1
|
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
|
2
|
Brokatzky D, Gomes MC, Robertin S, Albino C, Miles SL, Mostowy S. Septins promote macrophage pyroptosis by regulating gasdermin D cleavage and ninjurin-1-mediated plasma membrane rupture. Cell Chem Biol 2024; 31:1518-1528.e6. [PMID: 39106869 DOI: 10.1016/j.chembiol.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] [Received: 12/13/2023] [Revised: 05/20/2024] [Accepted: 07/11/2024] [Indexed: 08/09/2024]
Abstract
The septin cytoskeleton is primarily known for roles in cell division and host defense against bacterial infection. Despite recent insights, the full breadth of roles for septins in host defense is poorly understood. In macrophages, Shigella induces pyroptosis, a pro-inflammatory form of cell death dependent upon gasdermin D (GSDMD) pores at the plasma membrane and cell surface protein ninjurin-1 (NINJ1) for membrane rupture. Here, we discover that septins promote macrophage pyroptosis induced by lipopolysaccharide (LPS)/nigericin and Shigella infection, but do not affect cytokine expression or release. We observe that septin filaments assemble at the plasma membrane, and cleavage of GSDMD is impaired in septin-depleted cells. We found that septins regulate mitochondrial dynamics and the expression of NINJ1. Using a Shigella-zebrafish infection model, we show that septin-mediated pyroptosis is an in vivo mechanism of infection control. The discovery of septins as a mediator of pyroptosis may inspire innovative anti-bacterial and anti-inflammatory treatments.
Collapse
Affiliation(s)
- Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK.
| | - Margarida C Gomes
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Stevens Robertin
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Carolina Albino
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Sydney L Miles
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
3
|
Lee YT, Senturk M, Guan Y, Wang MC. Bacteria-organelle communication in physiology and disease. J Cell Biol 2024; 223:e202310134. [PMID: 38748249 PMCID: PMC11096858 DOI: 10.1083/jcb.202310134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/03/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
Bacteria, omnipresent in our environment and coexisting within our body, exert dual beneficial and pathogenic influences. These microorganisms engage in intricate interactions with the human body, impacting both human health and disease. Simultaneously, certain organelles within our cells share an evolutionary relationship with bacteria, particularly mitochondria, best known for their energy production role and their dynamic interaction with each other and other organelles. In recent years, communication between bacteria and mitochondria has emerged as a new mechanism for regulating the host's physiology and pathology. In this review, we delve into the dynamic communications between bacteria and host mitochondria, shedding light on their collaborative regulation of host immune response, metabolism, aging, and longevity. Additionally, we discuss bacterial interactions with other organelles, including chloroplasts, lysosomes, and the endoplasmic reticulum (ER).
Collapse
Affiliation(s)
- Yi-Tang Lee
- Waisman Center, University of Wisconsin, Madison, WI, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Integrative Program of Molecular and Biochemical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Mumine Senturk
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Youchen Guan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Meng C. Wang
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| |
Collapse
|
4
|
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
|
5
|
Holley CL, Emming S, Monteleone MM, Mellacheruvu M, Kenney KM, Lawrence GMEP, Coombs JR, Burgener SS, Schroder K. The septin modifier, forchlorfenuron, activates NLRP3 via a potassium-independent mitochondrial axis. Cell Chem Biol 2024; 31:962-972.e4. [PMID: 38759620 DOI: 10.1016/j.chembiol.2024.04.012] [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: 11/11/2023] [Revised: 04/07/2024] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
The Nod-like receptor protein 3 (NLRP3) inflammasome is activated by stimuli that induce perturbations in cell homeostasis, which commonly converge on cellular potassium efflux. NLRP3 has thus emerged as a sensor for ionic flux. Here, we identify forchlorfenuron (FCF) as an inflammasome activator that triggers NLRP3 signaling independently of potassium efflux. FCF triggers the rearrangement of septins, key cytoskeletal proteins that regulate mitochondrial function. We report that FCF triggered the rearrangement of SEPT2 into tubular aggregates and stimulated SEPT2-independent NLRP3 inflammasome signaling. Similar to imiquimod, FCF induced the collapse of the mitochondrial membrane potential and mitochondrial respiration. FCF thereby joins the imidazoquinolines as a structurally distinct class of molecules that triggers NLRP3 inflammasome signaling independent of potassium efflux, likely by inducing mitochondrial damage.
