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
|
Nenasheva VV, Stepanenko EA, Tarantul VZ. Multi-Directional Mechanisms of Participation of the TRIM Gene Family in Response of Innate Immune System to Bacterial Infections. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1283-1299. [PMID: 39218025 DOI: 10.1134/s0006297924070101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/30/2024] [Accepted: 06/08/2024] [Indexed: 09/04/2024]
Abstract
The multigene TRIM family is an important component of the innate immune system. For a long time, the main function of the genes belonging to this family was believed to be an antiviral defense of the host organism. The issue of their participation in the immune system response to bacterial invasion has been less studied. This review is the first comprehensive analysis of the mechanisms of functioning of the TRIM family genes in response to bacterial infections, which expands our knowledge about the role of TRIM in the innate immune system. When infected with different types of bacteria, individual TRIM proteins regulate inflammatory, interferon, and other responses of the immune system in the cells, and also affect autophagy and apoptosis. Functioning of TRIM proteins in response to bacterial infection, as well as viral infection, often includes ubiquitination and various protein-protein interactions with both bacterial proteins and host cell proteins. At the same time, some TRIM proteins, on the contrary, contribute to the infection development. Different members of the TRIM family possess similar mechanisms of response to viral and bacterial infection, and the final impact of these proteins could vary significantly. New data on the effect of TRIM proteins on bacterial infections make an important contribution to a more detailed understanding of the innate immune system functioning in animals and humans when interacting with pathogens. This data could also be used for the search of new targets for antibacterial defense.
Collapse
|
3
|
Fu Y, Li J, Cai W, Huang Y, Liu X, Ma Z, Tang Z, Bian X, Zheng J, Jiang J, Li C. The emerging tumor microbe microenvironment: From delineation to multidisciplinary approach-based interventions. Acta Pharm Sin B 2024; 14:1560-1591. [PMID: 38572104 PMCID: PMC10985043 DOI: 10.1016/j.apsb.2023.11.018] [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: 08/28/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024] Open
Abstract
Intratumoral microbiota has become research hotspots, and emerges as a non-negligent new component of tumor microenvironments (TME), due to its powerful influence on tumor initiation, metastasis, immunosurveillance and prognosis despite in low-biomass. The accumulations of microbes, and their related components and metabolites within tumor tissues, endow TME with additional pluralistic features which are distinct from the conventional one. Therefore, it's definitely necessary to comprehensively delineate the sophisticated landscapes of tumor microbe microenvironment, as well as their functions and related underlying mechanisms. Herein, in this review, we focused on the fields of tumor microbe microenvironment, including the heterogeneity of intratumor microbiota in different types of tumors, the controversial roles of intratumoral microbiota, the basic features of tumor microbe microenvironment (i.e., pathogen-associated molecular patterns (PAMPs), typical microbial metabolites, autophagy, inflammation, multi-faceted immunomodulation and chemoresistance), as well as the multidisciplinary approach-based intervention of tumor microbiome for cancer therapy by applying wild-type or engineered live microbes, microbiota metabolites, antibiotics, synthetic biology and rationally designed biomaterials. We hope our work will provide valuable insight to deeply understand the interplay of cancer-immune-microbial, and facilitate the development of microbes-based tumor-specific treatments.
Collapse
Affiliation(s)
- Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jia Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Wenyun Cai
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongyi Ma
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
4
|
Aguilera MO, Delgui LR, Reggiori F, Romano PS, Colombo MI. Autophagy as an innate immunity response against pathogens: a Tango dance. FEBS Lett 2024; 598:140-166. [PMID: 38101809 DOI: 10.1002/1873-3468.14788] [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: 08/23/2023] [Revised: 10/18/2023] [Accepted: 10/27/2023] [Indexed: 12/17/2023]
Abstract
Intracellular infections as well as changes in the cell nutritional environment are main events that trigger cellular stress responses. One crucial cell response to stress conditions is autophagy. During the last 30 years, several scenarios involving autophagy induction or inhibition over the course of an intracellular invasion by pathogens have been uncovered. In this review, we will present how this knowledge was gained by studying different microorganisms. We intend to discuss how the cell, via autophagy, tries to repel these attacks with the objective of destroying the intruder, but also how some pathogens have developed strategies to subvert this. These two fates can be compared with a Tango, a dance originated in Buenos Aires, Argentina, in which the partner dancers are in close connection. One of them is the leader, embracing and involving the partner, but the follower may respond escaping from the leader. This joint dance is indeed highly synchronized and controlled, perfectly reflecting the interaction between autophagy and microorganism.
Collapse
Affiliation(s)
- Milton O Aguilera
- Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia-Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Facultad de Odontología, Microbiología, Parasitología e Inmunología, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Laura R Delgui
- Instituto de Histología y Embriología de Mendoza, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Denmark
| | - Patricia S Romano
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología de Mendoza, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
- Facultad de Ciencias Médicas, Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| | - María I Colombo
- Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia-Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| |
Collapse
|
5
|
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
|
6
|
Tang B, Lu X, Tong Y, Feng Y, Mao Y, Dun G, Li J, Xu Q, Tang J, Zhang T, Deng L, He X, Lan Y, Luo H, Zeng L, Xiang Y, Li Q, Zeng D, Mao X. MicroRNA-31 induced by Fusobacterium nucleatum infection promotes colorectal cancer tumorigenesis. iScience 2023; 26:106770. [PMID: 37216106 PMCID: PMC10196571 DOI: 10.1016/j.isci.2023.106770] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/27/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Persistent Fusobacterium nucleatum infection is associated with the development of human colorectal cancer (CRC) and promotes tumorigenicity, but the underlying mechanisms remain unclear. Here, we reported that F. nucleatum promoted the tumorigenicity of CRC, which was associated with F. nucleatum-induced microRNA-31 (miR-31) expression in CRC tissues and cells. F. nucleatum infection inhibited autophagic flux by miR-31 through inhibiting syntaxin-12 (STX12) and was associated with the increased intracellular survival of F. nucleatum. Overexpression of miR-31 in CRC cells promoted their tumorigenicity by targeting eukaryotic initiation factor 4F-binding protein 1/2 (eIF4EBP1/2), whereas miR-31 knockout mice were resistant to the formation of colorectal tumors. In conclusion, F. nucleatum, miR-31, and STX12 form a closed loop in the autophagy pathway, and continuous F. nucleatum-induced miR-31 expression promotes the tumorigenicity of CRC cells by targeting eIF4EBP1/2. These findings reveal miR-31 as a potential diagnostic biomarker and therapeutic target in CRC patients with F. nucleatum infection.
Collapse
Affiliation(s)
- Bin Tang
- Department of Clinical Laboratory, Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin, Chongqing 402260, China
| | - Xiaoxue Lu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yanan Tong
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuyang Feng
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yilan Mao
- Class of 2021 undergraduate, Nursing College of Chongqing Medical University, Chongqing 400016, China
| | - Guodong Dun
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jing Li
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Qiaolin Xu
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Jie Tang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Tao Zhang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ling Deng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xiaoyi He
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuanzhi Lan
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Huaxing Luo
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Linghai Zeng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuanyuan Xiang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Dongzhu Zeng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xuhu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
7
|
Yuan W, Zhan X, Liu W, Ma R, Zhou Y, Xu G, Ge Z. Mmu-miR-25-3p promotes macrophage autophagy by targeting DUSP10 to reduce mycobacteria survival. Front Cell Infect Microbiol 2023; 13:1120570. [PMID: 37256106 PMCID: PMC10225524 DOI: 10.3389/fcimb.2023.1120570] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Background The present study aimed to investigate the regulation of miR-25-3p on macrophage autophagy and its effect on macrophage clearance of intracellular Mycobacterium bovis Bacillus Calmette-Guerin (BCG) retention based on the previous findings on the differential expression of exosomal miRNA in macrophages infected with BCG. Methods Through enrichment analysis and Hub gene analysis, key differentially expressed miRNA and its target genes were selected. The targeted binding ability of the screened mmu-miR-25-3p and its predicted target gene DUSP10 was determined through the TargetScan database, and this was further verified by dual luciferase reporter gene assay. mmu-miR-25-3p mimics, mmu-miR-25-3p inhibitor, si-DUSP10, miR-NC,si-NC and PD98059 (ERK Inhibitor) were used to intervene macrophages Raw264.7. Rt-qPCR was used to detect the expression levels of mmu-miR-25-3p and DUSP10 mRNA. Western blot was used to detect the expression levels of DUSP10, LC3-II, p-ERK1/2, beclin1, Atg5 and Atg7. The autophagy flux of macrophage Raw264.7 in each group was observed by confocal laser microscopy, and the expression distribution of DUSP10 and the structure of autophagosomes were observed by transmission electron microscopy. Finally, the intracellular BCG load of macrophage Raw264.7 was evaluated by colony-forming unit (CFU) assay. Results Bioinformatics analysis filtered and identified the differentially expressed exosomal miRNAs. As a result, mmu-miR-25-3p expression was significantly increased, and dual specificity phosphatase 10 (DUSP10) was predicted as its target gene that was predominantly involved in autophagy regulation. The dual luciferase reporter gene activity assay showed that mmu-miR-25-3p was targeted to the 3'-untranslated region (UTR) of DUSP10. The infection of BCG induced the upregulation of mmu-miR-25-3p and downregulation of DUSP10 in RAW264.7 cells, which further increased the expression of LC3-II and promoted autophagy. Upregulated mmu-miR-25-3p expression decreased the level of DUSP10 and enhanced the phosphorylation of ERK1/2, which in turn upregulated the expression of LC3-II, Atg5, Atg7, and Beclin1. Immuno-electron microscopy, transmission electron microscopy, and autophagic flux analysis further confirmed that the upregulation of mmu-miR-25-3p promotes the autophagy of macrophages after BCG infection. The CFU number indicated that upregulated mmu-miR-25-3p expression decreased the mycobacterial load and accelerated residual mycobacteria clearance. Conclusion mmu-miR-25-3p promotes the phosphorylation of ERK1/2 by inhibiting the expression of DUSP10, thus enhancing the BCG-induced autophagy of macrophages. These phenomena reduce the bacterial load of intracellular Mycobacterium and facilitate the clearance of residual mycobacteria. mmu-miR-25-3p has great potential as a target for anti-tuberculosis immunotherapy and can be the optimal miRNA loaded into exosomal drug delivery system in future studies.
