1
|
Ramanathan C, Thomas E, Henschen AE, Adelman JS, Zhang Y. Mycoplasma gallisepticum (MG) infection inhibits mitochondrial respiratory function in a wild songbird. J Exp Biol 2025; 228:jeb249705. [PMID: 40181769 DOI: 10.1242/jeb.249705] [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: 10/16/2024] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
An animal's immune function is vital for survival but is potentially metabolically expensive. Some pathogens can manipulate their hosts' immune and metabolic responses. One example is Mycoplasma gallisepticum (MG), which infects both the respiratory system and conjunctiva of the eye in house finches (Haemorhous mexicanus). MG has been shown to exhibit immune- and metabolic-suppressive properties, but the physiological mechanisms are still unknown. Recent studies demonstrated that mitochondria could serve as powerhouses for both ATP production and immunity, notably inflammatory processes, by regulating complex II and its metabolites. Consequently, in this study, we investigate the short-term (3 days post-inoculation) and long-term (34 days post-inoculation) effects of MG infection on the hepatic mitochondrial respiration of house finches from two populations infected with two different MG isolates. After short-term infection, MG-infected birds had significantly lower state 2 and state 4 respiration, but only when using complex II substrates. After long-term infection, MG-infected birds exhibited lower state 3 respiration with both complex I and II substrates, resulting in a lower respiratory control ratio compared with uninfected controls, which aligned with the hypothesized metabolic-suppressive properties of MG. Interestingly, there were limited differences in mitochondrial respiration regardless of house finch population of origin, MG isolate and whether birds recovered from infection or not. We propose that MG targets mitochondrial complex II for its immune-suppressive properties during the early stages of infection and inhibits mitochondrial respiration for its metabolic-suppressive properties at a later stage of infection, both of which should delay recovery of the host and extend infectious periods.
Collapse
Affiliation(s)
| | - Elina Thomas
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA
| | - Amberleigh E Henschen
- Department of Biological Science, University of Memphis, Memphis, TN 38152, USA
- Department of Biological Sciences, Eastern Illinois University, Charleston, IL 61920, USA
| | - James S Adelman
- Department of Biological Science, University of Memphis, Memphis, TN 38152, USA
| | - Yufeng Zhang
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|
2
|
Shahi A, Kidane D. Decoding mitochondrial DNA damage and repair associated with H. pylori infection. Front Cell Infect Microbiol 2025; 14:1529441. [PMID: 39906209 PMCID: PMC11790445 DOI: 10.3389/fcimb.2024.1529441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025] Open
Abstract
Mitochondrial genomic stability is critical to prevent various human inflammatory diseases. Bacterial infection significantly increases oxidative stress, driving mitochondrial genomic instability and initiating inflammatory human disease. Oxidative DNA base damage is predominantly repaired by base excision repair (BER) in the nucleus (nBER) as well as in the mitochondria (mtBER). In this review, we summarize the molecular mechanisms of spontaneous and H. pylori infection-associated oxidative mtDNA damage, mtDNA replication stress, and its impact on innate immune signaling. Additionally, we discuss how mutations located on mitochondria targeting sequence (MTS) of BER genes may contribute to mtDNA genome instability and innate immune signaling activation. Overall, the review summarizes evidence to understand the dynamics of mitochondria genome and the impact of mtBER in innate immune response during H. pylori-associated pathological outcomes.
Collapse
Affiliation(s)
| | - Dawit Kidane
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, United States
| |
Collapse
|
3
|
Zhou L, Zhuo H, Jin J, Pu A, Liu Q, Song J, Tong X, Tang H, Dai F. Temperature perception by ER UPR promotes preventive innate immunity and longevity. Cell Rep 2024; 43:115071. [PMID: 39675004 DOI: 10.1016/j.celrep.2024.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/02/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
Microbial infectivity increases with rising environmental temperature, heightening the risk of infection to host organisms. The host's basal immunity is activated accordingly to mitigate upcoming pathogenic threats; still, how animals sense temperature elevation to adjust their preventive immune response remains elusive. This study reports that high temperature enhances innate immunity differently from pathogen infection. Unlike pathogen invasion requiring the mitochondrial unfolded protein response (UPR), high temperature engages the endoplasmic reticulum (ER) UPR to trigger the innate immune response. Furthermore, chronic activation of the XBP-1 UPR branch represses nucleolar ribosome biogenesis, a highly energy-consuming process, leading to lipid accumulation. The subsequent increase in oleic acid promotes the activation of the PMK-1 immune pathway. Additionally, ribosome biogenesis was identified as a regulator of longevity, wherein its impact is dependent on lipid metabolism and innate immunity. Collectively, our findings reveal the crucial role of ER-nucleolus crosstalk in shaping preventive immune responses and lifespan regulation.
Collapse
Affiliation(s)
- Lei Zhou
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Haoyu Zhuo
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Jiaqi Jin
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Anrui Pu
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Qin Liu
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Jiangbo Song
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Xiaoling Tong
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Haiqing Tang
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| | - Fangyin Dai
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
4
|
Gao X, Feng S, Wu B, Liu L, Xu Y, Zhang J, Miao J. Staphylococcus aureus Conquers Host by Hijacking Mitochondria via PFKFB3 in Epithelial Cells. J Infect Dis 2024; 230:1488-1500. [PMID: 38805184 DOI: 10.1093/infdis/jiae263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/25/2024] [Indexed: 05/29/2024] Open
Abstract
Staphylococcus aureus persists within mammary epithelial cells for an extended duration, exploiting the host metabolic resources to facilitate replication. This study revealed a mechanism by which intracellular S aureus reprograms host metabolism, with PFKFB3 playing a crucial role in this process. Mechanistically, S aureus induced mitochondrial damage, leading to increased levels of mitochondrial reactive oxygen species and dysfunction in the electron transport chain. Moreover, S aureus shifted the balance of mitochondrial dynamics from fusion to fission, subsequently activating PINK1-PRKN-dependent mitophagy, causing loss of sirtuin 3 to stabilize hypoxic inducible factor 1α, and shifting the host metabolism toward enhanced glycolysis. The inhibition of PFKFB3 reversed the mitochondrial damage and degradation of sirtuin 3 induced by S aureus. Overall, our findings elucidate the mechanism by which S aureus reprograms host metabolism, thereby offering insights into the treatment of S aureus infection.
Collapse
Affiliation(s)
- Xing Gao
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Shiyuan Feng
- Sanya Research Institute, Nanjing Agricultural University, Sanya
| | - Binfeng Wu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Laizhen Liu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Yuanyuan Xu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Jinqiu Zhang
- Institute of Veterinary Immunology and Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing
- Jiangsu Key Laboratory for Food Quality and Safety, State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, China
| | - Jinfeng Miao
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| |
Collapse
|
5
|
Cheng C, Chen M, Sun J, Xu J, Deng S, Xia J, Han Y, Zhang X, Wang J, Lei L, Zhai R, Wu Q, Fang W, Song H. The MICOS Complex Subunit Mic60 is Hijacked by Intracellular Bacteria to Manipulate Mitochondrial Dynamics and Promote Bacterial Pathogenicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406760. [PMID: 39431455 PMCID: PMC11633497 DOI: 10.1002/advs.202406760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/20/2024] [Indexed: 10/22/2024]
Abstract
Host mitochondria undergo fission and fusion, which bacteria often exploit for their infections. In this study, the underlying molecular mechanisms are aimed to clarify through which Listeria monocytogenes (L. monocytogenes), a human bacterial pathogen, manipulates mitochondrial dynamics to enhance its pathogenicity. It is demonstrated that L. monocytogenes triggers transient mitochondrial fission through its virulence factor listeriolysin O (LLO), driven by LLO's interaction with Mic60, a core component of the mitochondrial contact site and the cristae organizing system (MICOS). Specifically, Phe251 within LLO is identify as a crucial residue for binding to Mic60, crucial for LLO-induced mitochondrial fragmentation and bacterial pathogenicity. Importantly, it is that Mic60 affect the formation of F-actin tails recruited by L. monocytogenes, thereby contributing to intracellular bacterial infection. Mic60 plays a critical role in mediating changes in mitochondrial morphology, membrane potential, and reactive oxidative species (ROS) production, and L. monocytogenes infection exacerbates these changes by affecting Mic60 expression. These findings unveil a novel mechanism through which intracellular bacteria exploit host mitochondria, shedding light on the complex interplay between hosts and microbes during infections. This knowledge holds promise for developing innovative strategies to combat bacterial infections.
