using proteome-analysis, we pinpointed this effect to a concerted downregulation of galectin-8 (LGALS8), DExD/H-box helicase 58 (DDX58), tumor protein P53 (TP53), and then MX dynamin-like GTPase 1 (MX1) by the three miRNAs. In summary, our results demonstrate a new miRNA-controlled immune network restricting Legionella replication in human macrophages.
Collapse
|
25
|
Nozawa T, Sano S, Minowa-Nozawa A, Toh H, Nakajima S, Murase K, Aikawa C, Nakagawa I. TBC1D9 regulates TBK1 activation through Ca 2+ signaling in selective autophagy. Nat Commun 2020; 11:770. [PMID: 32034138 PMCID: PMC7005872 DOI: 10.1038/s41467-020-14533-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 01/15/2020] [Indexed: 12/19/2022] Open
Abstract
Invading microbial pathogens can be eliminated selectively by xenophagy. Ubiquitin-mediated autophagy receptors are phosphorylated by TANK-binding kinase 1 (TBK1) and recruited to ubiquitinated bacteria to facilitate autophagosome formation during xenophagy, but the molecular mechanism underlying TBK1 activation in response to microbial infection is not clear. Here, we show that bacterial infection increases Ca2+ levels to activate TBK1 for xenophagy via the Ca2+-binding protein TBC1 domain family member 9 (TBC1D9). Mechanistically, the ubiquitin-binding region (UBR) and Ca2+-binding motif of TBC1D9 mediate its binding with ubiquitin-positive bacteria, and TBC1D9 knockout suppresses TBK1 activation and subsequent recruitment of the ULK1 complex. Treatment with a Ca2+ chelator impairs TBC1D9-ubiquitin interactions and TBK1 activation during xenophagy. TBC1D9 is also recruited to damaged mitochondria through its UBR and Ca2+-binding motif, and is required for TBK1 activation during mitophagy. These results indicate that TBC1D9 controls TBK1 activation during xenophagy and mitophagy through Ca2+-dependent ubiquitin-recognition.
Collapse
Affiliation(s)
- Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shunsuke Sano
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Atsuko Minowa-Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hirotaka Toh
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shintaro Nakajima
- Department of Life Science Dentistry, The Nippon Dental University, Tokyo, 102-8159, Japan.,Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, 102-8159, Japan
| | - Kazunori Murase
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Chihiro Aikawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
26
|
Jia J, Bissa B, Brecht L, Allers L, Choi SW, Gu Y, Zbinden M, Burge MR, Timmins G, Hallows K, Behrends C, Deretic V. AMPK, a Regulator of Metabolism and Autophagy, Is Activated by Lysosomal Damage via a Novel Galectin-Directed Ubiquitin Signal Transduction System. Mol Cell 2020; 77:951-969.e9. [PMID: 31995728 DOI: 10.1016/j.molcel.2019.12.028] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/27/2019] [Accepted: 12/24/2019] [Indexed: 12/29/2022]
Abstract
AMPK is a central regulator of metabolism and autophagy. Here we show how lysosomal damage activates AMPK. This occurs via a hitherto unrecognized signal transduction system whereby cytoplasmic sentinel lectins detect membrane damage leading to ubiquitination responses. Absence of Galectin 9 (Gal9) or loss of its capacity to recognize lumenal glycans exposed during lysosomal membrane damage abrogate such ubiquitination responses. Proteomic analyses with APEX2-Gal9 have revealed global changes within the Gal9 interactome during lysosomal damage. Gal9 association with lysosomal glycoproteins increases whereas interactions with a newly identified Gal9 partner, deubiquitinase USP9X, diminishes upon lysosomal injury. In response to damage, Gal9 displaces USP9X from complexes with TAK1 and promotes K63 ubiquitination of TAK1 thus activating AMPK on damaged lysosomes. This triggers autophagy and contributes to autophagic control of membrane-damaging microbe Mycobacterium tuberculosis. Thus, galectin and ubiquitin systems converge to activate AMPK and autophagy during endomembrane homeostasis.