Collapse
Affiliation(s)
- Wenqi Yuan
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xuehua Zhan
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wei Liu
- Clinical Medicine School, Ningxia Medical University, Yinchuan, China
| | - Rong Ma
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yueyong Zhou
- Clinical Medicine School, Ningxia Medical University, Yinchuan, China
| | - Guangxian Xu
- The First Dongguan Affiliated Hospital, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Zhaohui Ge
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
8
|
Lattos A, Feidantsis K, Giantsis IA, Theodorou JA, Michaelidis B. Seasonality in Synergism with Multi-Pathogen Presence Leads to Mass Mortalities of the Highly Endangered Pinna nobilis in Greek Coastlines: A Pathophysiological Approach. Microorganisms 2023; 11:1117. [PMID: 37317091 DOI: 10.3390/microorganisms11051117] [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: 03/30/2023] [Revised: 04/22/2023] [Accepted: 04/23/2023] [Indexed: 06/16/2023] Open
Abstract
Mortalities of Pinna nobilis populations set at risk the survival of the species from many Mediterranean coastline habitats. In many cases, both Haplosporidium pinnae and Mycobacterium spp. are implicated in mass mortalities of P. nobilis populations, leading the species into extinction. In the context of the importance of these pathogens' role in P. nobilis mortalities, the present study investigated two Greek populations of the species hosting different microbial loads (one only H. pinnae and the second both pathogens) by the means of pathophysiological markers. More specifically, the populations from Kalloni Gulf (Lesvos Island) and from Maliakos Gulf (Fthiotis), seasonally sampled, were chosen based on the host pathogens in order to investigate physiological and immunological biomarkers to assess those pathogens' roles. In order to determine if the haplosporidian parasite possesses a major role in the mortalities or if both pathogens are involved in these phenomena, a variety of biomarkers, including apoptosis, autophagy, inflammation and heat shock response were applied. The results indicated a decreased physiological performance of individuals hosting both pathogens in comparison with those hosting only H. pinnae. Our findings provide evidence for the synergistic role of those pathogens in the mortality events, which is also enhanced by the influence of seasonality.
Collapse
Affiliation(s)
- Athanasios Lattos
- Laboratory of Animal Physiology, Department of Zoology, Faculty of Science, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Konstantinos Feidantsis
- Laboratory of Animal Physiology, Department of Zoology, Faculty of Science, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Ioannis A Giantsis
- Department of Animal Science, Faculty of Agricultural Sciences, University of Western Macedonia, GR-53100 Florina, Greece
| | - John A Theodorou
- Department of Fisheries & Aquaculture, University of Patras, GR-23200 Mesolonghi, Greece
| | - Basile Michaelidis
- Laboratory of Animal Physiology, Department of Zoology, Faculty of Science, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| |
Collapse
|
9
|
Wang B, Zhou C, Wu Q, Lin P, Pu Q, Qin S, Gao P, Wang Z, Liu Y, Arel J, Chen Y, Chen T, Wu M. cGAS modulates cytokine secretion and bacterial burdens by altering the release of mitochondrial DNA in Pseudomonas pulmonary infection. Immunology 2022; 166:408-423. [PMID: 35420160 DOI: 10.1111/imm.13482] [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] [Received: 07/26/2021] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 11/28/2022] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) is essential for fighting against viruses and bacteria, but how cGAS is involved in host immune response remains largely elusive. Here, we uncover the crucial role of cGAS in host immunity based on a Pseudomonas aeruginosa pulmonary infection model. cGAS-/- mice showed more heavy bacterial burdens and serious lung injury accompanied with exorbitant proinflammatory cytokines than wild-type mice. cGAS deficiency caused an accumulation of mitochondrial DNA in cytoplasm, which in turn induced excessive secretion of proinflammatory factors by activating inflammasome and TLR9 signaling. Mechanistically, cGAS deficiency inhibited the recruitment of LC3 by reducing the binding capacity of TBK-1 to p62, leading to impaired mitophagy and augmented release of mitochondrial DNA. Importantly, cytoplasmic mitochondrial DNA also acted as a feedback signal that induced the activation of cGAS. Altogether, these findings identify protective and homeostasis functions of cGAS against Pseudomonas aeruginosa infection, adding significant insight into the pathogenesis of bacterial infectious diseases.
Collapse
Affiliation(s)
- Biao Wang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, P. R. China.,Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Chuanmin Zhou
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA.,Wuhan University School of Health Sciences, Wuhan, Hubei Province, P. R. China
| | - Qun Wu
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Ping Lin
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Qinqin Pu
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Shugang Qin
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Pan Gao
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Zhihan Wang
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Yingying Liu
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Jacob Arel
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Yanjiong Chen
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, P. R. China
| | - Teng Chen
- Forensic Medicine College of Xi'an Jiaotong University, Key Laboratory of the Health Ministry for Forensic Medicine, Xi'an, P. R. China
| | - Min Wu
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| |
Collapse
|
10
|
Ramser A, Greene E, Alrubaye AA, Wideman R, Dridi S. Role of Autophagy Machinery Dysregulation in Bacterial Chondronecrosis with Osteomyelitis (BCO). Poult Sci 2022; 101:101750. [PMID: 35278754 PMCID: PMC8914211 DOI: 10.1016/j.psj.2022.101750] [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] [Received: 07/15/2021] [Revised: 12/27/2021] [Accepted: 01/23/2022] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a cell survival and homeostasis mechanism involving lysosomal degradation of cellular components and foreign bodies. It plays a role in bone homeostasis, skeletal diseases, and bacterial infections as both a cell-survival or cell-death pathway. This study sought to determine if autophagy played a role in bacterial chondronecrosis with osteomyelitis (BCO). BCO is a prominent cause of lameness in modern broilers and results from bacterial infection of mechanically stressed leg bone growth plates. The protein and gene expression of key autophagy machinery was analyzed in both normal and BCO-affected broilers using real-time qPCR and immunoblot, respectively. Gene expression showed a significant downregulation of key target signatures involved in every stage of autophagy in BCO-affected bone, such as ATG13, SQSTM1 (p62), ATG9B, ATG16L, ATG12, LC3C, and RAB7A. Additionally, protein expression for LC3 was also significantly lower in BCO. An in vitro study using human fetal osteoblast cells challenged with BCO isolate, Staphylococcus agnetis 908, showed a similar dysregulation of autophagy machinery along with a significant decrease in cell viability. When autophagy was inhibited via 3-methyladenine or chloroquine, comparable decreases in cell viability were seen along with dysregulation of autophagy machinery. Together, these results are the first to implicate autophagy machinery dysregulation in the pathology of BCO.
Collapse
|
11
|
Cis-9, Trans-11 CLA Alleviates Lipopolysaccharide-Induced Depression of Fatty Acid Synthesis by Inhibiting Oxidative Stress and Autophagy in Bovine Mammary Epithelial Cells. Antioxidants (Basel) 2021; 11:antiox11010055. [PMID: 35052560 PMCID: PMC8773093 DOI: 10.3390/antiox11010055] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
Lipopolysaccharide (LPS) is the dominating endotoxin of Gram-negative bacteria, which can cause mastitis. Bovine mammary epithelial cells (BMECs), as major components of the mammary gland, usually suffer LPS challenge. Cis-9, trans-11 conjugated linoleic acid (CLA) has been reported to have anti-inflammatory characteristics, while its anti-oxidative ability to maintain cellular homeostasis in BMECs under LPS challenge is limited. Therefore, we studied whether cis-9, trans-11 CLA can restore the disturbance of cellular homeostasis indicated by the redox status and autophagy level caused by LPS and have an effect on cellular function- milk fat metabolism. For oxidative stress, LPS challenge promoted the formation of reactive oxygen species (ROS) and thiobarbituric acid reactive substances (TBARS) and decreased the concentration of glutathione. Anti-oxidative signaling regulated by transcription factor nuclear factor, erythroid 2 like 2 (Nrf2) was also depressed by LPS at the mRNA and protein level. However, cis-9, trans-11 CLA pretreatment downregulated the formation of ROS and TBARS and upregulated the expression of antioxidative enzymes. As a part of innate immunity, autophagy was also motivated by LPS challenge, while CLA decreased the autophagy level. LPS and H2O2 inhibited milk fat synthesis-related transcription factor sterol regulatory element binding protein (SREBP1), peroxisome proliferator activated receptor gamma (PPARG) and their downstream enzymes. Furthermore, 50 uM cis-9, trans-11 CLA promoted the mRNA and protein abundance of milk fat synthesis-related genes and lipid droplet formation in BMECs. In conclusion, LPS challenge disturbed the cellular homeostasis and depressed milk fat synthesis in BMECs; while cis-9, trans-11 CLA alleviated oxidative stress and decreased autophagy level, thus promoting milk fat synthesis, which offers a natural therapeutic strategy for mastitis.
Collapse
|
12
|
Abstract
Nutritional symbionts are restricted to specialized host cells called bacteriocytes in various insect orders. These symbionts can provide essential nutrients to the host. However, the cellular mechanisms underlying the regulation of these insect-symbiont metabolic associations remain largely unclear. The whitefly, Bemisia tabaci MEAM1, hosts Portiera and Hamiltonella bacteria in the same bacteriocyte. In this study, the induction of autophagy by chemical treatment and gene silencing decreased symbiont titers, and essential amino acid (EAA) and B vitamin contents. In contrast, the repression of autophagy in bacteriocytes via Atg8 silencing increased symbiont titers, and amino acid and B vitamin contents. Furthermore, dietary supplementation with non-EAAs or B vitamins alleviated autophagy in whitefly bacteriocytes, elevated TOR (target of rapamycin) expression and increased symbiont titers. TOR silencing restored symbiont titers in whiteflies after dietary supplementation with B vitamins. These data suggest that Portiera and Hamiltonella evade autophagy of the whitefly bacteriocytes by activating the TOR pathway via providing essential nutrients. Taken together, we demonstrated that autophagy plays a critical role in regulating the metabolic interactions between the whitefly and two intracellular symbionts. Therefore, this study reveals that autophagy is an important cellular basis for bacteriocyte evolution and symbiosis persistence in whiteflies. The whitefly symbiosis unravels the interactions between cellular and metabolic functions of bacteriocytes. Importance Nutritional symbionts, which are restricted to specialized host cells called bacteriocytes, can provide essential nutrients for many hosts. However, the cellular mechanisms of regulation of animal-symbiont metabolic associations have been largely unexplored. Here, using the whitefly-Portiera/Hamiltonella endosymbiosis, we demonstrate autophagy regulates the symbiont titers, and thereby alters the essential amino acid and B vitamin contents. For persistence in the whitefly bacteriocytes, Portiera and Hamiltonella alleviate autophagy by activating the TOR (target of rapamycin) pathway through providing essential nutrients. Therefore, we demonstrate that autophagy plays a critical role in regulating the metabolic interactions between the whitefly and two intracellular symbionts. This study also provides insight into the cellular basis of bacteriocyte evolution and symbiosis persistence in the whitefly. The mechanisms underlying the role of autophagy in whitefly symbiosis could be widespread in many insect nutritional symbioses. These findings provide new avenue for whitefly control via regulating autophagy in the future.