Collapse
Affiliation(s)
- Changyong Cheng
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Mianmian Chen
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Jing Sun
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Jiali Xu
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Simin Deng
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Jing Xia
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Yue Han
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Xian Zhang
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Jing Wang
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Lei Lei
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Ruidong Zhai
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Qin Wu
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Weihuan Fang
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| | - Houhui Song
- Key Laboratory of Applied Technology on Green‐Eco‐Healthy Animal Husbandry of Zhejiang ProvinceZhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced TechnologyZhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health ManagementChina‐Australia Joint Laboratory for Animal Health Big Data AnalyticsCollege of Veterinary Medicine of Zhejiang A&F University666 Wusu Street, Lin'an DistrictHangzhouZhejiang Province311300China
| |
Collapse
|
6
|
Iebba V. Bacterial predators and BALOs: Growth protocol and relation with mitochondria. Methods Cell Biol 2024; 194:151-167. [PMID: 40058958 DOI: 10.1016/bs.mcb.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The microbial world is characterized by mechanisms of competition and predation, akin to the animal world. However, while predation's ecological role is well-established in animals, it's less understood in bacteria due to fewer known predators and unclear phylogenetic affiliations. Nevertheless, microorganisms can prey on bacterial cells, including Bacteriophages, Protists, and Predatory Prokaryotes. These predators inhabit various habitats and may play vital roles in bacterial ecology and ecosystem regulation. Predatory interactions between host and parasite are common in nature. Predatory bacteria, such as Bdellovibrio and like organisms (BALOs), employ various strategies, including epibiotic predation and direct invasion. BALOs, which thrive in the periplasmic space of Gram-negative bacterial cells, modulate bacterial populations and could serve as preventive or therapeutic agents against Gram-negative infections. While primarily active against extracellular prey, BALOs may also target mitochondria, which are crucial for cellular processes. The relationship between intracellular bacteria and host mitochondria, including morphology, function, and apoptosis, warrants further exploration. Protocols for growing, propagating, and detecting predatory activities of BALOs, particularly Bdellovibrio bacteriovorus, are provided to assess their presence and activities against potential prey.
Collapse
|
7
|
Wang J, Cui M, Liu Y, Chen M, Xu J, Xia J, Sun J, Jiang L, Fang W, Song H, Cheng C. The mitochondrial carboxylase PCCA interacts with Listeria monocytogenes phospholipase PlcB to modulate bacterial survival. Appl Environ Microbiol 2024; 90:e0213523. [PMID: 38727222 PMCID: PMC11218614 DOI: 10.1128/aem.02135-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/19/2024] [Indexed: 06/19/2024] Open
Abstract
Listeria monocytogenes, a prominent foodborne pathogen responsible for zoonotic infections, owes a significant portion of its virulence to the presence of the phospholipase PlcB. In this study, we performed an in-depth examination of the intricate relationship between L. monocytogenes PlcB and host cell mitochondria, unveiling a novel participant in bacterial survival: the mitochondrial carboxylase propionyl-coenzyme A carboxylase (PCCA). Our investigation uncovered previously unexplored levels of interaction and colocalization between PCCA and PlcB within host cells, with particular emphasis on the amino acids 504-508 of PCCA, which play a pivotal role in this partnership. To assess the effect of PCCA expression on L. monocytogenes proliferation, PCCA expression levels were manipulated by siRNA-si-PCCA or pCMV-N-HA-PCCA plasmid transfection. Our findings demonstrated a clear inverse correlation between PCCA expression levels and the proliferation of L. monocytogenes. Furthermore, the effect of L. monocytogenes infection on PCCA expression was investigated by assessing PCCA mRNA and protein expression in HeLa cells infected with L. monocytogenes. These results indicate that L. monocytogenes infection did not significantly alter PCCA expression. These findings led us to propose that PCCA represents a novel participant in L. monocytogenes survival, and its abundance has a detrimental impact on bacterial proliferation. This suggests that L. monocytogenes may employ PlcB-PCCA interactions to maintain stable PCCA expression, representing a unique pro-survival strategy distinct from that of other intracellular bacterial pathogens. IMPORTANCE Mitochondria represent attractive targets for pathogenic bacteria seeking to modulate host cellular processes to promote their survival and replication. Our current study has uncovered mitochondrial carboxylase propionyl-coenzyme A carboxylase (PCCA) as a novel host cell protein that interacts with L. monocytogenes PlcB. The results demonstrate that PCCA plays a negative regulatory role in L. monocytogenes infection, as heightened PCCA levels are associated with reduced bacterial survival and persistence. However, L. monocytogenes may exploit the PlcB-PCCA interaction to maintain stable PCCA expression and establish a favorable intracellular milieu for bacterial infection. Our findings shed new light on the intricate interplay between bacterial pathogens and host cell mitochondria, while also highlighting the potential of mitochondrial metabolic enzymes as antimicrobial agents.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Mingzhu Cui
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Yucong Liu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Mianmian Chen
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jiali Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jing Xia
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jing Sun
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Lingli Jiang
- Ningbo College of Health Sciences, Ningbo, Zhejiang, China
| | - Weihuan Fang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Houhui Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Changyong Cheng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Lu L, Qi Z, Chen Z, Wang H, Wei X, Zhao B, Wang Z, Shao Y, Tu J, Song X. Avian pathogenic Escherichia coli T6SS effector protein Hcp2a causes mitochondrial dysfunction through interaction with LETM1 protein in DF-1 cells. Poult Sci 2024; 103:103514. [PMID: 38367471 PMCID: PMC10879833 DOI: 10.1016/j.psj.2024.103514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/19/2024] Open
Abstract
The type VI secretion system (T6SS) of avian pathogenic Escherichia coli (APEC) can affect the functions of eukaryotic cells by secreting or injecting effectors. Hemolysin co-regulatory protein (Hcp), one of the markers of the T6SS, is both a structural protein and an effector protein of the T6SS. According to previous studies, mitochondria in eukaryotic cells are targeted by pathogenic bacteria. However, little is known about the regulation of mitochondria in eukaryotic host cells by the T6SS effector protein Hcp of APEC. In our study, DF-1 cells co-incubated with Hcp2a protein for 6 h showed decreased mitochondrial membrane potential, increased Ca2+ concentration, and increased cellular reactive oxygen species (ROS) levels. We therefore conclude that Hcp2a protein causes dysfunction to mitochondria in DF-1 cells. To explain the mechanism that causes mitochondrial dysfunction, we reanalyzed the Hcp2a interaction protein dataset in DF-1 cells, and the Leucine zipper EF-hand-containing transmembrane protein 1 (LETM1), which is associated with mitochondria, was screened. The protein and molecular docking results showed that Hcp2a protein and LETM1 protein have better binding. Finally, subcellular localization results showed that Hcp2a was localized to mitochondria. In summary, Hcp2a effector proteins caused dysfunction to DF-1 cellular mitochondria, and we hypothesize that the interaction of Hcp2a protein with LETM1 protein induces mitochondrial dysfunction and promotes mitochondrial localization of Hcp2a in DF-1 cells.
Collapse
Affiliation(s)
- Liting Lu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Zhao Qi
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Zhe Chen
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Haiyang Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Xiyang Wei
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Bingyu Zhao
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Zhenyu Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Ying Shao
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Jian Tu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, PR China
| | - Xiangjun Song
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, PR China.
| |
Collapse
|
9
|
Hartsoe P, Holguin F, Chu HW. Mitochondrial Dysfunction and Metabolic Reprogramming in Obesity and Asthma. Int J Mol Sci 2024; 25:2944. [PMID: 38474191 PMCID: PMC10931700 DOI: 10.3390/ijms25052944] [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: 01/01/2024] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Mitochondrial dysfunction and metabolic reprogramming have been extensively studied in many disorders ranging from cardiovascular to neurodegenerative disease. Obesity has previously been associated with mitochondrial fragmentation, dysregulated glycolysis, and oxidative phosphorylation, as well as increased reactive oxygen species production. Current treatments focus on reducing cellular stress to restore homeostasis through the use of antioxidants or alterations of mitochondrial dynamics. This review focuses on the role of mitochondrial dysfunction in obesity particularly for those suffering from asthma and examines mitochondrial transfer from mesenchymal stem cells to restore function as a potential therapy. Mitochondrial targeted therapy to restore healthy metabolism may provide a unique approach to alleviate dysregulation in individuals with this unique endotype.