Collapse
Affiliation(s)
- Jingyue Jia
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Bhawana Bissa
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Lukas Brecht
- Munich Cluster of Systems Neurology, Munich, Germany
| | - Lee Allers
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Seong Won Choi
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Yuexi Gu
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Mark Zbinden
- Human Metabolome Technologies America, Boston, MA, USA
| | - Mark R Burge
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Graham Timmins
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; School pf Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Kenneth Hallows
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Vojo Deretic
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| |
Collapse
|
27
|
Li FY, Wang SF, Bernardes ES, Liu FT. Galectins in Host Defense Against Microbial Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:141-167. [DOI: 10.1007/978-981-15-1580-4_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Group A Streptococcus Induces LAPosomes via SLO/β1 Integrin/NOX2/ROS Pathway in Endothelial Cells That Are Ineffective in Bacterial Killing and Suppress Xenophagy. mBio 2019; 10:mBio.02148-19. [PMID: 31575768 PMCID: PMC6775456 DOI: 10.1128/mbio.02148-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Our previous reports showed that the LC3-associated GAS-containing single membrane vacuoles are inefficient for bacterial clearance in endothelial cells, which may result in bacteremia. However, the characteristics and the induction mechanisms of these LC3-positive vacuoles are still largely unknown. Here we provide the first evidence that these LC3-positive GAS-containing single membrane compartments appear to be LAPosomes, which are induced by NOX2 and ROS. Through NOX2- and ROS-mediated signaling, GAS preferentially induces LAP and inhibits bacteriostatic xenophagy in endothelial cells. We also provide the first demonstration that β1 integrin acts as the receptor for LAP induction through GAS-produced SLO stimulation in endothelial cells. Our findings reveal the underlying mechanisms of LAP induction and autophagy evasion for GAS multiplication in endothelial cells. Group A streptococcus (GAS) is an important human pathogen which can cause fatal diseases after invasion into the bloodstream. Although antibiotics and immune surveillance are the main defenses against GAS infection, GAS utilizes internalization into cells as a major immune evasion strategy. Our previous findings revealed that light chain 3 (LC3)-associated single membrane GAS-containing vacuoles in endothelial cells are compromised for bacterial clearance due to insufficient acidification after fusion with lysosomes. However, the characteristics and the activation mechanisms of these LC3-positive compartments are still largely unknown. In the present study, we demonstrated that the LC3-positive GAS is surrounded by single membrane and colocalizes with NADPH oxidase 2 (NOX2) complex but without ULK1, which are characteristics of LC3-associated phagocytosis (LAP). Inhibition of NOX2 or reactive oxygen species (ROS) significantly reduces GAS multiplication and enhances autolysosome acidification in endothelial cells through converting LAP to conventional xenophagy, which is revealed by enhancement of ULK1 recruitment, attenuation of p70s6k phosphorylation, and formation of the isolation membrane. We also clarify that the inactivation of mTORC1, which is the initiation signal of autophagy, is inhibited by NOX2- and ROS-activated phosphatidylinositol 3-kinase (PI3K)/AKT and MEK/extracellular signal-regulated kinase (ERK) pathways. In addition, streptolysin O (SLO) of GAS is identified as a crucial inducer of ROS for β1 integrin-mediated LAP induction. After downregulation of β1 integrin, GAS multiplication is reduced, accompanied with LAP inhibition and xenophagy induction. These results demonstrate that GAS infection preferentially induces ineffective LAP to evade xenophagic killing in endothelial cells through the SLO/β1 integrin/NOX2/ROS pathway.
Collapse
|
29
|
Tribulatti MV, Carabelli J, Prato CA, Campetella O. Galectin-8 in the onset of the immune response and inflammation. Glycobiology 2019; 30:134-142. [DOI: 10.1093/glycob/cwz077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Galectins (Gals), a family of mammalian lectins, have emerged as key regulators of the immune response, being implicated in several physiologic and pathologic conditions. Lately, there is increasing data regarding the participation of Galectin-8 (Gal-8) in both the adaptive and innate immune responses, as well as its high expression in inflammatory disorders. Here, we focus on the pro- and anti-inflammatory properties of Gal-8 and discuss the potential use of this lectin in order to shape the immune response, according to the context.