Collapse
|
13
|
Malkeyeva D, Kiseleva E, Fedorova SA. Loss of Hsp67Bc leads to autolysosome enlargement in the Drosophila brain. Cell Biol Int 2021; 46:203-212. [PMID: 34719095 DOI: 10.1002/cbin.11721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/18/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Hsp67Bc is a small heat shock protein found in Drosophila melanogaster. Apart from performing a function (common for all small heat shock proteins) of preventing aggregation of misfolded proteins, it is involved in macroautophagy regulation alongside the Starvin protein. Overexpression of the D. melanogaster Hsp67Bc gene has been shown to stimulate macroautophagy in S2 cell culture. Nonetheless, it has been unknown how the absence of the Hsp67Bc gene may affect it. Here, we studied the effect of Hsp67Bc gene deletion on the macroautophagy induced by the pathogenic Wolbachia wMelPop strain in D. melanogaster. We detected Wolbachia inside autophagic vacuoles in fly neurons, thereby proving that these endosymbionts were being eliminated via macroautophagy. Nevertheless, we did not register any difference in brain bacterial load between Hsp67Bc-null and control flies at all tested stages of ontogenesis. Moreover, the abundance of autophagic vacuoles was similar between neurons of the mutant and control flies, yet the cross-sectional area of autolysosomes on ultrathin sections was more than 1.5-fold larger in Hsp67Bc-null fly brains than in the control line. Our findings suggest that the product of the Hsp67Bc gene does not participate in the initiation of endosymbiont-induced macroautophagy but may mediate autophagosome maturation: the deletion of the Hsp67Bc gene leads to the increase in autolysosome size.
Collapse
Affiliation(s)
- Dina Malkeyeva
- Cell Biology Department, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Elena Kiseleva
- Cell Biology Department, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Svetlana A Fedorova
- Cell Biology Department, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| |
Collapse
|
14
|
Inhibiting miR-129-5p alleviates inflammation and modulates autophagy by targeting ATG14 in fungal keratitis. Exp Eye Res 2021; 211:108731. [PMID: 34411602 DOI: 10.1016/j.exer.2021.108731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 07/16/2021] [Accepted: 08/13/2021] [Indexed: 01/04/2023]
Abstract
To investigate the role of miR-129-5p in inflammation and autophagy in fungal keratitis, we established a keratitis mouse model infected with Fusarium solani (F. solani) and conducted experiments on corneal stromal cells infected with F. solani. The expression of miR-129-5p was detected via quantitative real-time polymerase chain reaction (PCR). The miR-129-5p antagomir was used to transfect cells and mice to study the regulatory role of miR-129-5p in autophagy and inflammation after fungal infection. The expression of Beclin1 and LC3B and colocalization of LC3B with lysosomes were detected via Western blotting and immunofluorescence. CCK-8 was used to determine the viability of corneal stromal cells. The expression of IL-1β were detected by ELISA. Bioinformatics software was used to predict the potential targets of miR-129-5p, which were verified by a luciferase reporter gene assay. RT-PCR showed that miR-129-5p expression in mouse corneas was significantly increased after infection with F. solani. Subconjunctival injection of the miR-129-5p antagomir significantly enhanced the proteins Beclin-1 and LC3B. At the same time, inhibiting miR-129-5p expression could reduce the inflammatory response in FK and significantly increase the viability of corneal stromal cells infected with F. solan. Moreover, the dual luciferase reporter assay indicated that Atg14 was a direct target of miR-129-5p. Our study shows that miR-129-5p is a novel small molecule that regulates autophagy by targeting Atg14, indicating that it may be a proinflammatory and therapeutic target for fungal keratitis.
Collapse
|
15
|
Yokota M, Häffner N, Kassier M, Brunner M, Shambat SM, Brennecke F, Schniering J, Marques Maggio E, Distler O, Zinkernagel AS, Maurer B. Staphylococcus aureus impairs dermal fibroblast functions with deleterious effects on wound healing. FASEB J 2021; 35:e21695. [PMID: 34160101 DOI: 10.1096/fj.201902836r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/16/2021] [Accepted: 05/10/2021] [Indexed: 12/28/2022]
Abstract
Chronic wounds are a major disease burden worldwide. The breach of the epithelial barrier facilitates transition of skin commensals to invasive facultative pathogens. Therefore, we investigated the potential effects of Staphylococcus aureus (SA) on dermal fibroblasts as key cells for tissue repair. In co-culture systems combining live or heat-killed SA with dermal fibroblasts derived from the BJ-5ta cell line, healthy individuals, and patients with systemic sclerosis, we assessed tissue repair including pro-inflammatory cytokines, matrix metalloproteases (MMPs), myofibroblast functions, and host defense responses. Only live SA induced the upregulation of IL-1β/-6/-8 and MMP1/3 as co-factors of tissue degradation. Additionally, the increased cell death reduced collagen production, proliferation, migration, and contractility, prerequisite mechanisms for wound closure. Intracellular SA triggered inflammatory and type I IFN responses via intracellular dsDNA sensor molecules and MyD88 and STING signaling pathways. In conclusion, live SA affected various key tissue repair functions of dermal fibroblasts from different sources to a similar extent. Thus, SA infection of dermal fibroblasts should be taken into account for future wound management strategies.
Collapse
Affiliation(s)
- Masaya Yokota
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland.,Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Nicola Häffner
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthew Kassier
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Matthias Brunner
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Srikanth Mairpady Shambat
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Fabian Brennecke
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Janine Schniering
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Ewerton Marques Maggio
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Annelies Sophie Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Britta Maurer
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland.,Department of Rheumatology and Immunology, University Hospital Bern, University Bern, Bern, Switzerland
| |
Collapse
|
16
|
Han L, Ma Q, Yu J, Gong Z, Ma C, Xu Y, Deng G, Wu X. Autophagy plays a protective role during Pseudomonas aeruginosa-induced apoptosis via ROS-MAPK pathway. Innate Immun 2020; 26:580-591. [PMID: 32878509 PMCID: PMC7556189 DOI: 10.1177/1753425920952156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 07/18/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa infection can induce alveolar macrophage apoptosis and autophagy, which play a vital role in eliminating pathogens. These two processes are usually not independent. Recently, autophagy has been found to interact with apoptosis during pathogen infections. Nevertheless, the role of autophagy in P. aeruginosa-infected cell apoptosis is unclear. In this study, we explored the impact of P. aeruginosa infection on autophagy and apoptosis in RAW264.7 cells. The autophagy activator rapamycin was used to stimulate autophagy and explore the role of autophagy on apoptosis in P. aeruginosa-infected RAW264.7 cells. The results indicated that P. aeruginosa infection induced autophagy and apoptosis in RAW264.7 cells, and that rapamycin could suppress P. aeruginosa-induced apoptosis by regulating the expression of apoptosis-related proteins. In addition, rapamycin scavenged the cellular reactive oxygen species (ROS) and diminished p-JNK, p-ERK1/2 and p-p38 expression of MAPK pathways in RAW264.7 cells infected with P. aeruginosa. In conclusion, the promotion of autophagy decreased P. aeruginosa-induced ROS accumulation and further attenuated the apoptosis of RAW264.7 cells through MAPK pathway. These results provide novel insights into host-pathogen interactions and highlight a potential role of autophagy in eliminating P. aeruginosa.
Collapse
Affiliation(s)
- Lu Han
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Qinmei Ma
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Jialin Yu
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Zhaoqian Gong
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Chenjie Ma
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Yanan Xu
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Guangcun Deng
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Xiaoling Wu
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| |
Collapse
|
17
|
Chou YJ, Lin CC, Dzhagalov I, Chen NJ, Lin CH, Lin CC, Chen ST, Chen KH, Fu SL. Vaccine adjuvant activity of a TLR4-activating synthetic glycolipid by promoting autophagy. Sci Rep 2020; 10:8422. [PMID: 32439945 PMCID: PMC7242473 DOI: 10.1038/s41598-020-65422-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 04/20/2020] [Indexed: 11/09/2022] Open
Abstract
Toll-like receptors (TLRs) play crucial roles in host immune defenses. Recently, TLR-mediated autophagy is reported to promote immune responses via increasing antigen processing and presentation in antigen presenting cells. The present study examined whether the synthetic TLR4 activator (CCL-34) could induce autophagy to promote innate and adaptive immunity. In addition, the potential of CCL-34 as an immune adjuvant in vivo was also investigated. Our data using RAW264.7 cells and bone marrow-derived macrophages showed that CCL-34 induced autophagy through a TLR4-NF-κB pathway. The autophagy-related molecules (Nrf2, p62 and Beclin 1) were activated in RAW264.7 cells and bone marrow-derived macrophages under CCL-34 treatment. CCL-34-stimulated macrophages exhibited significant antigen-processing activity and induced the proliferation of antigen-specific CD4+T cells as well as the production of activated T cell-related cytokines, IL-2 and IFN-γ. Furthermore, CCL-34 immunization in mice induced infiltration of monocytes in the peritoneal cavity and elevation of antigen-specific IgG in the serum. CCL-34 treatment in vivo did not cause toxicity based on serum biochemical profiles. Notably, the antigen-specific responses induced by CCL-34 were attenuated by the autophagy inhibitor, 3-methyladenine. In summary, we demonstrated CCL-34 can induce autophagy to promote antigen-specific immune responses and act as an efficient adjuvant.
Collapse
Affiliation(s)
- Yi-Ju Chou
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, 11221, Taiwan
| | - Ching-Cheng Lin
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Ivan Dzhagalov
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Chao-Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Szu-Ting Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Kuo-Hsin Chen
- Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan.
| | - Shu-Ling Fu
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, 11221, Taiwan. .,Institute of Traditional Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.
| |
Collapse
|
18
|
Guan XL, Zhang BC, Sun L. Japanese flounder pol-miR-3p-2 suppresses Edwardsiella tarda infection by regulation of autophagy via p53. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103531. [PMID: 31668931 DOI: 10.1016/j.dci.2019.103531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 06/10/2023]
Abstract
MicroRNAs (miRNAs) are post-transcriptional regulators that play vital roles in diverse physiological processes including immunity. In this study, we investigated the regulatory mechanism and function of a novel Japanese flounder (Paralichthys olivaceus) miRNA, pol-miR-3p-2. pol-miR-3p-2 was responsive in expression to the infection of the bacterial pathogen Edwardsiella tarda. pol-miR-3p-2 negatively regulated the expression of p53 through interaction with the 3'UTR of p53. Overexpression of pol-miR-3p-2 promoted autophagy, resulting in augmented production of LC3-II, while knockdown of p53 increased the level of beclin, a key factor of autophagy. In vivo and in vitro studies showed that E. tarda infection induced autophagy in flounder, and pol-miR-3p-2 inhibited the infectivity of E. tarda. Together these results indicate that pol-miR-3p-2 regulates autophagy through the target gene p53, thus revealing a regulatory link between p53 and autophagy in teleost, and that pol-miR-3p-2 plays an important role in the immune defense against E. tarda.