Collapse
Affiliation(s)
- Paige Hartsoe
- Department of Medicine, National Jewish Health, Denver, CO 80222, USA
| | - Fernando Holguin
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO 80222, USA
| |
Collapse
|
10
|
Gao S, Gao L, Yuan D, Lin X, van der Veen S. Gonococcal OMV-delivered PorB induces epithelial cell mitophagy. Nat Commun 2024; 15:1669. [PMID: 38396029 PMCID: PMC10891091 DOI: 10.1038/s41467-024-45961-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The bacterial pathogen Neisseria gonorrhoeae is able to invade epithelial cells and survive intracellularly. During this process, it secretes outer membrane vesicles (OMVs), however, the mechanistic details for interactions between gonococcal OMVs and epithelial cells and their impact on intracellular survival are currently not established. Here, we show that gonococcal OMVs induce epithelial cell mitophagy to reduce mitochondrial secretion of reactive oxygen species (ROS) and enhance intracellular survival. We demonstrate that OMVs deliver PorB to mitochondria to dissipate the mitochondrial membrane potential, resulting in mitophagy induction through a conventional PINK1 and OPTN/NDP52 mechanism. Furthermore, PorB directly recruits the E3 ubiquitin ligase RNF213, which decorates PorB lysine residue 171 with K63-linked polyubiquitin to induce mitophagy in a p62-dependent manner. These results demonstrate a mechanism in which polyubiquitination of a bacterial virulence factor that targets mitochondria directs mitophagy processes to this organelle to prevent its secretion of deleterious ROS.
Collapse
Affiliation(s)
- Shuai Gao
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Lingyu Gao
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Dailin Yuan
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, PR China
| | - Xu'ai Lin
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Stijn van der Veen
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China.
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, PR China.
| |
Collapse
|
11
|
Candeias E, Pereira-Santos AR, Empadinhas N, Cardoso SM, Esteves ARF. The Gut-Brain Axis in Alzheimer's and Parkinson's Diseases: The Catalytic Role of Mitochondria. J Alzheimers Dis 2024; 100:413-429. [PMID: 38875045 DOI: 10.3233/jad-240524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Accumulating evidence suggests that gut inflammation is implicated in neuroinflammation in Alzheimer's and Parkinson's diseases. Despite the numerous connections it remains unclear how the gut and the brain communicate and whether gut dysbiosis is the cause or consequence of these pathologies. Importantly, several reports highlight the importance of mitochondria in the gut-brain axis, as well as in mechanisms like gut epithelium self-renewal, differentiation, and homeostasis. Herein we comprehensively address the important role of mitochondria as a cellular hub in infection and inflammation and as a link between inflammation and neurodegeneration in the gut-brain axis. The role of mitochondria in gut homeostasis and as well the crosstalk between mitochondria and gut microbiota is discussed. Significantly, we also review studies highlighting how gut microbiota can ultimately affect the central nervous system. Overall, this review summarizes novel findings regarding this cross-talk where the mitochondria has a main role in the pathophysiology of both Alzheimer's and Parkinson's disease strengthen by cellular, animal and clinical studies.
Collapse
Affiliation(s)
- Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Pereira-Santos
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sandra Morais Cardoso
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Fernandes Esteves
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
12
|
Tian Y, Zhang T, Li J, Tao Y. Advances in development of exosomes for ophthalmic therapeutics. Adv Drug Deliv Rev 2023; 199:114899. [PMID: 37236425 DOI: 10.1016/j.addr.2023.114899] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/18/2023] [Accepted: 05/21/2023] [Indexed: 05/28/2023]
Abstract
Exosomes contain multiple bioactive molecules and maintain the connection between cells. Recent advances in exosome-based therapeutics have witnessed unprecedented opportunities in treating ophthalmic diseases, including traumatic diseases, autoimmune diseases, chorioretinal diseases and others. Utilization of exosomes as delivery vectors to encapsulate both drugs and therapeutic genes could yield higher efficacy and avoid the unnecessary immune responses. However, exosome-based therapies also come with some potential ocular risks. In this review, we first present a general introduction to exosomes. Then we provide an overview of available applications and discuss their potential risks. Moreover, we review recently reported exosomes as delivery vectors for ophthalmic diseases. Finally, we put forward future perspectives to grapple with its translation and underlying issues.
Collapse
Affiliation(s)
- Ying Tian
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Tao Zhang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Jing Li
- Beijing Key Laboratory of DNA Damage Response, College of Life Sciences, Capital Normal University, Beijing 100048, PR China
| | - Yong Tao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China.
| |
Collapse
|
13
|
Verbeke J, Fayt Y, Martin L, Yilmaz O, Sedzicki J, Reboul A, Jadot M, Renard P, Dehio C, Renard H, Letesson J, De Bolle X, Arnould T. Host cell egress of Brucella abortus requires BNIP3L-mediated mitophagy. EMBO J 2023; 42:e112817. [PMID: 37232029 PMCID: PMC10350838 DOI: 10.15252/embj.2022112817] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
The facultative intracellular pathogen Brucella abortus interacts with several organelles of the host cell to reach its replicative niche inside the endoplasmic reticulum. However, little is known about the interplay between the intracellular bacteria and the host cell mitochondria. Here, we showed that B. abortus triggers substantive mitochondrial network fragmentation, accompanied by mitophagy and the formation of mitochondrial Brucella-containing vacuoles during the late steps of cellular infection. Brucella-induced expression of the mitophagy receptor BNIP3L is essential for these events and relies on the iron-dependent stabilisation of the hypoxia-inducible factor 1α. Functionally, BNIP3L-mediated mitophagy appears to be advantageous for bacterial exit from the host cell as BNIP3L depletion drastically reduces the number of reinfection events. Altogether, these findings highlight the intricate link between Brucella trafficking and the mitochondria during host cell infection.
Collapse
Affiliation(s)
- Jérémy Verbeke
- Research Unit in Cell Biology (URBC)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Youri Fayt
- Research Unit in Cell Biology (URBC)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Lisa Martin
- Research Unit in Cell Biology (URBC)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Oya Yilmaz
- Research Unit in Cell Biology (URBC)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | | | - Angéline Reboul
- Research Unit in Microorganisms Biology (URBM)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Michel Jadot
- Research Unit in Molecular Physiology (URPhyM)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Patricia Renard
- Research Unit in Cell Biology (URBC)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | | | - Henri‐François Renard
- Research Unit in Cell Biology (URBC)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Jean‐Jacques Letesson
- Research Unit in Microorganisms Biology (URBM)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Xavier De Bolle
- Research Unit in Microorganisms Biology (URBM)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| | - Thierry Arnould
- Research Unit in Cell Biology (URBC)—Namur Research Institute for Life Sciences (NARILIS)University of NamurNamurBelgium
| |
Collapse
|
14
|
Uzum Z, Ershov D, Pavia MJ, Mallet A, Gorgette O, Plantard O, Sassera D, Stavru F. Three-dimensional images reveal the impact of the endosymbiont Midichloria mitochondrii on the host mitochondria. Nat Commun 2023; 14:4133. [PMID: 37438329 DOI: 10.1038/s41467-023-39758-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
The hard tick, Ixodes ricinus, a main Lyme disease vector, harbors an intracellular bacterial endosymbiont. Midichloria mitochondrii is maternally inherited and resides in the mitochondria of I. ricinus oocytes, but the consequences of this endosymbiosis are not well understood. Here, we provide 3D images of wild-type and aposymbiotic I. ricinus oocytes generated with focused ion beam-scanning electron microscopy. Quantitative image analyses of endosymbionts and oocyte mitochondria at different maturation stages show that the populations of both mitochondrion-associated bacteria and bacterium-hosting mitochondria increase upon vitellogenisation, and that mitochondria can host multiple bacteria in later stages. Three-dimensional reconstructions show symbiosis-dependent morphologies of mitochondria and demonstrate complete M. mitochondrii inclusion inside a mitochondrion. Cytoplasmic endosymbiont located close to mitochondria are not oriented towards the mitochondria, suggesting that bacterial recolonization is unlikely. We further demonstrate individual globular-shaped mitochondria in the wild type oocytes, while aposymbiotic oocytes only contain a mitochondrial network. In summary, our study suggests that M. mitochondrii modulates mitochondrial fragmentation in oogenesis possibly affecting organelle function and ensuring its presence over generations.