Collapse
Affiliation(s)
- María V Tribulatti
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Campus Miguelete, Avenida 25 de Mayo y Francia, San Martín, Buenos Aires B1650HMP, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Julieta Carabelli
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Campus Miguelete, Avenida 25 de Mayo y Francia, San Martín, Buenos Aires B1650HMP, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Cecilia A Prato
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Campus Miguelete, Avenida 25 de Mayo y Francia, San Martín, Buenos Aires B1650HMP, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Oscar Campetella
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Campus Miguelete, Avenida 25 de Mayo y Francia, San Martín, Buenos Aires B1650HMP, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| |
Collapse
|
30
|
Pancholi V. Group A Streptococcus-Mediated Host Cell Signaling. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0021-2018. [PMID: 30767846 PMCID: PMC11590744 DOI: 10.1128/microbiolspec.gpp3-0021-2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Indexed: 12/14/2022] Open
Abstract
In the past decade, the field of the cellular microbiology of group A Streptococcus (S. pyogenes) infection has made tremendous advances and touched upon several important aspects of pathogenesis, including receptor biology, invasive and evasive phenomena, inflammasome activation, strain-specific autophagic bacterial killing, and virulence factor-mediated programmed cell death. The noteworthy aspect of S. pyogenes-mediated cell signaling is the recognition of the role of M protein in a variety of signaling events, starting with the targeting of specific receptors on the cell surface and on through the induction and evasion of NETosis, inflammasome, and autophagy/xenophagy to pyroptosis and apoptosis. Variations in reports on S. pyogenes-mediated signaling events highlight the complex mechanism of pathogenesis and underscore the importance of the host cell and S. pyogenes strain specificity, as well as in vitro/in vivo experimental parameters. The severity of S. pyogenes infection is, therefore, dependent on the virulence gene expression repertoire in the host environment and on host-specific dynamic signaling events in response to infection. Commonly known as an extracellular pathogen, S. pyogenes finds host macrophages as safe havens wherein it survives and even multiplies. The fact that endothelial cells are inherently deficient in autophagic machinery compared to epithelial cells and macrophages underscores the invasive nature of S. pyogenes and its ability to cause severe systemic diseases. S. pyogenes is still one of the top 10 causes of infectious mortality. Understanding the orchestration of dynamic host signaling networks will provide a better understanding of the increasingly complex mechanism of S. pyogenes diseases and novel ways of therapeutically intervening to thwart severe and often fatal infections.
Collapse
Affiliation(s)
- Vijay Pancholi
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210
| |
Collapse
|
31
|
Mycobacterium tuberculosis Type VII Secretion System Effectors Differentially Impact the ESCRT Endomembrane Damage Response. mBio 2018; 9:mBio.01765-18. [PMID: 30482832 PMCID: PMC6282207 DOI: 10.1128/mbio.01765-18] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Intracellular pathogens have varied strategies to breach the endolysosomal barrier so that they can deliver effectors to the host cytosol, access nutrients, replicate in the cytoplasm, and avoid degradation in the lysosome. In the case of Mycobacterium tuberculosis, the bacterium perforates the phagosomal membrane shortly after being taken up by macrophages. Phagosomal damage depends upon the mycobacterial ESX-1 type VII secretion system (T7SS). Sterile insults, such as silica crystals or membranolytic peptides, can also disrupt phagosomal and endolysosomal membranes. Recent work revealed that the host endosomal sorting complex required for transport (ESCRT) machinery rapidly responds to sterile endolysosomal damage and promotes membrane repair. We hypothesized that ESCRTs might also respond to pathogen-induced phagosomal damage and that M. tuberculosis could impair this host response. Indeed, we found that ESCRT-III proteins were recruited to M. tuberculosis phagosomes in an ESX-1-dependent manner. We previously demonstrated that the mycobacterial effectors EsxG/TB9.8 and EsxH/TB10.4, both secreted by the ESX-3 T7SS, can inhibit ESCRT-dependent trafficking of receptors to the lysosome. Here, we additionally show that ESCRT-III recruitment to sites of endolysosomal damage is antagonized by EsxG and EsxH, both within the context of M. tuberculosis infection and sterile injury. Moreover, EsxG and EsxH themselves respond within minutes to membrane damage in a manner that is independent of calcium and ESCRT-III recruitment. Thus, our study reveals that T7SS effectors and ESCRT participate in a series of measures and countermeasures for control of phagosome integrity.IMPORTANCE Mycobacterium tuberculosis causes tuberculosis, which kills more people than any other infection. M. tuberculosis grows in macrophages, cells that specialize in engulfing and degrading microorganisms. Like many intracellular pathogens, in order to cause disease, M. tuberculosis damages the membrane-bound compartment (phagosome) in which it is enclosed after macrophage uptake. Recent work showed that when chemicals damage this type of intracellular compartment, cells rapidly detect and repair the damage, using machinery called the endosomal sorting complex required for transport (ESCRT). Therefore, we hypothesized that ESCRT might also respond to pathogen-induced damage. At the same time, our previous work showed that the EsxG-EsxH heterodimer of M. tuberculosis can inhibit ESCRT, raising the possibility that M. tuberculosis impairs this host response. Here, we show that ESCRT is recruited to damaged M. tuberculosis phagosomes and that EsxG-EsxH undermines ESCRT-mediated endomembrane repair. Thus, our studies demonstrate a battle between host and pathogen over endomembrane integrity.