Collapse
Affiliation(s)
- Xiao-Lu Guan
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Bao-Cun Zhang
- Department of Biomedicine and Aarhus Research Center for Innate Immunity, Aarhus University, Aarhus, Denmark
| | - Li Sun
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
19
|
Pu Q, Lin P, Wang Z, Gao P, Qin S, Cui L, Wu M. Interaction among inflammasome, autophagy and non-coding RNAs: new horizons for drug. PRECISION CLINICAL MEDICINE 2019; 2:166-182. [PMID: 31598387 PMCID: PMC6770284 DOI: 10.1093/pcmedi/pbz019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/22/2019] [Accepted: 08/25/2019] [Indexed: 02/07/2023] Open
Abstract
Autophagy and inflammasomes are shown to interact in various situations including
infectious disease, cancer, diabetes and neurodegeneration. Since multiple layers of
molecular regulators contribute to the interplay between autophagy and inflammasome
activation, the detail of such interplay remains largely unknown. Non-coding RNAs
(ncRNAs), which have been implicated in regulating an expanding list of cellular processes
including immune defense against pathogens and inflammatory response in cancer and
metabolic diseases, may join in the crosstalk between inflammasomes and autophagy in
physiological or disease conditions. In this review, we summarize the latest research on
the interlink among ncRNAs, inflammasomes and autophagy and discuss the emerging role of
these three in multiple signaling transduction pathways involved in clinical conditions.
By analyzing these intriguing interconnections, we hope to unveil the mechanism
inter-regulating these multiple processes and ultimately discover potential drug targets
for some refractory diseases.
Collapse
Affiliation(s)
- Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Lin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Zhihan Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Pan Gao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shugang Qin
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luqing Cui
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
20
|
Tan DX. Aging: An evolutionary competition between host cells and mitochondria. Med Hypotheses 2019; 127:120-128. [PMID: 31088635 DOI: 10.1016/j.mehy.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/20/2019] [Accepted: 04/11/2019] [Indexed: 12/19/2022]
Abstract
Here, a new theory of aging is proposed. This new theory is referred as the Host-Mitochondria Intracellular Innate Immune Theory of Aging (HMIIITA). The main point of this theory is that the aging is rooted from an evolutionary competition, that is, a never ending coevolutionary race between host cells and mitochondria. Mitochondria are the descendants of bacteria. The host cells will inevitably sense their bacterial origin, particularly their circular mtDNA. The host intracellular innate immune pressure (HIIIP) aims to eliminate mtDNA as more as possible while mitochondria have to adapt the HIIIP for survival. Co-evolution is required for both of them. From biological point of view, the larger, the mtDNA, the higher, the chance, it becomes the target of HIIIP. As a result, mitochondria have to reduce their mtDNA size via deletion. This process has last for 1.5-2 billion yeas and the result is that mitochondria have lost excessive 95% of their DNA. This mtDNA deletion is not associated with free radical attack but a unique trait acquired during evolution. In the postmitotic cells, the deletion is passively selected by the mitochondrial fission-fusion cycles. Eventually, the accumulation of deletion will significantly jeopardize the mitochondrial function. The dysfunctional mitochondria no longer provide sufficient ATP to support host cells' continuous demanding for growth. At this stage, the cell or the organism aging is inevitable.
Collapse
Affiliation(s)
- Dun-Xian Tan
- The Department of Cell System and Anatomy, The University of Texas, Health, San Antonio, TX 78229, USA.
| |
Collapse
|
21
|
Synergistic clearance of intracellular pathogens by hyaluronan-streptomycin micelles encapsulated with rapamycin. Carbohydr Polym 2019; 210:364-371. [PMID: 30732772 DOI: 10.1016/j.carbpol.2019.01.068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 02/02/2023]
|
22
|
Hsieh CL, Huang HM, Hsieh SY, Zheng PX, Lin YS, Chiang-Ni C, Tsai PJ, Wang SY, Liu CC, Wu JJ. NAD-Glycohydrolase Depletes Intracellular NAD + and Inhibits Acidification of Autophagosomes to Enhance Multiplication of Group A Streptococcus in Endothelial Cells. Front Microbiol 2018; 9:1733. [PMID: 30123194 PMCID: PMC6085451 DOI: 10.3389/fmicb.2018.01733] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/11/2018] [Indexed: 12/17/2022] Open
Abstract
Group A Streptococcus (GAS) is a human pathogen causing a wide spectrum of diseases, from mild pharyngitis to life-threatening necrotizing fasciitis. GAS has been shown to evade host immune killing by invading host cells. However, how GAS resists intracellular killing by endothelial cells is still unclear. In this study, we found that strains NZ131 and A20 have higher activities of NADase and intracellular multiplication than strain SF370 in human endothelial cells (HMEC-1). Moreover, nga mutants of NZ131 (SW957 and SW976) were generated to demonstrate that NADase activity is required for the intracellular growth of GAS in endothelial cells. We also found that intracellular levels of NAD+ and the NAD+/NADH ratio of NZ131-infected HMEC-1 cells were both lower than in cells infected by the nga mutant. Although both NZ131 and its nga mutant were trapped by LC3-positive vacuoles, only nga mutant vacuoles were highly co-localized with acidified lysosomes. On the other hand, intracellular multiplication of the nga mutant was increased by bafilomycin A1 treatment. These results indicate that NADase causes intracellular NAD+ imbalance and impairs acidification of autophagosomes to escape autophagocytic killing and enhance multiplication of GAS in endothelial cells.
Collapse
Affiliation(s)
- Cheng-Lu Hsieh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsuan-Min Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Ying Hsieh
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Xing Zheng
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Yee-Shin Lin
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chuan Chiang-Ni
- Department of Microbiology & Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Ying Wang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Chuan Liu
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jiunn-Jong Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
23
|
Siqueira MDS, Ribeiro RDM, Travassos LH. Autophagy and Its Interaction With Intracellular Bacterial Pathogens. Front Immunol 2018; 9:935. [PMID: 29875765 PMCID: PMC5974045 DOI: 10.3389/fimmu.2018.00935] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022] Open
Abstract
Cellular responses to stress can be defined by the overwhelming number of changes that cells go through upon contact with and stressful conditions such as infection and modifications in nutritional status. One of the main cellular responses to stress is autophagy. Much progress has been made in the understanding of the mechanisms involved in the induction of autophagy during infection by intracellular bacteria. This review aims to discuss recent findings on the role of autophagy as a cellular response to intracellular bacterial pathogens such as, Streptococcus pyogenes, Mycobacterium tuberculosis, Shigella flexneri, Salmonella typhimurium, Listeria monocytogenes, and Legionella pneumophila, how the autophagic machinery senses these bacteria directly or indirectly (through the detection of bacteria-induced nutritional stress), and how some of these bacterial pathogens manage to escape from autophagy.
Collapse
Affiliation(s)
- Mariana da Silva Siqueira
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato de Moraes Ribeiro
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo H Travassos
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Palmela C, Chevarin C, Xu Z, Torres J, Sevrin G, Hirten R, Barnich N, Ng SC, Colombel JF. Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut 2018; 67:574-587. [PMID: 29141957 DOI: 10.1136/gutjnl-2017-314903] [Citation(s) in RCA: 338] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/20/2017] [Accepted: 10/28/2017] [Indexed: 02/06/2023]
Abstract
Intestinal microbiome dysbiosis has been consistently described in patients with IBD. In the last decades, Escherichia coli, and the adherent-invasive E coli (AIEC) pathotype in particular, has been implicated in the pathogenesis of IBD. Since the discovery of AIEC, two decades ago, progress has been made in unravelling these bacteria characteristics and its interaction with the gut immune system. The mechanisms of adhesion of AIEC to intestinal epithelial cells (via FimH and cell adhesion molecule 6) and its ability to escape autophagy when inside macrophages are reviewed here. We also explore the existing data on the prevalence of AIEC in patients with Crohn's disease and UC, and the association between the presence of AIEC and disease location, activity and postoperative recurrence. Finally, we highlight potential therapeutic strategies targeting AIEC colonisation of gut mucosa, including the use of phage therapy, bacteriocins and antiadhesive molecules. These strategies may open new avenues for the prevention and treatment of IBD in the future.
Collapse
Affiliation(s)
- Carolina Palmela
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Caroline Chevarin
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Zhilu Xu
- Department of Medicine and Therapeutics, Institute of Digestive Diseases, LKS Institute of Health Science, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Joana Torres
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Gwladys Sevrin
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Robert Hirten
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Nicolas Barnich
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Siew C Ng
- Department of Medicine and Therapeutics, Institute of Digestive Diseases, LKS Institute of Health Science, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
25
|
Zhang C, Yang L, Zhao N, Zhao Y, Shi C. Insights into Macrophage Autophagy in Latent Tuberculosis Infection: Role of Heat Shock Protein 16.3. DNA Cell Biol 2018; 37:442-448. [PMID: 29461881 DOI: 10.1089/dna.2017.4066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis (TB) is a major bacterial infectious disease worldwide that is predominantly caused by Mycobacterium tuberculosis (Mtb). The comorbidity of multiple drug-resistant TB strains with HIV and diabetes is widespread. In the presence of these diseases, host immunity is weakened, allowing the recovery of dormant bacilli and leading to recurrent TB infection. As an important component of the host innate and adaptive immune responses, macrophage autophagy plays a significant role in protecting the host against TB. However, dormant bacilli can escape from autophagosomes and/or suppress autophagy, thus surviving within the host for an extended period of time, although the underlying mechanism remains elusive. Heat shock protein 16.3 (Hsp16.3, HspX, Rv2031c, and Acr) is one of the immunodominant proteins expressed during latent TB infection (LTBI). It may help maintain the protein stability and long-term viability of Mtb by inhibiting macrophage autophagy, resulting in LBTI. In this review, we discuss how dormant bacilli escape from autophagosomes, and we focus on the role of Hsp16.3 in regulating macrophage autophagy in LTBI so as to provide a firm basis for further studies. Hsp16.3 may represent a potential biomarker of LTBI and novel pharmacological target for anti-tubercular drugs.