Collapse
Affiliation(s)
- Zerrin Uzum
- Unit Evolutionary Biology of the Microbial Cell, Institut Pasteur; CNRS UMR2001, Paris, France.
| | - Dmitry Ershov
- Image Analysis Hub, Cell Biology and Infection Department, Institut Pasteur, Paris, France
- Bioinformatics and Biostatistics HUB, Department of Computational Biology, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Michael J Pavia
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Adeline Mallet
- Ultrastructural BioImaging Core Facility, Institut Pasteur, Paris, France
| | - Olivier Gorgette
- Ultrastructural BioImaging Core Facility, Institut Pasteur, Paris, France
| | | | - Davide Sassera
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Fabrizia Stavru
- Unit Evolutionary Biology of the Microbial Cell, Institut Pasteur; CNRS UMR2001, Paris, France
| |
Collapse
|
15
|
Martin M, deVisch A, Boudehen YM, Barthe P, Gutierrez C, Turapov O, Aydogan T, Heriaud L, Gracy J, Neyrolles O, Mukamolova GV, Letourneur F, Cohen-Gonsaud M. A Mycobacterium tuberculosis Effector Targets Mitochondrion, Controls Energy Metabolism, and Limits Cytochrome c Exit. Microbiol Spectr 2023; 11:e0106623. [PMID: 37036353 PMCID: PMC10269737 DOI: 10.1128/spectrum.01066-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 04/11/2023] Open
Abstract
Host metabolism reprogramming is a key feature of Mycobacterium tuberculosis (Mtb) infection that enables the survival of this pathogen within phagocytic cells and modulates the immune response facilitating the spread of the tuberculosis disease. Here, we demonstrate that a previously uncharacterized secreted protein from Mtb, Rv1813c, manipulates the host metabolism by targeting mitochondria. When expressed in eukaryotic cells, the protein is delivered to the mitochondrial intermembrane space and promotes the enhancement of host ATP production by boosting the oxidative phosphorylation metabolic pathway. Furthermore, the release of cytochrome c from mitochondria, an early apoptotic event in response to short-term oxidative stress, is delayed in Rv1813c-expressing cells. This study reveals a novel class of mitochondria targeting effectors from Mtb that might participate in host cell metabolic reprogramming and apoptosis control during Mtb infections. IMPORTANCE In this article, using a combination of techniques (bioinformatics, structural biology, and cell biology), we identified and characterized a new class of effectors present only in intracellular mycobacteria. These proteins specifically target host cell mitochondria when ectopically expressed in cells. We showed that one member of this family (Rv1813c) affects mitochondria metabolism in a way that might twist the immune response. This effector also inhibits the cytochrome c exit from mitochondria, suggesting that it might alter normal host cell apoptotic capacities, one of the first defenses of immune cells against Mtb infection.
Collapse
Affiliation(s)
- Marianne Martin
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Angelique deVisch
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Yves-Marie Boudehen
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Philippe Barthe
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Claude Gutierrez
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Obolbek Turapov
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Talip Aydogan
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Laurène Heriaud
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Jerome Gracy
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Galina V. Mukamolova
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - François Letourneur
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Martin Cohen-Gonsaud
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
16
|
Zhang Y, Zhang J, Duan L. The role of microbiota-mitochondria crosstalk in pathogenesis and therapy of intestinal diseases. Pharmacol Res 2022; 186:106530. [DOI: 10.1016/j.phrs.2022.106530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/17/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
|
17
|
Infection Dynamics of Cotransmitted Reproductive Symbionts Are Mediated by Sex, Tissue, and Development. Appl Environ Microbiol 2022; 88:e0052922. [PMID: 35730939 PMCID: PMC9275221 DOI: 10.1128/aem.00529-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
One of the most prevalent intracellular infections on earth is with Wolbachia, a bacterium in the Rickettsiales that infects a range of insects, crustaceans, chelicerates, and nematodes. Wolbachia is maternally transmitted to offspring and has profound effects on the reproduction and physiology of its hosts, which can result in reproductive isolation, altered vectorial capacity, mitochondrial sweeps, and even host speciation. Some populations stably harbor multiple Wolbachia strains, which can further contribute to reproductive isolation and altered host physiology. However, almost nothing is known about the requirements for multiple intracellular microbes to be stably maintained across generations while they likely compete for space and resources. Here, we use a coinfection of two Wolbachia strains (“wHa” and “wNo”) in Drosophila simulans to define the infection and transmission dynamics of an evolutionarily stable double infection. We find that a combination of sex, tissue, and host development contributes to the infection dynamics of the two microbes and that these infections exhibit a degree of niche partitioning across host tissues. wHa is present at a significantly higher titer than wNo in most tissues and developmental stages, but wNo is uniquely dominant in ovaries. Unexpectedly, the ratio of wHa to wNo in embryos does not reflect those observed in the ovaries, indicative of strain-specific transmission dynamics. Understanding how Wolbachia strains interact to establish and maintain stable infections has important implications for the development and effective implementation of Wolbachia-based vector biocontrol strategies, as well as more broadly defining how cooperation and conflict shape intracellular communities. IMPORTANCEWolbachia is a maternally transmitted intracellular bacterium that manipulates the reproduction and physiology of arthropods, resulting in drastic effects on the fitness, evolution, and even speciation of its hosts. Some hosts naturally harbor multiple strains of Wolbachia that are stably transmitted across generations, but almost nothing is known about the factors that limit or promote these coinfections, which can have profound effects on the host’s biology and evolution and are under consideration as an insect-management tool. Here, we define the infection dynamics of a known stably transmitted double infection in Drosophila simulans with an eye toward understanding the patterns of infection that might facilitate compatibility between the two microbes. We find that a combination of sex, tissue, and development all contributes to infection dynamics of the coinfection.
Collapse
|
18
|
Mahanta DK, Jangra S, Priti, Ghosh A, Sharma PK, Iquebal MA, Jaiswal S, Baranwal VK, Kalia VK, Chander S. Groundnut Bud Necrosis Virus Modulates the Expression of Innate Immune, Endocytosis, and Cuticle Development-Associated Genes to Circulate and Propagate in Its Vector, Thrips palmi. Front Microbiol 2022; 13:773238. [PMID: 35369489 PMCID: PMC8969747 DOI: 10.3389/fmicb.2022.773238] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Thrips palmi (Thysanoptera: Thripidae) is the predominant tospovirus vector in Asia-Pacific region. It transmits economically damaging groundnut bud necrosis virus (GBNV, family Tospoviridae) in a persistent propagative manner. Thrips serve as the alternate host, and virus reservoirs making tospovirus management very challenging. Insecticides and host plant resistance remain ineffective in managing thrips–tospoviruses. Recent genomic approaches have led to understanding the molecular interactions of thrips–tospoviruses and identifying novel genetic targets. However, most of the studies are limited to Frankliniella species and tomato spotted wilt virus (TSWV). Amidst the limited information available on T. palmi–tospovirus relationships, the present study is the first report of the transcriptome-wide response of T. palmi associated with GBNV infection. The differential expression analyses of the triplicate transcriptome of viruliferous vs. nonviruliferous adult T. palmi identified a total of 2,363 (1,383 upregulated and 980 downregulated) significant transcripts. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses showed the abundance of differentially expressed genes (DEGs) involved in innate immune response, endocytosis, cuticle development, and receptor binding and signaling that mediate the virus invasion and multiplication in the vector system. Also, the gene regulatory network (GRN) of most significant DEGs showed the genes like ABC transporter, cytochrome P450, endocuticle structural glycoprotein, gamma-aminobutyric acid (GABA) receptor, heat shock protein 70, larval and pupal cuticle proteins, nephrin, proline-rich protein, sperm-associated antigen, UHRF1-binding protein, serpin, tyrosine–protein kinase receptor, etc., were enriched with higher degrees of interactions. Further, the expression of the candidate genes in response to GBNV infection was validated in reverse transcriptase-quantitative real-time PCR (RT-qPCR). This study leads to an understanding of molecular interactions between T. palmi and GBNV and suggests potential genetic targets for generic pest control.