Collapse
|
32
|
Casals C, Campanero-Rhodes MA, García-Fojeda B, Solís D. The Role of Collectins and Galectins in Lung Innate Immune Defense. Front Immunol 2018; 9:1998. [PMID: 30233589 PMCID: PMC6131309 DOI: 10.3389/fimmu.2018.01998] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022] Open
Abstract
Different families of endogenous lectins use complementary defense strategies against pathogens. They may recognize non-self glycans typically found on pathogens and/or host glycans. The collectin and galectin families are prominent examples of these two lectin categories. Collectins are C-type lectins that contain a carbohydrate recognition domain and a collagen-like domain. Members of this group include surfactant protein A (SP-A) and D (SP-D), secreted by the alveolar epithelium to the alveolar fluid. Lung collectins bind to several microorganisms, which results in pathogen aggregation and/or killing, and enhances phagocytosis of pathogens by alveolar macrophages. Moreover, SP-A and SP-D influence macrophage responses, contributing to resolution of inflammation, and SP-A is essential for tissue-repair functions of macrophages. Galectins also function by interacting directly with pathogens or by modulating the immune system in response to the infection. Direct binding may result in enhanced or impaired infection of target cells, or can have microbicidal effects. Immunomodulatory effects of galectins include recruitment of immune cells to the site of infection, promotion of neutrophil function, and stimulation of the bactericidal activity of infected macrophages. Moreover, intracellular galectins can serve as danger receptors, promoting autophagy of the invading pathogen. This review will focus on the role of collectins and galectins in pathogen clearance and immune response activation in infectious diseases of the respiratory system.
Collapse
Affiliation(s)
- Cristina Casals
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
| | - María A Campanero-Rhodes
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| | - Belén García-Fojeda
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
| | - Dolores Solís
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| |
Collapse
|
33
|
Hong MH, Weng IC, Liu FT. Galectins as Intracellular Regulators of Cellular Responses through the Detection of Damaged Endocytic Vesicles. TRENDS GLYCOSCI GLYC 2018. [DOI: 10.4052/tigg.1733.1se] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
| | - I-Chun Weng
- Institute of Biomedical Sciences, Academia Sinica
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica
| |
Collapse
|
34
|
Abstract
Galectins are carbohydrate-binding proteins that are involved in many physiological functions, such as inflammation, immune responses, cell migration, autophagy and signalling. They are also linked to diseases such as fibrosis, cancer and heart disease. How such a small family of only 15 members can have such widespread effects remains a conundrum. In this Cell Science at a Glance article, we summarise recent literature on the many cellular activities that have been ascribed to galectins. As shown on the accompanying poster, these include carbohydrate-independent interactions with cytosolic or nuclear targets and carbohydrate-dependent interactions with extracellular glycoconjugates. We discuss how these intra- and extracellular activities might be linked and point out the importance of unravelling molecular mechanisms of galectin function to gain a true understanding of their contributions to the physiology of the cell. We close with a short outlook on the organismal functions of galectins and a perspective on the major challenges in the field.
Collapse
Affiliation(s)
- Ludger Johannes
- Institut Curie, PSL Research University, Cellular and Chemical Biology unit, U1143 INSERM, UMR3666 CNRS, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Ralf Jacob
- Philipps-Universität Marburg, Institut für Zytobiologie, Robert-Koch-Str. 6, 35037 Marburg, Germany
| | - Hakon Leffler
- Sect. MIG (Microbiology, Immunology, Glycobiology), Dept Laboratory Medicine, Lund University, POB 117, 22100 Lund, Sweden
| |
Collapse
|