Collapse
Affiliation(s)
- Caiqin Zhang
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Li Yang
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Ningning Zhao
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Yong Zhao
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Changhong Shi
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| |
Collapse
|
26
|
Sunahara S, Watanabe E, Hatano M, Swanson PE, Oami T, Fujimura L, Teratake Y, Shimazui T, Lee C, Oda S. Influence of autophagy on acute kidney injury in a murine cecal ligation and puncture sepsis model. Sci Rep 2018; 8:1050. [PMID: 29348412 PMCID: PMC5773584 DOI: 10.1038/s41598-018-19350-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022] Open
Abstract
The role of autophagy in the maintenance of renal homeostasis during sepsis is not well understood. We therefore aimed to determine the influence of autophagy on kidney during sepsis using a murine sepsis model, i.e. cecal ligation and puncture (CLP). In CLP treated animals, the number of autolysosomes observed by electron microscopy increased over time. The number of autophagosomes in CLP animals decreased relative sham operated controls at 24 hrs after CLP, indicating that autophagy flux is already diminishing by that time. Moreover, CLP induced an increase in LC3-II/LC3-I ratio at 6-8 hrs, demonstrated in western blots, as well as an increase in GFP-LC3 dots at 6-8 hrs and 24 hrs, using immunofluorescence and anti-LC3 and LAMP1 antibodies on tissue sections from GFP-LC3 transgenic mice. LC3-II/LC3-I ratio and the number of co-localized GFP-LC3 dots and LAMP1 signals (GFP LC3 + LAMP1 dots) in CLP mice at 24 hrs were significantly reduced compared with data obtained at 6-8 hrs. Notably, acceleration of autophagy by rapamycin resulted in improvement of renal function that was associated with improvement in the histologic severity of tubular epithelial injury in CLP treated animals. Autophagy in the kidney was significantly slowed in the kidney during the acute phase of sepsis; nonetheless, autophagy in kidney appears to play a protective role against sepsis.
Collapse
Affiliation(s)
- Satoshi Sunahara
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Eizo Watanabe
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Masahiko Hatano
- Biomedical Research Center, Chiba University, Chiba, Japan.,Department of Biomedical Science, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Paul E Swanson
- Department of Pathology, University of Washington School of Medicine, Seattle, USA
| | - Takehiko Oami
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Lisa Fujimura
- Biomedical Research Center, Chiba University, Chiba, Japan
| | | | - Takashi Shimazui
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Chiwei Lee
- Department of Nephrology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shigeto Oda
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
27
|
Yang L, Zhang C, Zhao Y, Zhao N, Wu P, Zhang H, Shi C. Effects of Mycobacterium tuberculosis Mutant Strain Hsp16.3 Gene on Murine RAW 264.7 Macrophage Autophagy. DNA Cell Biol 2017; 37:7-14. [PMID: 29068712 DOI: 10.1089/dna.2016.3599] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Heat shock protein Hsp16.3 is closely related to latent Mycobacterium tuberculosis (MTB) infection and plays an important role in sustained survival when MTB is dormant. In this study, the Hsp16.3 gene mutant MTB H37Rv strain (Hsp16.3ΔMTB) was obtained through gene recombination and infected into murine RAW 264.7 macrophages. Western blotting and immunofluorescence showed increased expression of the autophagy-related protein LC3, and transmission electron microscopy showed significantly increased macrophage autophagosomes, suggesting that Hsp16.3ΔMTB facilitates murine macrophage autophagy. These findings have implications for preventing and controlling tuberculosis.
Collapse
Affiliation(s)
- Li Yang
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Caiqin Zhang
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Yong Zhao
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Ningning Zhao
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Pengpeng Wu
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Hai Zhang
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| | - Changhong Shi
- Division of Infection and Immunology, Laboratory Animals Center, Fourth Military Medical University , Xi'an, China
| |
Collapse
|
28
|
Abstract
The secretion of proteins that damage host tissue is well established as integral to the infectious processes of many bacterial pathogens. However, recent advances in our understanding of the activity of toxins suggest that the attributes we have assigned to them from early in vitro experimentation have misled us into thinking of them as merely destructive tools. Here, we will discuss the multifarious ways in which toxins contribute to the lifestyle of bacteria and, by considering their activity from an evolutionary perspective, demonstrate how this extends far beyond their ability to destroy host tissue.
Collapse
|
29
|
Abstract
Group A streptococcus (GAS) is an important human pathogen that causes a wide variety of cutaneous and systemic infections. Although originally thought to be an extracellular bacterium, numerous studies have demonstrated that GAS can trigger internalization into nonimmune cells to escape from immune surveillance or antibiotic-mediated killing. Epithelial cells possess a defense mechanism involving autophagy-mediated targeting and killing of GAS within lysosome-fused autophagosomes. In endothelial cells, in contrast, we previously showed that autophagy is not sufficient for GAS killing. In the present study, we showed higher galectin-3 (Gal-3) expression and lower Gal-8 expression in endothelial cells than in epithelial cells. The recruitment of Gal-3 to GAS is higher and the recruitment of Gal-8 to GAS is lower in endothelial cells than in epithelial cells. We further showed that Gal-3 promotes GAS replication and diminishes the recruitment of Gal-8 and ubiquitin, the latter of which is a critical protein for autophagy sequestration. After knockdown of Gal-3 in endothelial cells, the colocalization of Gal-8, parkin, and ubiquitin-decorated GAS is significantly increased, as is the interaction of Gal-8 and parkin, an E3 ligase. Furthermore, inhibition of Gal-8 in epithelial cells attenuates recruitment of parkin; both Gal-8 and parkin contribute to ubiquitin recruitment and GAS elimination. Animal studies confirmed that Gal-3-knockout mice develop less-severe skin damage and that GAS replication can be detected only in the air pouch and not in organs and endothelial cells. These results demonstrate that Gal-3 inhibits ubiquitin recruitment by blocking Gal-8 and parkin recruitment, resulting in GAS replication in endothelial cells. In epithelial cells, GAS can be efficiently killed within the lysosome-fused autophaosome compartment. However, we previously showed that, in spite of LC-3 recruitment, the autophagic machinery is not sufficient for GAS killing in endothelial cells. In this report, we provide the first evidence that Gal-3, highly expressed in endothelial cells, blocks the tagging of ubiquitin to GAS by inhibiting recruitment of Gal-8 and parkin, leading to an enhancement of GAS replication. We also provide the first demonstration that Gal-8 can interact with parkin, the critical E3 ligase, for resistance to intracellular bacteria by facilitating the decoration of bacteria with ubiquitin chains. Our findings reveal that differential levels of Gal-3 and Gal-8 expression and recruitment to GAS between epithelial cells and endothelial cells may contribute to the different outcomes of GAS elimination or survival and growth of GAS in these two types of cells.
Collapse
|
30
|
Lu Z, Xie D, Chen Y, Tian E, Muhammad I, Chen X, Miao Y, Hu W, Wu Z, Ni H, Xin J, Li Y, Li J. TLR2 mediates autophagy through ERK signaling pathway in Mycoplasma gallisepticum-infected RAW264.7 cells. Mol Immunol 2017; 87:161-170. [PMID: 28478286 DOI: 10.1016/j.molimm.2017.04.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 04/16/2017] [Accepted: 04/22/2017] [Indexed: 12/11/2022]
Abstract
Toll-like receptor 2 (TLR2) plays a crucial role in early innate immune response of host to various microorganisms. Mycoplasma gallisepticum (MG) is one of the major pathogen that can cause chronic respiratory diseases in chickens, but the molecular mechanism of MG infection still remained unclear. In this study, we examined the typical hallmarks of autophagy and multiple signaling pathways by western blot, immunofluorescence microscopy and electron microscopy. The results indicated that infection of mouse macrophage cell line RAW264.7 with MG activated autophagy and mitogen-activated protein kinases (MAPKs). Silencing of TLR2 by siRNA substantially down-regulated MG-triggered autophagy in macrophages, and markedly reduced MG-induced extracellular regulated protein kinase (ERK) in macrophages but did not down-regulate c-Jun N-terminal kinase (JNK) and p38. Importantly, in macrophages, inhibition of ERK by PD98059 (ERK inhibitor) also significantly attenuated the level of autophagy upon MG infection, and the simultaneous treatment of TLR2 siRNA and PD98059 showed a similar effect on MG-induced autophagy as compared with TLR2 siRNA treatment alone. These findings thus demonstrate that TLR2 may mediate MG-induced autophagy through ERK signaling pathway in macrophage.
Collapse
Affiliation(s)
- Ziyin Lu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Daoyuan Xie
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Ying Chen
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Erjie Tian
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Ishfaq Muhammad
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Xueping Chen
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Yusong Miao
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Wanjun Hu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Ziyong Wu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Huili Ni
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China
| | - Jiuqing Xin
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 678 Heping Road, Xiangfang District, Harbin 150030, PR China
| | - Yuan Li
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 678 Heping Road, Xiangfang District, Harbin 150030, PR China
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Xiangfang District, Harbin 150030, PR China.
| |
Collapse
|
31
|
Vu CB, Bridges RJ, Pena-Rasgado C, Lacerda AE, Bordwell C, Sewell A, Nichols AJ, Chandran S, Lonkar P, Picarella D, Ting A, Wensley A, Yeager M, Liu F. Fatty Acid Cysteamine Conjugates as Novel and Potent Autophagy Activators That Enhance the Correction of Misfolded F508del-Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). J Med Chem 2016; 60:458-473. [PMID: 27976892 DOI: 10.1021/acs.jmedchem.6b01539] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A depressed autophagy has previously been reported in cystic fibrosis patients with the common F508del-CFTR mutation. This report describes the synthesis and preliminary biological characterization of a novel series of autophagy activators involving fatty acid cysteamine conjugates. These molecular entities were synthesized by first covalently linking cysteamine to docosahexaenoic acid. The resulting conjugate 1 synergistically activated autophagy in primary homozygous F508del-CFTR human bronchial epithelial (hBE) cells at submicromolar concentrations. When conjugate 1 was used in combination with the corrector lumacaftor and the potentiator ivacaftor, it showed an additive effect, as measured by the increase in the chloride current in a functional assay. In order to obtain a more stable form for oral dosing, the sulfhydryl group in conjugate 1 was converted into a functionalized disulfide moiety. The resulting conjugate 5 is orally bioavailable in the mouse, rat, and dog and allows a sustained delivery of the biologically active conjugate 1.