Collapse
|
19
|
Mourenza Á, Lorente-Torres B, Durante E, Llano-Verdeja J, Aparicio JF, Fernández-López A, Gil JA, Mateos LM, Letek M. Understanding microRNAs in the Context of Infection to Find New Treatments against Human Bacterial Pathogens. Antibiotics (Basel) 2022; 11:356. [PMID: 35326819 PMCID: PMC8944844 DOI: 10.3390/antibiotics11030356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
The development of RNA-based anti-infectives has gained interest with the successful application of mRNA-based vaccines. Small RNAs are molecules of RNA of <200 nucleotides in length that may control the expression of specific genes. Small RNAs include small interference RNAs (siRNAs), Piwi-interacting RNAs (piRNAs), or microRNAs (miRNAs). Notably, the role of miRNAs on the post-transcriptional regulation of gene expression has been studied in detail in the context of cancer and many other genetic diseases. However, it is also becoming apparent that some human miRNAs possess important antimicrobial roles by silencing host genes essential for the progress of bacterial or viral infections. Therefore, their potential use as novel antimicrobial therapies has gained interest during the last decade. The challenges of the transport and delivery of miRNAs to target cells are important, but recent research with exosomes is overcoming the limitations in RNA-cellular uptake, avoiding their degradation. Therefore, in this review, we have summarised the latest developments in the exosomal delivery of miRNA-based therapies, which may soon be another complementary treatment to pathogen-targeted antibiotics that could help solve the problem caused by multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Álvaro Mourenza
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Blanca Lorente-Torres
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Elena Durante
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- L’Università di Urbino Carlo Bo, Via Aurelio Saffi, 2, 61029 Urbino, Italy
| | - Jesús Llano-Verdeja
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Jesús F. Aparicio
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Arsenio Fernández-López
- Departamento de Biología Molecular, Área de Biología Celular, Universidad de León, 24071 León, Spain;
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Neural Therapies SL, Campus de Vegazana s/n, 24071 León, Spain
| | - José A. Gil
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Luis M. Mateos
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Michal Letek
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
| |
Collapse
|
20
|
Zhao W, He F, Barkema HW, Xu S, Gao J, Liu G, Deng Z, Shahid M, Shi Y, Kastelic JP, Han B. Prototheca spp. induce an inflammatory response via mtROS-mediated activation of NF-κB and NLRP3 inflammasome pathways in bovine mammary epithelial cell cultures. Vet Res 2021; 52:144. [PMID: 34895324 PMCID: PMC8666081 DOI: 10.1186/s13567-021-01014-9] [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: 08/30/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Emergence of bovine mastitis caused by Prototheca algae is the impetus to better understand these infections. Both P. bovis and P. ciferrii belong to Prototheca algae, but they differ in their pathogenicity to induce inflammatory responses. The objective was to characterize and compare pathogenesis of inflammatory responses in bMECs induced by P. bovis versus P. ciferrii. Mitochondrial ultrastructure, activity and mtROS in bMECs were assessed with transmission electron microscopy and laser scanning confocal microscopy. Cytokines, including TNF-α, IL-1β and IL-18, were measured by ELISA and real-time PCR, whereas expressions of various proteins in the NF-κB and NLRP3 inflammasome pathways were detected with immunofluorescence or Western blot. Infection with P. bovis or P. ciferrii damaged mitochondria, including dissolution and vacuolation of cristae, and decreased mitochondrial activity, with P. bovis being more pathogenic and causing greater destruction. There were increases in NADPH production and mtROS accumulation in infected bMECs, with P. bovis causing greater increases and also inducing higher cytokine concentrations. Expressions of NF-κB-p65, p-NF-κB-p65, IκBα and p-IκBα proteins in the NF-κB pathway, as well as NLRP3, Pro Caspase1, Caspase1 p20, ASC, Pro IL-1β, and IL-1β proteins in the NLRP3 inflammasome pathway, were significantly higher in P. bovis-infected bMECs. However, mito-TEMPO significantly inhibited production of cytokines and decreased expression of proteins in NF-κB and NLRP3 inflammasome pathways in bMECs infected with either P. bovis or P. ciferrii. In conclusion, P. bovis or P. ciferrii infections induced inflammatory responses in bMECs, with increased mtROS in damaged mitochondria and activated NF-κB and NLRP3 inflammasome pathways, with P. bovis causing a more severe reaction.
Collapse
Affiliation(s)
- Wenpeng Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Fumeng He
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Herman W Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Siyu Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Gang Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Zhaoju Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Muhammad Shahid
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yuxiang Shi
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, 056038, Hebei, China
| | - John P Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
21
|
Escoll P, Platon L, Dramé M, Sahr T, Schmidt S, Rusniok C, Buchrieser C. Reverting the mode of action of the mitochondrial F OF 1-ATPase by Legionella pneumophila preserves its replication niche. eLife 2021; 10:e71978. [PMID: 34882089 PMCID: PMC8718111 DOI: 10.7554/elife.71978] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/06/2021] [Indexed: 01/17/2023] Open
Abstract
Legionella pneumophila, the causative agent of Legionnaires' disease, a severe pneumonia, injects via a type 4 secretion system (T4SS) more than 300 proteins into macrophages, its main host cell in humans. Certain of these proteins are implicated in reprogramming the metabolism of infected cells by reducing mitochondrial oxidative phosphorylation (OXPHOS) early after infection. Here. we show that despite reduced OXPHOS, the mitochondrial membrane potential (Δψm) is maintained during infection of primary human monocyte-derived macrophages (hMDMs). We reveal that L. pneumophila reverses the ATP-synthase activity of the mitochondrial FOF1-ATPase to ATP-hydrolase activity in a T4SS-dependent manner, which leads to a conservation of the Δψm, preserves mitochondrial polarization, and prevents macrophage cell death. Analyses of T4SS effectors known to target mitochondrial functions revealed that LpSpl is partially involved in conserving the Δψm, but not LncP and MitF. The inhibition of the L. pneumophila-induced 'reverse mode' of the FOF1-ATPase collapsed the Δψm and caused cell death in infected cells. Single-cell analyses suggested that bacterial replication occurs preferentially in hMDMs that conserved the Δψm and showed delayed cell death. This direct manipulation of the mode of activity of the FOF1-ATPase is a newly identified feature of L. pneumophila allowing to delay host cell death and thereby to preserve the bacterial replication niche during infection.
Collapse
Affiliation(s)
- Pedro Escoll
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR 3525ParisFrance
| | - Lucien Platon
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR 3525ParisFrance
- Faculté des Sciences, Université de MontpellierMontpellierFrance
| | - Mariatou Dramé
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR 3525ParisFrance
- Faculté des Sciences, Université de ParisParisFrance
| | - Tobias Sahr
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR 3525ParisFrance
| | - Silke Schmidt
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR 3525ParisFrance
- Sorbonne Université, Collège doctoralParisFrance
| | - Christophe Rusniok
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR 3525ParisFrance
| | - Carmen Buchrieser
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR 3525ParisFrance
| |
Collapse
|
22
|
Spier A, Connor MG, Steiner T, Carvalho F, Cossart P, Eisenreich W, Wai T, Stavru F. Mitochondrial respiration restricts Listeria monocytogenes infection by slowing down host cell receptor recycling. Cell Rep 2021; 37:109989. [PMID: 34758302 PMCID: PMC8595641 DOI: 10.1016/j.celrep.2021.109989] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/26/2021] [Accepted: 10/21/2021] [Indexed: 01/06/2023] Open
Abstract
Mutations in mitochondrial genes impairing energy production cause mitochondrial diseases (MDs), and clinical studies have shown that MD patients are prone to bacterial infections. However, the relationship between mitochondrial (dys)function and infection remains largely unexplored, especially in epithelial cells, the first barrier to many pathogens. Here, we generate an epithelial cell model for one of the most common mitochondrial diseases, Leigh syndrome, by deleting surfeit locus protein 1 (SURF1), an assembly factor for respiratory chain complex IV. We use this genetic model and a complementary, nutrient-based approach to modulate mitochondrial respiration rates and show that impaired mitochondrial respiration favors entry of the human pathogen Listeria monocytogenes, a well-established bacterial infection model. Reversely, enhanced mitochondrial energy metabolism decreases infection efficiency. We further demonstrate that endocytic recycling is reduced in mitochondrial respiration-dependent cells, dampening L. monocytogenes infection by slowing the recycling of its host cell receptor c-Met, highlighting a previously undescribed role of mitochondrial respiration during infection. Enhanced mitochondrial respiration decreases L. monocytogenes infection Bacterial entry is affected by the host cell metabolism Mitochondrial respiration restricts host cell receptor recycling and thus infection
Collapse
Affiliation(s)
- Anna Spier
- Evolutionary Biology of the Microbial Cell Unit, Institut Pasteur, Paris, France; Bacteria-Cell Interactions Unit, Institut Pasteur, Paris, France; Université de Paris, Paris, France; UMR2001, CNRS, Paris, France
| | - Michael G Connor
- Université de Paris, Paris, France; Chromatin and Infection Unit, Institut Pasteur, Paris, France
| | - Thomas Steiner
- Bavarian NMR Center - Structural Membrane Biochemistry, Department of Chemistry, Technische Universität München, Garching, Germany
| | - Filipe Carvalho
- Bacteria-Cell Interactions Unit, Institut Pasteur, Paris, France
| | - Pascale Cossart
- Bacteria-Cell Interactions Unit, Institut Pasteur, Paris, France; Université de Paris, Paris, France.