Collapse
Affiliation(s)
- Chi B Vu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Robert J Bridges
- Chicago Medical School, Rosalind Franklin University of Medicine and Science , 3333 Green Bay Road, North Chicago, Illinois 60064, United States
| | - Cecilia Pena-Rasgado
- Chicago Medical School, Rosalind Franklin University of Medicine and Science , 3333 Green Bay Road, North Chicago, Illinois 60064, United States
| | - Antonio E Lacerda
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Curtis Bordwell
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Abby Sewell
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Andrew J Nichols
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Sachin Chandran
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Pallavi Lonkar
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Dominic Picarella
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Amal Ting
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Allison Wensley
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Maisy Yeager
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Feng Liu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
32
|
Tang B, Wang K, Jia YP, Zhu P, Fang Y, Zhang ZJ, Mao XH, Li Q, Zeng DZ. Fusobacterium nucleatum-Induced Impairment of Autophagic Flux Enhances the Expression of Proinflammatory Cytokines via ROS in Caco-2 Cells. PLoS One 2016; 11:e0165701. [PMID: 27828984 PMCID: PMC5102440 DOI: 10.1371/journal.pone.0165701] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 10/17/2016] [Indexed: 12/16/2022] Open
Abstract
Fusobacterium nucleatum (F. nucleatum) plays a critical role in gastrointestinal inflammation. However, the exact mechanism by which F. nucleatum contributes to inflammation is unclear. In the present study, it was revealed that F. nucleatum could induce the production of proinflammatory cytokines (IL-8, IL-1β and TNF-α) and reactive oxygen species (ROS) in Caco-2 colorectal) adenocarcinoma cells. Furthermore, ROS scavengers (NAC or Tiron) could decrease the production of proinflammatory cytokines during F. nucleatum infection. In addition, we observed that autophagy is impaired in Caco-2 cells after F. nucleatum infection. The production of proinflammatory cytokines and ROS induced by F. nucleatum was enhanced with either autophagy pharmacologic inhibitors (3-methyladenine, bafilomycin A1) or RNA interference in essential autophagy genes (ATG5 or ATG12) in Caco-2 cells. Taken together, these results indicate that F. nucleatum-induced impairment of autophagic flux enhances the expression of proinflammatory cytokines via ROS in Caco-2 Cells.
Collapse
Affiliation(s)
- Bin Tang
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
- Emei Sanatorium of PLA Rocket Force, Emeishan, China
| | - Kun Wang
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
| | - Yin-ping Jia
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
| | - Pan Zhu
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
| | - Yao Fang
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
| | - Zhu-jun Zhang
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
| | - Xu-hu Mao
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, Southwest Hospital & College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
- * E-mail: (DZZ); (QL)
| | - Dong-Zhu Zeng
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
- * E-mail: (DZZ); (QL)
| |
Collapse
|
33
|
Strobel M, Pförtner H, Tuchscherr L, Völker U, Schmidt F, Kramko N, Schnittler HJ, Fraunholz MJ, Löffler B, Peters G, Niemann S. Post-invasion events after infection with Staphylococcus aureus are strongly dependent on both the host cell type and the infecting S. aureus strain. Clin Microbiol Infect 2016; 22:799-809. [PMID: 27393124 DOI: 10.1016/j.cmi.2016.06.020] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022]
Abstract
Host cell invasion is a major feature of Staphylococcus aureus and contributes to infection development. The intracellular metabolically active bacteria can induce host cell activation and death but they can also persist for long time periods. In this study a comparative analysis was performed of different well-characterized S. aureus strains in their interaction with a variety of host cell types. Staphylococcus aureus (strains 6850, USA300, LS1, SH1000, Cowan1) invasion was compared in different human cell types (epithelial and endothelial cells, keratinocytes, fibroblasts, osteoblasts). The number of intracellular bacteria was determined, cell inflammation was investigated, as well as cell death and phagosomal escape of bacteria. To explain strain-dependent differences in the secretome, a proteomic approach was used. Barrier cells took up high amounts of bacteria and were killed by aggressive strains. These strains expressed high levels of toxins, and possessed the ability to escape from phagolysosomes. Osteoblasts and keratinocytes ingested less bacteria, and were not killed, even though the primary osteoblasts were strongly activated by S. aureus. In all cell types S. aureus was able to persist. Strong differences in uptake, cytotoxicity, and inflammatory response were observed between primary cells and their corresponding cell lines, demonstrating that cell lines reflect only partially the functions and physiology of primary cells. This study provides a contribution for a better understanding of the pathomechanisms of S. aureus infections. The proteomic data provide important basic knowledge on strains commonly used in the analysis of S. aureus-host cell interaction.
Collapse
Affiliation(s)
- M Strobel
- University Hospital of Muenster, Institute of Medical Microbiology, Muenster, Germany
| | - H Pförtner
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - L Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, Germany
| | - U Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - F Schmidt
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - N Kramko
- Westfaelische-Wilhelms University, Institute of Anatomy and Vascular Biology, Muenster, Germany
| | - H-J Schnittler
- Westfaelische-Wilhelms University, Institute of Anatomy and Vascular Biology, Muenster, Germany
| | - M J Fraunholz
- Department of Microbiology, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | - B Löffler
- Institute of Medical Microbiology, Jena University Hospital, Germany
| | - G Peters
- University Hospital of Muenster, Institute of Medical Microbiology, Muenster, Germany; Cluster of Excellence EXC 1003, Cells in Motion, Muenster, Germany
| | - S Niemann
- University Hospital of Muenster, Institute of Medical Microbiology, Muenster, Germany.
| |
Collapse
|
34
|
The staphylococcal surface-glycopolymer wall teichoic acid (WTA) is crucial for complement activation and immunological defense against Staphylococcus aureus infection. Immunobiology 2016; 221:1091-101. [PMID: 27424796 DOI: 10.1016/j.imbio.2016.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 11/22/2022]
Abstract
Staphylococcus aureus is a Gram-positive bacterial pathogen that is decorated by glycopolymers, including wall teichoic acid (WTA), peptidoglycan, lipoteichoic acid, and capsular polysaccharides. These bacterial surface glycopolymers are recognized by serum antibodies and a variety of pattern recognition molecules, including mannose-binding lectin (MBL). Recently, we demonstrated that human serum MBL senses staphylococcal WTA. Whereas MBL in infants who have not yet fully developed adaptive immunity binds to S. aureus WTA and activates complement serum, MBL in adults who have fully developed adaptive immunity cannot bind to WTA because of an inhibitory effect of serum anti-WTA IgG. Furthermore, we showed that human anti-WTA IgGs purified from pooled adult serum IgGs triggered activation of classical complement-dependent opsonophagocytosis against S. aureus. Because the epitopes of WTA that are recognized by anti-WTA IgG and MBL have not been determined, we constructed several S. aureus mutants with altered WTA glycosylation. Our intensive biochemical studies provide evidence that the β-GlcNAc residues of WTA are required for the induction of anti-WTA IgG-mediated opsonophagocytosis and that both β- and α-GlcNAc residues are required for MBL-mediated complement activation. The molecular interactions of other S. aureus cell wall components and host recognition proteins are also discussed. In summary, in this review, we discuss the biological importance of S. aureus cell surface glycopolymers in complement activation and host defense responses.
Collapse
|
35
|
Qin Z, Yang Y, Wang H, Luo J, Huang X, You J, Wang B, Li M. Role of Autophagy and Apoptosis in the Postinfluenza Bacterial Pneumonia. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3801026. [PMID: 27376082 PMCID: PMC4916274 DOI: 10.1155/2016/3801026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/05/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
The risk of influenza A virus (IAV) is more likely caused by secondary bacterial infections. During the past decades, a great amount of studies have been conducted on increased morbidity from secondary bacterial infections following influenza and provide an increasing number of explanations for the mechanisms underlying the infections. In this paper, we first review the recent research progress that IAV infection increased susceptibility to bacterial infection. We then propose an assumption that autophagy and apoptosis manipulation are beneficial to antagonize post-IAV bacterial infection and discuss the clinical significance.
Collapse
Affiliation(s)
- Zhen Qin
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Yang
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongren Wang
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Luo
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaojun Huang
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiangzhou You
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Baoning Wang
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mingyuan Li
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
36
|
Liu PF, Cheng JS, Sy CL, Huang WC, Yang HC, Gallo RL, Huang CM, Shu CW. IsaB Inhibits Autophagic Flux to Promote Host Transmission of Methicillin-Resistant Staphylococcus aureus. J Invest Dermatol 2015; 135:2714-2722. [PMID: 26134948 PMCID: PMC4641007 DOI: 10.1038/jid.2015.254] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 06/17/2015] [Indexed: 12/19/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has emerged as a major nosocomial pathogen that is widespread in both health-care facilities and in the community at large, as a result of direct host-to-host transmission. Several virulence factors are associated with pathogen transmission to naive hosts. Immunodominant surface antigen B (IsaB) is a virulence factor that helps Staphylococcus aureus to evade the host defense system. However, the mechanism of IsaB on host transmissibility remains unclear. We found that IsaB expression was elevated in transmissible MRSA. Wild-type isaB strains inhibited autophagic flux to promote bacterial survival and elicit inflammation in THP-1 cells and mouse skin. MRSA isolates with increased IsaB expression showed decreased autophagic flux, and the MRSA isolate with the lowest IsaB expression showed increased autophagic flux. In addition, recombinant IsaB rescued the virulence of the isaB deletion strain and increased the group A streptococcus (GAS) virulence in vivo. Together, these results reveal that IsaB diminishes autophagic flux, thereby allowing MRSA to evade host degradation. These findings suggest that IsaB is a suitable target for preventing or treating MRSA infection.
Collapse
Affiliation(s)
- Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Medicine, Division of Dermatology, University of California, San Diego, USA
| | - Jin-Shiung Cheng
- Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Cheng-Len Sy
- Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Chun Huang
- Cardiovascular Medical Center, Kaohsiung Veterans General Hospital, Kaohsiung City
- School of Medicine, National Yang-Ming University, Taipei City
| | - Hsiu-Chen Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Richard L. Gallo
- Department of Medicine, Division of Dermatology, University of California, San Diego, USA
- VA San Diego Healthcare Center, San Diego, USA
| | - Chun-Ming Huang
- Department of Medicine, Division of Dermatology, University of California, San Diego, USA
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| |
Collapse
|
37
|
Antibiotic resistance breakers: can repurposed drugs fill the antibiotic discovery void? Nat Rev Drug Discov 2015; 14:821-32. [PMID: 26493767 DOI: 10.1038/nrd4675] [Citation(s) in RCA: 227] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
38
|
Insufficient Acidification of Autophagosomes Facilitates Group A Streptococcus Survival and Growth in Endothelial Cells. mBio 2015; 6:e01435-15. [PMID: 26419882 PMCID: PMC4611045 DOI: 10.1128/mbio.01435-15] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Group A streptococcus (GAS) is an important human pathogen, and its invasion via blood vessels is critically important in serious events such as bacteremia or multiorgan failure. Although GAS was identified as an extracellular bacterium, the internalization of GAS into nonphagocytic cells may provide a strategy to escape from immune surveillance and antibiotic killing. However, GAS has also been reported to induce autophagy and is efficiently killed within lysosome-fused autophagosomes in epithelial cells. In this study, we show that GAS can replicate in endothelial cells and that streptolysin O is required for GAS growth. Bacterial replication can be suppressed by altering GAS gene expression in an acidic medium before internalization into endothelial cells. The inhibitory effect on GAS replication can be reversed by treatment with bafilomycin A1, a specific inhibitor of vacuolar-type H+-ATPase. Compared with epithelial cells in which acidification causes autophagy-mediated clearance of GAS, there was a defect in acidification of GAS-containing vesicles in endothelial cells. Consequently, endothelial cells fail to maintain low pH in GAS-containing autophagosomes, thereby permitting GAS replication inside LAMP-1- and LC3-positive vesicles. Furthermore, treatment of epithelial cells with bafilomycin A1 resulted in defective GAS clearance by autophagy, with subsequent bacterial growth intracellularly. Therefore, low pH is a key factor for autophagy-mediated suppression of GAS growth inside epithelial cells, while defective acidification of GAS-containing vesicles results in bacterial growth in endothelial cells. Previous reports showed that GAS can induce autophagy and is efficiently killed within lysosome-fused autophagosomes in epithelial cells. In endothelial cells, in contrast, induction of autophagy is not sufficient for GAS killing. In this study, we provide the first evidence that low pH is required to prevent intracellular growth of GAS in epithelial cells and that this mechanism is defective in endothelial cells. Treatment of GAS with low pH altered GAS growth rate and gene expression of virulence factors and resulted in enhanced susceptibility of GAS to intracellular lysosomal killing. Our findings reveal the existence of different mechanisms of host defense against GAS invasion between epithelial and endothelial cells.
Collapse
|
39
|
Duan X, Zhang T, Ding S, Wei J, Su C, Liu H, Xu G. microRNA-17-5p Modulates Bacille Calmette-Guerin Growth in RAW264.7 Cells by Targeting ULK1. PLoS One 2015; 10:e0138011. [PMID: 26384021 PMCID: PMC4575043 DOI: 10.1371/journal.pone.0138011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/24/2015] [Indexed: 01/07/2023] Open
Abstract
To explore the potential roles of miRNAs in controlling the survival of mycobacteria in macrophages, miR-17-5p in the regulation of Bacillus Calmette-Guérin(BCG)growth in the macrophage RAW264.7 cells was interrogated. Our results reveal that an infection of BCG shows a time-dependent up-regulation of miR-17-5p in RAW264.7 cells in early phase; importantly, excessive expression of miR-17-5p in these cells exhibits an increased propagation of intracellular BCG. Mechanistically, the Unc-51 like autophagy activating kinase 1 (ULK1), an initial molecular of autophagy are identified as novel target of miR-17-5p, the miR-17-5p is capable of targeting down-regulating the expression of ULK1 protein. In addition, an overexpression of miR-17-5p in RAW264.7 cells is correlated with repression of ULK1 and the autophagosome related proteins LC3I/II. These results imply that miR-17-5p may be able to arrest the maturation of mycobacterial phagosomes in part by targeting ULK1, subsequently reduces the ability of host cells to kill intracellular BCG.
Collapse
Affiliation(s)
- Xiangguo Duan
- School of LaboratoryMedicine, Ningxia Medical University, Yinchuan, 750004, China
- General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
| | - Tao Zhang
- School of LaboratoryMedicine, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
- Taian City Central Hospital,Taian City, Shandong Province, 271000, China
| | - Shuqin Ding
- School of LaboratoryMedicine, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
| | - Jun Wei
- General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
| | - Chunxia Su
- School of LaboratoryMedicine, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
| | - Hongpeng Liu
- School of LaboratoryMedicine, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
| | - Guangxian Xu
- School of LaboratoryMedicine, Ningxia Medical University, Yinchuan, 750004, China
- General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
- * E-mail:
| |
Collapse
|
40
|
Autophagy and inflammatory bowel disease: Association between variants of the autophagy-related IRGM gene and susceptibility to Crohn's disease. Dig Liver Dis 2015; 47:744-50. [PMID: 26066377 DOI: 10.1016/j.dld.2015.05.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/25/2015] [Accepted: 05/14/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Crohn's disease and ulcerative colitis are inflammatory bowel diseases involving a genetically determined inappropriate mucosal immune response towards luminal antigens, including resident bacterial flora. Recent studies identified susceptibility genes involved in autophagy. AIMS We analyzed known autophagic loci (IRGM, ULK1 and AMBRA1) previously described as associated with inflammatory bowel diseases or with other autoimmune and/or inflammatory disorders in a sample of Italian inflammatory bowel diseases patients in order to confirm their possible involvement and relative contribution in the disease. METHODS We performed a case-control association study, a sub-phenotype correlation and a haplotype analysis. The analysis included 263 Crohn's disease, 206 ulcerative colitis patients and 245 matched healthy controls. Five polymorphisms were genotyped by allelic discrimination assays. RESULTS IRGM was the most strongly associated with Crohn's disease susceptibility [rs13361189: P=0.011, OR=1.66 [95% CI: (1.12-2.45)]; rs4958847: P=0.05, OR=1.43 [95% CI: (1-2.03)]. The SNP rs13361189 was also found to increase the risk of Crohn's disease clinical sub-phenotype (fibrostricturing behaviour, ileal disease, perianal disease, intestinal resection). These findings suggest that IRGM variants may modulate clinical characteristics of Crohn's disease. CONCLUSIONS Our study confirms IRGM rs13361189 and rs4958847 polymorphisms to be important for Crohn's disease susceptibility and phenotype modulation, in accordance with previous findings.
Collapse
|
41
|
Abstract
Autophagy, a lysosomal degradative pathway that maintains cellular homeostasis, has emerged as an innate immune defense against pathogens. The role of autophagy in the deregulated HIV-infected central nervous system (CNS) is unclear. We have found that HIV-1-induced neuro-glial (neurons and astrocytes) damage involves modulation of the autophagy pathway. Neuro-glial stress induced by HIV-1 led to biochemical and morphological dysfunctions. X4 HIV-1 produced neuro-glial toxicity coupled with suppression of autophagy, while R5 HIV-1-induced toxicity was restricted to neurons. Rapamycin, a specific mTOR inhibitor (autophagy inducer) relieved the blockage of the autophagy pathway caused by HIV-1 and resulted in neuro-glial protection. Further understanding of the regulation of autophagy by cytokines and chemokines or other signaling events may lead to recognition of therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Rajeev Mehla
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Ashok Chauhan
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|
42
|
Tuchscherr L, Bischoff M, Lattar SM, Noto Llana M, Pförtner H, Niemann S, Geraci J, Van de Vyver H, Fraunholz MJ, Cheung AL, Herrmann M, Völker U, Sordelli DO, Peters G, Löffler B. Sigma Factor SigB Is Crucial to Mediate Staphylococcus aureus Adaptation during Chronic Infections. PLoS Pathog 2015; 11:e1004870. [PMID: 25923704 PMCID: PMC4414502 DOI: 10.1371/journal.ppat.1004870] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/09/2015] [Indexed: 12/26/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen that causes a range of infections from acute invasive to chronic and difficult-to-treat. Infection strategies associated with persisting S. aureus infections are bacterial host cell invasion and the bacterial ability to dynamically change phenotypes from the aggressive wild-type to small colony variants (SCVs), which are adapted for intracellular long-term persistence. The underlying mechanisms of the bacterial switching and adaptation mechanisms appear to be very dynamic, but are largely unknown. Here, we analyzed the role and the crosstalk of the global S. aureus regulators agr, sarA and SigB by generating single, double and triple mutants, and testing them with proteome analysis and in different in vitro and in vivo infection models. We were able to demonstrate that SigB is the crucial factor for adaptation in chronic infections. During acute infection, the bacteria require the simultaneous action of the agr and sarA loci to defend against invading immune cells by causing inflammation and cytotoxicity and to escape from phagosomes in their host cells that enable them to settle an infection at high bacterial density. To persist intracellularly the bacteria subsequently need to silence agr and sarA. Indeed agr and sarA deletion mutants expressed a much lower number of virulence factors and could persist at high numbers intracellularly. SigB plays a crucial function to promote bacterial intracellular persistence. In fact, ΔsigB-mutants did not generate SCVs and were completely cleared by the host cells within a few days. In this study we identified SigB as an essential factor that enables the bacteria to switch from the highly aggressive phenotype that settles an acute infection to a silent SCV-phenotype that allows for long-term intracellular persistence. Consequently, the SigB-operon represents a possible target to develop preventive and therapeutic strategies against chronic and therapy-refractory infections.
Collapse
Affiliation(s)
- Lorena Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, University of Saarland Medical Center, Homburg, Germany
| | - Santiago M. Lattar
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM UBA-CONICET) y Facultad de Medicina, University of Buenos Aires, Buenos Aires, Argentina
| | - Mariangeles Noto Llana
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM UBA-CONICET) y Facultad de Medicina, University of Buenos Aires, Buenos Aires, Argentina
| | - Henrike Pförtner
- Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Jennifer Geraci
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Hélène Van de Vyver
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Martin J. Fraunholz
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Ambrose L. Cheung
- Dartmouth Medical School, Hanover, New Hampshire, United States of America
| | - Mathias Herrmann
- Institute of Medical Microbiology and Hygiene, University of Saarland Medical Center, Homburg, Germany
| | - Uwe Völker
- Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Daniel O. Sordelli
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM UBA-CONICET) y Facultad de Medicina, University of Buenos Aires, Buenos Aires, Argentina
| | - Georg Peters
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
- * E-mail:
| |
Collapse
|
43
|
Abstract
Skin is the most common site of Staphylococcus aureus infection. While most of these infections are self-limited, recurrent infections are common. Keratinocytes and recruited immune cells participate in skin defense against infection. We postulated that S. aureus is able to adapt to the milieu within human keratinocytes to avoid keratinocyte-mediated clearance. From a collection of S. aureus isolated from chronically infected patients with atopic dermatitis, we noted 22% had an agr mutant-like phenotype. Using several models of human skin infection, we demonstrate that toxin-deficient, agr mutants of methicillin-resistant S. aureus (MRSA) USA300 are able to persist within keratinocytes by stimulating autophagy and evading caspase-1 and inflammasome activation. MRSA infection induced keratinocyte autophagy, as evidenced by galectin-8 and LC3 accumulation. Autophagy promoted the degradation of inflammasome components and facilitated staphylococcal survival. The recovery of more than 58% agr or RNAIII mutants (P < 0.0001) of an inoculum of wild-type (WT) MRSA from within wortmannin-treated keratinocytes compared to control keratinocytes reflected the survival advantage for mutants no longer expressing agr-dependent toxins. Our results illustrate the dynamic interplay between S. aureus and keratinocytes that can result in the selection of mutants that have adapted specifically to evade keratinocyte-mediated clearance mechanisms. Human skin is a major site of staphylococcal infection, and keratinocytes actively participate in eradication of these pathogens. We demonstrate that methicillin-resistant Staphylococcus aureus (MRSA) is ingested by keratinocytes and activates caspase-1-mediated clearance through pyroptosis. Toxin-deficient MRSA mutants are selected within keratinocytes that fail to induce caspase-1 activity and keratinocyte-mediated clearance. These intracellular staphylococci induce autophagy that enhances their intracellular survival by diminishing inflammasome components. These findings suggest that S. aureus mutants, by exploiting autophagy, can persist within human keratinocytes.