| | - Wolfgang Eisenreich
- Bavarian NMR Center - Structural Membrane Biochemistry, Department of Chemistry, Technische Universität München, Garching, Germany
| | - Timothy Wai
- Université de Paris, Paris, France; Mitochondrial Biology Unit, Institut Pasteur, Paris, France.
| | - Fabrizia Stavru
- Evolutionary Biology of the Microbial Cell Unit, Institut Pasteur, Paris, France; Bacteria-Cell Interactions Unit, Institut Pasteur, Paris, France; Université de Paris, Paris, France; UMR2001, CNRS, Paris, France
| |
Collapse
|
23
|
Mitochondria as a Cellular Hub in Infection and Inflammation. Int J Mol Sci 2021; 22:ijms222111338. [PMID: 34768767 PMCID: PMC8583510 DOI: 10.3390/ijms222111338] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are the energy center of the cell. They are found in the cell cytoplasm as dynamic networks where they adapt energy production based on the cell’s needs. They are also at the center of the proinflammatory response and have essential roles in the response against pathogenic infections. Mitochondria are a major site for production of Reactive Oxygen Species (ROS; or free radicals), which are essential to fight infection. However, excessive and uncontrolled production can become deleterious to the cell, leading to mitochondrial and tissue damage. Pathogens exploit the role of mitochondria during infection by affecting the oxidative phosphorylation mechanism (OXPHOS), mitochondrial network and disrupting the communication between the nucleus and the mitochondria. The role of mitochondria in these biological processes makes these organelle good targets for the development of therapeutic strategies. In this review, we presented a summary of the endosymbiotic origin of mitochondria and their involvement in the pathogen response, as well as the potential promising mitochondrial targets for the fight against infectious diseases and chronic inflammatory diseases.
Collapse
|
24
|
Wang W, Hou S, Chen G, Xia L, Chen J, Wang Z, Lu Y. Characterization and function study of a glutamyl endopeptidase homolog from Nocardia seriolae. JOURNAL OF FISH DISEASES 2021; 44:813-821. [PMID: 33245800 DOI: 10.1111/jfd.13311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 06/11/2023]
Abstract
Glutamic endopeptidases (Glu), belonging to the class of serine proteases, are a subfamily of chymotrypsin-like proteolytic enzymes, which are regarded as important virulence factors in bacteria. However, the roles of glutamic endopeptidases of Nocardia seriolae in pathogenic process still remain uncertain. Here, a glutamic endopeptidase homolog from N. seriolae (GluNS) was cloned and its function was elucidated. GluNS encoded a 414-aa protein which shared 93% identity to N. concava. In the phylogenetic tree, the glutamic endopeptidases of genus Nocardia clustered together firstly and then clustered with Streptomyces species. Moreover, GluNS was identified to be a secreted protein of N. seriolae and localized in the mitochondria of FHM cells. The transient overexpression of GluNS significantly induced increase in caspase-3 activity and decrease in ΔΨm values in FHM cells. The number of apoptotic bodies was remarkably higher than that in control group. Taken together, GluNS overexpression induced apoptotic characteristics in FHM cells. This study provided new insights into the function of glutamic endopeptidase from N. seriolae.
Collapse
Affiliation(s)
- Wenji Wang
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Suying Hou
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Guoquan Chen
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Liqun Xia
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning, China
| | - Jianlin Chen
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Zhiwen Wang
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Yishan Lu
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning, China
| |
Collapse
|
25
|
Wang H, Xiao Q, Wei C, Chen H, Chen X, Dai C, Wen J, Ma C, Tu J, Fu T, Shen J, Yi B. A mitochondria-localized pentatricopeptide repeat protein is required to restore hau cytoplasmic male sterility in Brassica napus. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2021; 134:1377-1386. [PMID: 33725137 DOI: 10.1007/s00122-021-03777-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 06/12/2023]
Abstract
A mitochondria-localized pentatricopeptide repeat protein was identified by positional cloning and transferred into the hau CMS line, where it successfully restored fertility Cytoplasmic male sterility (CMS) is a maternally inherited trait that can be controlled by restorer-of-fertility (Rf) genes present in the nucleus. The hau CMS was identified as a new form of CMS associated with the mitochondrial transcript orf288; however, a lack of a restorer gene has limited its utilization in Brassica crops. Here, the combination of Brassica 60 K array with bulk segregant analysis and map-based cloning was used to delimit the Rfh locus to an 82.2-kb region on chromosome A09. A candidate gene encoding a mitochondria-localized pentatricopeptide repeat (PPR) protein was identified and transferred into the hau CMS line, where it successfully restored the fertility of the hau CMS plants. Furthermore, the expression analysis showed that Rfh was highly expressed in the flower buds, and the sequence analysis results implied that functional divergence between RFH and rfh could be due to 59 amino acid residue differences in the deduced protein sequences. In addition, a co-separated molecular marker was developed based on the divergent sequences between the dominant and recessive alleles. These results will help enable the heterosis of Brassica crops in the future.
Collapse
Affiliation(s)
- Huadong Wang
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Qing Xiao
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Chao Wei
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
- College of Life Science, Zhaoqing University, Zhaoqing, 526061, People's Republic of China
| | - Hui Chen
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xiaohan Chen
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Cheng Dai
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jing Wen
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Chaozhi Ma
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jinxing Tu
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Tingdong Fu
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jinxiong Shen
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| | - Bin Yi
- National Key Laboratory of Crop Genetic Improvement, National Centre of Rapeseed Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
26
|
Stavru F, Riemer J, Jex A, Sassera D. When bacteria meet mitochondria: The strange case of the tick symbiont Midichloria mitochondrii †. Cell Microbiol 2021; 22:e13189. [PMID: 32185904 DOI: 10.1111/cmi.13189] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/28/2022]
Abstract
Mitochondria are key eukaryotic organelles that perform several essential functions. Not surprisingly, many intracellular bacteria directly or indirectly target mitochondria, interfering with innate immunity, energy production or apoptosis, to make the host cell a more hospitable niche for bacterial replication. The alphaproteobacterium Midichloria mitochondrii has taken mitochondrial targeting to another level by physically colonising mitochondria, as shown by transmission electron micrographs of bacteria residing in the mitochondrial intermembrane space. This unique localization provokes a number of questions around the mechanisms allowing, and reasons driving intramitochondrial tropism. We suggest possible scenarios that could lead to this peculiar localization and hypothesize potential costs and benefits of mitochondrial colonisation for the bacterium and its host.