Collapse
|
44
|
Warnking K, Klemm C, Löffler B, Niemann S, van Krüchten A, Peters G, Ludwig S, Ehrhardt C. Super-infection with Staphylococcus aureus inhibits influenza virus-induced type I IFN signalling through impaired STAT1-STAT2 dimerization. Cell Microbiol 2014; 17:303-17. [PMID: 25293394 DOI: 10.1111/cmi.12375] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/02/2014] [Accepted: 10/05/2014] [Indexed: 12/24/2022]
Abstract
Bacterial super-infections are a major complication in influenza virus-infected patients. In response to infection with influenza viruses and bacteria, a complex interplay of cellular signalling mechanisms is initiated, regulating the anti-pathogen response but also pathogen-supportive functions. Here, we show that influenza viruses replicate to a higher efficiency in cells co-infected with Staphylococcus aureus (S. aureus). While cells initially respond with increased induction of interferon beta upon super-infection, subsequent interferon signalling and interferon-stimulated gene expression are rather impaired due to a block of STAT1-STAT2 dimerization. Thus, S. aureus interrupts the first line of defence against influenza viruses, resulting in a boost of viral replication, which may lead to enhanced viral pathogenicity.
Collapse
Affiliation(s)
- Kathrin Warnking
- Institute of Molecular Virology (IMV), Center for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von Esmarch-Str. 56, D-48149, Muenster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Targeting autophagy in skin diseases. J Mol Med (Berl) 2014; 93:31-8. [PMID: 25404245 DOI: 10.1007/s00109-014-1225-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/02/2014] [Accepted: 11/05/2014] [Indexed: 12/19/2022]
Abstract
Autophagy is a major intracellular degradative process by which cytoplasmic materials are sequestered in double-membraned vesicles and degraded upon fusion with lysosomes. Under normal circumstances, basal autophagy is necessary to maintain cellular homeostasis by scavenging dysfunctional or damaged organelles or proteins. In addition to its vital homeostatic role, this degradation pathway has been implicated in many different cellular processes such as cell apoptosis, inflammation, pathogen clearance, and antigen presentation and thereby has been linked to a variety of human disorders, including metabolic conditions, neurodegenerative diseases, cancers, and infectious diseases. The skin, the largest organ of the body, serves as the first line of defense against many different environmental insults; however, only a few studies have examined the effect of autophagy on the pathogenesis of skin diseases. This review provides an overview of the mechanisms of autophagy and highlights recent findings relevant to the role of autophagy in skin diseases and strategies for therapeutic modulation.
Collapse
|
46
|
Tawfik A, Flanagan PK, Campbell BJ. Escherichia coli-host macrophage interactions in the pathogenesis of inflammatory bowel disease. World J Gastroenterol 2014; 20:8751-8763. [PMID: 25083050 PMCID: PMC4112894 DOI: 10.3748/wjg.v20.i27.8751] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/07/2014] [Accepted: 04/03/2014] [Indexed: 02/06/2023] Open
Abstract
Multiple studies have demonstrated alterations in the intestinal microbial community (termed the microbiome) in Crohn’s disease (CD) and several lines of evidence suggest these changes may have a significant role in disease pathogenesis. In active and quiescent disease, both the faecal and mucosa-associated microbiome are discordant with matched controls with reduced biodiversity, changes in dominant organisms and increased temporal variation described. Mucosa-associated adherent, invasive Escherichia coli (E. coli) (AIEC), pro-inflammatory and resistant to killing by mucosal macrophages, appear to be particularly important. AIEC possess several virulence factors which may confer pathogenic potential in CD. Type-1 pili (FimH) allow adherence to intestinal cells via cell-surface carcinoembryonic antigen-related cell adhesion molecules and possession of long polar fimbrae promotes translocation across the intestinal mucosa via microfold (M)-cells of the follicle-associated epithelium. Resistance to stress genes (htrA, dsbA and hfq) and tolerance of an acidic pH may contribute to survival within the phagolysosomal environment. Here we review the current understanding of the role of mucosa-associated E. coli in Crohn’s pathogenesis, the role of the innate immune system, factors which may contribute to prolonged bacterial survival and therapeutic strategies to target intracellular E. coli.
Collapse
|
47
|
Chen X, Khambu B, Zhang H, Gao W, Li M, Chen X, Yoshimori T, Yin XM. Autophagy induced by calcium phosphate precipitates targets damaged endosomes. J Biol Chem 2014; 289:11162-11174. [PMID: 24619419 PMCID: PMC4036255 DOI: 10.1074/jbc.m113.531855] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/24/2014] [Indexed: 11/06/2022] Open
Abstract
Calcium phosphate precipitates (CPPs) form complexes with DNA, which enter cells via endocytosis. Under this condition CPPs induce autophagy via the canonic autophagy machinery. Here we showed that CPP-induced autophagy was also dependent on endocytosis as the process was significantly inhibited by methyl-β-cyclodextrin and dynasore, which suppress clathrin-dependent endocytosis. Consistently, CPP treatment triggered the formation of filipin-positive intracellular vesicles whose membranes are rich in cholesterol. Unexpectedly, these vesicles were also positive for galectin 3, suggesting that they were damaged and the membrane glycans became accessible to galectins to bind. Endosome damage was caused by endocytosis of CPPs and was reversed by calcium chelators or by endocytosis inhibitors. Notably, CPP-induced LC3-positive autophagosomes were colocalized with galectin 3, ubiquitin, and p62/SQSTM1. Inhibition of galectin 3 reduced p62 puncta and autophagosome formation. Knockdown of p62 additionally inhibited the colocalization of autophagosomes with galectins. Furthermore, most of the galectin 3-positive vesicles were colocalized with Rab7 or LAMP1. Agents that affect endosome/lysosome maturation and function, such as bafilomycin A1, also significantly affected CPP-induced tubulovesicular autophagosome formation. These findings thus indicate that endocytosed CPPs caused endosome damage and recruitment of galectins, particularly at the later endosome stage, which led to the interaction of the autophagosomal membranes with the damaged endosome in the presence of p62.
Collapse
Affiliation(s)
- Xi Chen
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China,; Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Bilon Khambu
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hao Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Wentao Gao
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Min Li
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202,; Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China, and
| | - Xiaoyun Chen
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Tamotsu Yoshimori
- Department of Genetics, Osaka University, Suita, Osaka 565-0871, Japan
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202,.
| |
Collapse
|
48
|
Bai R, You W, Chen J, Huang H, Ke C. Molecular cloning and expression analysis of GABA(A) receptor-associated protein (GABARAP) from small abalone, Haliotis diversicolor. FISH & SHELLFISH IMMUNOLOGY 2012; 33:675-682. [PMID: 22771962 DOI: 10.1016/j.fsi.2012.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 04/03/2012] [Accepted: 05/07/2012] [Indexed: 06/01/2023]
Abstract
GABA(A) receptor-associated protein (GABARAP), a multifunctional protein participating in autophagy process, is evolutionarily conserved and involves in innate immunity in eukaryotic cells, but currently there is no research on the relationship between GABARAP and innate immunity in mollusc. In the present study, the GABARAP full-length cDNA and its genomic DNA were firstly cloned from small abalone (Haliotis diversicolor), which was named as saGABARAP. Its full-length cDNA is 963 bp with a 354 bp open reading frame encoding a protein of 117 aa, a 276 bp 5'-UTR, and a 333 bp 3'-UTR including a poly(A) tail, two typical polyadenylation signals (AATAA) and two RNA instability motifs (ATTTA). The deduced protein has an estimated molecular weight of 13.9 kDa and a predicted PI of 8.73. Its genomic DNA comprises 4352 bp, containing three exons and two introns. Quantitative real-time PCR analysis revealed that saGABARAP was constitutively expressed in all examined tissues, with the highest expression level in hepatopancreas, and was upregulated in hepatopancreas and hemocytes after bacterial challenge. In addition, saGABARAP was ubiquitously expressed at all examined embryonic and larval development stages. These results suggested that saGABARAP could respond to bacteria challenge and may play a vital role in the adult innate immune system against pathogens and the development process of abalone embryo and larvae.
Collapse
Affiliation(s)
- Rongyao Bai
- Department of Biochemistry and Biotechnology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | | | | | | | | |
Collapse
|
49
|
Variant in the 5' untranslated region of insulin-like growth factor 1 receptor is associated with susceptibility to mastitis in cattle. G3-GENES GENOMES GENETICS 2012; 2:1077-84. [PMID: 22973545 PMCID: PMC3429922 DOI: 10.1534/g3.112.003095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/29/2012] [Indexed: 11/18/2022]
Abstract
Mastitis is a common infectious disease of the mammary gland and generates large losses in the dairy industry. By means of positional cloning and functional analysis techniques, we here show that insulin-like growth factor 1 receptor (IGF1R) can possibly mediate susceptibility to mastitis through autophagy. Scanning the whole genome of cows (Bos taurus) that were susceptible or resistant to mastitis in the half-sib families revealed that susceptible cows had a relatively long stretch of cytosine residues (C stretch) in the 5' untranslated region of IGF1R. The forebrain embryonic zinc finger-like (FEZL) transcription factor, which was previously identified as a factor controlling mastitis resistance in the same half-sib families, bound the C stretch of IGF1R. The susceptible type of FEZL with a glycine stretch containing 13 glycines (13G) and the longer C stretch of IGF1R together enhanced expression of IGF1R. Enhancing IGF1R inhibited autophagy in response to Streptococcus agalactiae invasion of mammary epithelial cells, whereas treatment with rapamycin, a known inducer of autophagy, rescued it. Cows carrying the variant combination of 13GFEZL might be more susceptible to mastitis as the result of impaired autophagy. Our results suggest that IGF1R could control innate immunity in mammals and serve as a potential tool for preventing mastitis.
Collapse
|
50
|
Staphylococcus-aureus-Adhärenz, Wirtszellinvasion und Persistenz. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2012. [DOI: 10.1007/s00398-012-0943-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|