Collapse
Affiliation(s)
- Fabrizia Stavru
- Unité de Biologie Evolutive de la Cellule Microbienne, Institut Pasteur, Paris, France.,CNRS ERL6002, Paris, France
| | - Jan Riemer
- Department for Chemistry, Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Aaron Jex
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia.,Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Davide Sassera
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
27
|
Patrick KL, Watson RO. Mitochondria: Powering the Innate Immune Response to Mycobacterium tuberculosis Infection. Infect Immun 2021; 89:e00687-20. [PMID: 33558322 PMCID: PMC8090963 DOI: 10.1128/iai.00687-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Within the last decade, we have learned that damaged mitochondria activate many of the same innate immune pathways that evolved to sense and respond to intracellular pathogens. These shared responses include cytosolic nucleic acid sensing and type I interferon (IFN) expression, inflammasome activation that leads to pyroptosis, and selective autophagy (called mitophagy when mitochondria are the cargo). Because mitochondria were once bacteria, parallels between how cells respond to mitochondrial and bacterial ligands are not altogether surprising. However, the potential for cross talk or synergy between bacterium- and mitochondrion-driven innate immune responses during infection remains poorly understood. This interplay is particularly striking, and intriguing, in the context of infection with the intracellular bacterial pathogen Mycobacterium tuberculosis (Mtb). Multiple studies point to a role for Mtb infection and/or specific Mtb virulence factors in disrupting the mitochondrial network in macrophages, leading to metabolic changes and triggering potent innate immune responses. Research from our laboratories and others argues that mutations in mitochondrial genes can exacerbate mycobacterial disease severity by hyperactivating innate responses or activating them at the wrong time. Indeed, growing evidence supports a model whereby different mitochondrial defects or mutations alter Mtb infection outcomes in distinct ways. By synthesizing the current literature in this minireview, we hope to gain insight into the molecular mechanisms driving, and consequences of, mitochondrion-dependent immune polarization so that we might better predict tuberculosis patient outcomes and develop host-directed therapeutics designed to correct these imbalances.
Collapse
Affiliation(s)
- Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
| |
Collapse
|
28
|
Cheng J, Zhang J, Yang J, Yi B, Liu G, Zhou M, Kastelic JP, Han B, Gao J. Klebsiella pneumoniae infection causes mitochondrial damage and dysfunction in bovine mammary epithelial cells. Vet Res 2021; 52:17. [PMID: 33568211 PMCID: PMC7876824 DOI: 10.1186/s13567-021-00898-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/15/2021] [Indexed: 11/10/2022] Open
Abstract
Klebsiella pneumoniae, an important cause of bovine mastitis worldwide, is strongly pathogenic to bovine mammary epithelial cells (bMECs). Our objective was to determine the role of mitochondrial damage in the pathogenicity of K. pneumoniae on bMECs, by assessing several classical indicators of mitochondrial dysfunction, as well as differentially expressed genes (DEGs). Two K. pneumoniae strains (HLJ-D2 and HB-AF5), isolated from cows with clinical mastitis (CM), were used to infect bMECs (MAC-T line) cultured in vitro. In whole-transcriptome analysis of bMECs at 6 h post-infection (hpi), there were 3453 up-regulated and 3470 down-regulated genes for HLJ-D2, whereas for HB-AF5, there were 2891 up-regulated and 3278 down-regulated genes (P < 0.05). Based on GO term enrichment of differentially expressed genes (DEGs), relative to the controls, the primary categories altered in K. pneumoniae-infected bMECs included cellular macromolecule metabolism, metabolic process, binding, molecular function, etc. Infections increased (P < 0.05) malondialdehyde concentrations and formation of reactive oxygen species in bMECs. Additionally, both bacterial strains decreased (P < 0.05) total antioxidant capacity in bMECs at 6 and 12 hpi. Furthermore, infections decreased (P < 0.05) mitochondrial membrane potential and increased (P < 0.01) mitochondrial calcium concentrations. Finally, severe mitochondrial swelling and vacuolation, as well as mitochondrial rupture and cristae degeneration, were detected in infected bMECs. In conclusion, K. pneumoniae infections induced profound mitochondrial damage and dysfunction in bMECs; we inferred that this caused cellular damage and contributes to the pathogenesis of K. pneumoniae-induced CM in dairy cows.
Collapse
Affiliation(s)
- Jia Cheng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jv Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jingyue Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Bing Yi
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Gang Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Man Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - John P Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
29
|
Tiku V, Kofoed EM, Yan D, Kang J, Xu M, Reichelt M, Dikic I, Tan MW. Outer membrane vesicles containing OmpA induce mitochondrial fragmentation to promote pathogenesis of Acinetobacter baumannii. Sci Rep 2021; 11:618. [PMID: 33436835 PMCID: PMC7804284 DOI: 10.1038/s41598-020-79966-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Acinetobacter baumannii is a highly antibiotic resistant Gram-negative bacterium that causes life-threatening infections in humans with a very high mortality rate. A. baumannii is an extracellular pathogen with poorly understood virulence mechanisms. Here we report that A. baumannii employs the release of outer membrane vesicles (OMVs) containing the outer membrane protein A (OmpAAb) to promote bacterial pathogenesis and dissemination. OMVs containing OmpAAb are taken up by mammalian cells where they activate the host GTPase dynamin-related protein 1 (DRP1). OmpAAb mediated activation of DRP1 enhances its accumulation on mitochondria that causes mitochondrial fragmentation, elevation in reactive oxygen species (ROS) production and cell death. Loss of DRP1 rescues these phenotypes. Our data show that OmpAAb is sufficient to induce mitochondrial fragmentation and cytotoxicity since its expression in E. coli transfers its pathogenic properties to E. coli. A. baumannii infection in mice also induces mitochondrial damage in alveolar macrophages in an OmpAAb dependent manner. We finally show that OmpAAb is also required for systemic dissemination in the mouse lung infection model. In this study we uncover the mechanism of OmpAAb as a virulence factor in A. baumannii infections and further establish the host cell factor required for its pathogenic effects.
Collapse
Affiliation(s)
- Varnesh Tiku
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Eric M Kofoed
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Donghong Yan
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jing Kang
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Min Xu
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Mike Reichelt
- Department of Pathology, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Ivan Dikic
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern- Kai 7, 60590, Frankfurt am Main, Germany.
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
- Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany.
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
30
|
Fielden LF, Scott NE, Palmer CS, Khoo CA, Newton HJ, Stojanovski D. Proteomic Identification of Coxiella burnetii Effector Proteins Targeted to the Host Cell Mitochondria During Infection. Mol Cell Proteomics 2020; 20:100005. [PMID: 33177156 PMCID: PMC7950127 DOI: 10.1074/mcp.ra120.002370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/11/2020] [Indexed: 11/06/2022] Open
Abstract
Modulation of the host cell is integral to the survival and replication of microbial pathogens. Several intracellular bacterial pathogens deliver bacterial proteins, termed "effector proteins" into the host cell during infection by sophisticated protein translocation systems, which manipulate cellular processes and functions. The functional contribution of individual effectors is poorly characterized, particularly in intracellular bacterial pathogens with large effector protein repertoires. Technical caveats have limited the capacity to study these proteins during a native infection, with many effector proteins having only been demonstrated to be translocated during over-expression of tagged versions. Here, we developed a novel strategy to examine effector proteins in the context of infection. We coupled a broad, unbiased proteomics-based screen with organelle purification to study the host-pathogen interactions occurring between the host cell mitochondrion and the Gram-negative, Q fever pathogen Coxiella burnetii. We identify four novel mitochondrially-targeted C. burnetii effector proteins, renamed Mitochondrial Coxiella effector protein (Mce) B to E. Examination of the subcellular localization of ectopically expressed proteins confirmed their mitochondrial localization, demonstrating the robustness of our approach. Subsequent biochemical analysis and affinity enrichment proteomics of one of these effector proteins, MceC, revealed the protein localizes to the inner membrane and can interact with components of the mitochondrial quality control machinery. Our study adapts high-sensitivity proteomics to study intracellular host-pathogen interactions, providing a robust strategy to examine the subcellular localization of effector proteins during native infection. This approach could be applied to a range of pathogens and host cell compartments to provide a rich map of effector dynamics throughout infection.
Collapse
Affiliation(s)
- Laura F Fielden
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Catherine S Palmer
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Chen Ai Khoo
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hayley J Newton
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| | - Diana Stojanovski
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
31
|
Xu G, Zhong X, Shi Y, Liu Z, Jiang N, Liu J, Ding B, Li Z, Kang H, Ning Y, Liu W, Guo Z, Wang GL, Wang X. A fungal effector targets a heat shock-dynamin protein complex to modulate mitochondrial dynamics and reduce plant immunity. SCIENCE ADVANCES 2020; 6:6/48/eabb7719. [PMID: 33239288 PMCID: PMC7688324 DOI: 10.1126/sciadv.abb7719] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/13/2020] [Indexed: 05/31/2023]
Abstract
Mitochondria are essential for animal and plant immunity. Here, we report that the effector MoCDIP4 of the fungal pathogen Magnaporthe oryzae targets the mitochondria-associated OsDjA9-OsDRP1E protein complex to reduce rice immunity. The DnaJ protein OsDjA9 interacts with the dynamin-related protein OsDRP1E and promotes the degradation of OsDRP1E, which functions in mitochondrial fission. By contrast, MoCDIP4 binds OsDjA9 to compete with OsDRP1E, resulting in OsDRP1E accumulation. Knockout of OsDjA9 or overexpression of OsDRP1E or MoCDIP4 in transgenic rice results in shortened mitochondria and enhanced susceptibility to M. oryzae Overexpression of OsDjA9 or knockout of OsDRP1E in transgenic rice, in contrast, leads to elongated mitochondria and enhanced resistance to M. oryzae Our study therefore reveals a previously unidentified pathogen-infection strategy in which the pathogen delivers an effector into plant cells to target an HSP40-DRP complex; the targeting leads to the perturbation of mitochondrial dynamics, thereby inhibiting mitochondria-mediated plant immunity.
Collapse
Affiliation(s)
- Guojuan Xu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Plant Pathology, Department of Plant Pathology, China Agricultural University, Beijing 100193, China
| | - Xionghui Zhong
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanlong Shi
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhuo Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Jiang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jing Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Bo Ding
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhiqiang Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Houxiang Kang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yuese Ning
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Wende Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zejian Guo
- Key Laboratory of Plant Pathology, Department of Plant Pathology, China Agricultural University, Beijing 100193, China
| | - Guo-Liang Wang
- Department of Plant Pathology, The Ohio State University, Columbus, OH 43210, USA.
| | - Xuli Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
32
|
McKay DM, Mancini NL, Shearer J, Shutt T. Perturbed mitochondrial dynamics, an emerging aspect of epithelial-microbe interactions. Am J Physiol Gastrointest Liver Physiol 2020; 318:G748-G762. [PMID: 32116020 DOI: 10.1152/ajpgi.00031.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mitochondria exist in a complex network that is constantly remodeling via the processes of fission and fusion in response to intracellular conditions and extracellular stimuli. Excessive fragmentation of the mitochondrial network because of an imbalance between fission and fusion reduces the cells' capacity to generate ATP and can be a forerunner to cell death. Given the critical roles mitochondria play in cellular homeostasis and innate immunity, it is not surprising that many microbial pathogens can disrupt mitochondrial activity. Here we note the putative contribution of mitochondrial dysfunction to gut disease and review data showing that infection with microbial pathogens can alter the balance between mitochondrial fragmentation and fusion, preventing normal remodeling (i.e., dynamics) and can lead to cell death. Current data indicate that infection of epithelia or macrophages with microbial pathogens will ultimately result in excessive fragmentation of the mitochondrial network. Concerted research efforts are required to elucidate fully the processes that regulate mitochondrial dynamics, the mechanisms by which microbes affect epithelial mitochondrial fission and/or fusion, and the implications of this for susceptibility to infectious disease. We speculate that the commensal microbiome of the gut may be important for normal epithelial mitochondrial form and function. Drugs designed to counteract the effect of microbial pathogen interference with mitochondrial dynamics may be a new approach to infectious disease at mucosal surfaces.
Collapse
Affiliation(s)
- Derek M McKay
- Gastrointestinal Research Group (GIRG) and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole L Mancini
- Gastrointestinal Research Group (GIRG) and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jane Shearer
- Department of Biochemistry and Molecular Biology, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Timothy Shutt
- Department of Medical Genetics and Biochemistry & Molecular Biology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
33
|
Listeria monocytogenes Exploits Mitochondrial Contact Site and Cristae Organizing System Complex Subunit Mic10 To Promote Mitochondrial Fragmentation and Cellular Infection. mBio 2020; 11:mBio.03171-19. [PMID: 32019800 PMCID: PMC7002346 DOI: 10.1128/mbio.03171-19] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Pathogenic bacteria can target host cell organelles to take control of key cellular processes and promote their intracellular survival, growth, and persistence. Mitochondria are essential, highly dynamic organelles with pivotal roles in a wide variety of cell functions. Mitochondrial dynamics and function are intimately linked. Our previous research showed that Listeria monocytogenes infection impairs mitochondrial function and triggers fission of the mitochondrial network at an early infection stage, in a process that is independent of the presence of the main mitochondrial fission protein Drp1. Here, we analyzed how mitochondrial proteins change in response to L. monocytogenes infection and found that infection raises the levels of Mic10, a mitochondrial inner membrane protein involved in formation of cristae. We show that Mic10 is important for L. monocytogenes-dependent mitochondrial fission and infection of host cells. Our findings thus offer new insight into the mechanisms used by L. monocytogenes to hijack mitochondria to optimize host infection. Mitochondrial function adapts to cellular demands and is affected by the ability of the organelle to undergo fusion and fission in response to physiological and nonphysiological cues. We previously showed that infection with the human bacterial pathogen Listeria monocytogenes elicits transient mitochondrial fission and a drop in mitochondrion-dependent energy production through a mechanism requiring the bacterial pore-forming toxin listeriolysin O (LLO). Here, we performed quantitative mitochondrial proteomics to search for host factors involved in L. monocytogenes-induced mitochondrial fission. We found that Mic10, a critical component of the mitochondrial contact site and cristae organizing system (MICOS) complex, is significantly enriched in mitochondria isolated from cells infected with wild-type but not with LLO-deficient L. monocytogenes. Increased mitochondrial Mic10 levels did not correlate with upregulated transcription, suggesting a posttranscriptional mechanism. We then showed that Mic10 is necessary for L. monocytogenes-induced mitochondrial network fragmentation and that it contributes to L. monocytogenes cellular infection independently of MICOS proteins Mic13, Mic26, and Mic27. In conclusion, investigation of L. monocytogenes infection allowed us to uncover a role for Mic10 in mitochondrial fission.
Collapse
|
34
|
Ramond E, Jamet A, Coureuil M, Charbit A. Pivotal Role of Mitochondria in Macrophage Response to Bacterial Pathogens. Front Immunol 2019; 10:2461. [PMID: 31708919 PMCID: PMC6819784 DOI: 10.3389/fimmu.2019.02461] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/02/2019] [Indexed: 12/23/2022] Open
Abstract
Mitochondria are essential organelles that act as metabolic hubs and signaling platforms within the cell. Numerous mitochondrial functions, including energy metabolism, lipid synthesis, and autophagy regulation, are intimately linked to mitochondrial dynamics, which is shaped by ongoing fusion and fission events. Recently, several intracellular bacterial pathogens have been shown to modulate mitochondrial functions to maintain their replicative niche. Through selected examples of human bacterial pathogens, we will discuss how infection induces mitochondrial changes in infected macrophages, triggering modifications of the host metabolism that lead to important immunological reprogramming.
Collapse
Affiliation(s)
- Elodie Ramond
- Université de Paris, Paris, France.,INSERM U1151, Institut Necker-Enfants Malades, Team 7, Pathogenesis of Systemic Infections, Paris, France.,CNRS UMR 8253, Paris, France
| | - Anne Jamet
- Université de Paris, Paris, France.,INSERM U1151, Institut Necker-Enfants Malades, Team 7, Pathogenesis of Systemic Infections, Paris, France.,CNRS UMR 8253, Paris, France
| | - Mathieu Coureuil
- Université de Paris, Paris, France.,INSERM U1151, Institut Necker-Enfants Malades, Team 7, Pathogenesis of Systemic Infections, Paris, France.,CNRS UMR 8253, Paris, France
| | - Alain Charbit
- Université de Paris, Paris, France.,INSERM U1151, Institut Necker-Enfants Malades, Team 7, Pathogenesis of Systemic Infections, Paris, France.,CNRS UMR 8253, Paris, France
| |
Collapse
|