Collapse
Affiliation(s)
- Caroline L Holley
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Stefan Emming
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mercedes M Monteleone
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Manasa Mellacheruvu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kirsten M Kenney
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Grace M E P Lawrence
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jared R Coombs
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sabrina S Burgener
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
6
|
Xian W, Fu J, Zhang Q, Li C, Zhao YB, Tang Z, Yuan Y, Wang Y, Zhou Y, Brzoic PS, Zheng N, Ouyang S, Luo ZQ, Liu X. The Shigella kinase effector OspG modulates host ubiquitin signaling to escape septin-cage entrapment. Nat Commun 2024; 15:3890. [PMID: 38719850 PMCID: PMC11078946 DOI: 10.1038/s41467-024-48205-4] [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: 09/28/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
Shigella flexneri is a Gram-negative bacterium causing severe bloody dysentery. Its pathogenesis is largely dictated by a plasmid-encoded type III secretion system (T3SS) and its associated effectors. Among these, the effector OspG has been shown to bind to the ubiquitin conjugation machinery (E2~Ub) to activate its kinase activity. However, the cellular targets of OspG remain elusive despite years of extensive efforts. Here we show by unbiased phosphoproteomics that a major target of OspG is CAND1, a regulatory protein controlling the assembly of cullin-RING ubiquitin ligases (CRLs). CAND1 phosphorylation weakens its interaction with cullins, which is expected to impact a large panel of CRL E3s. Indeed, global ubiquitome profiling reveals marked changes in the ubiquitination landscape when OspG is introduced. Notably, OspG promotes ubiquitination of a class of cytoskeletal proteins called septins, thereby inhibiting formation of cage-like structures encircling cytosolic bacteria. Overall, we demonstrate that pathogens have evolved an elaborate strategy to modulate host ubiquitin signaling to evade septin-cage entrapment.
Collapse
Affiliation(s)
- Wei Xian
- Department of Microbiology and Infectious Disease Center, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Jiaqi Fu
- Department of Respiratory Medicine, Center for Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, 130021, Changchun, China
| | - Qinxin Zhang
- Department of Microbiology and Infectious Disease Center, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Chuang Li
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Yan-Bo Zhao
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Zhiheng Tang
- Department of Microbiology and Infectious Disease Center, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Yi Yuan
- Department of Microbiology and Infectious Disease Center, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Ying Wang
- Department of Microbiology and Infectious Disease Center, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Yan Zhou
- Institute of Microbiology, College of Life Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Peter S Brzoic
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Ning Zheng
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| | - Songying Ouyang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Zhao-Qing Luo
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| | - Xiaoyun Liu
- Department of Microbiology and Infectious Disease Center, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
| |
Collapse
|
7
|
Robertin S, Brokatzky D, Lobato-Márquez D, Mostowy S. Regulation of integrin α5β1-mediated Staphylococcus aureus cellular invasion by the septin cytoskeleton. Eur J Cell Biol 2023; 102:151359. [PMID: 37683588 DOI: 10.1016/j.ejcb.2023.151359] [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: 04/05/2023] [Revised: 08/10/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Staphylococcus aureus, a Gram-positive bacterial pathogen, is an urgent health threat causing a wide range of clinical infections. Originally viewed as a strict extracellular pathogen, accumulating evidence has revealed S. aureus to be a facultative intracellular pathogen subverting host cell signalling to support invasion. The majority of clinical isolates produce fibronectin-binding proteins A and B (FnBPA and FnBPB) to interact with host integrin α5β1, a key component of focal adhesions. S. aureus binding of integrin α5β1 promotes its clustering on the host cell surface, triggering activation of focal adhesion kinase (FAK) and cytoskeleton rearrangements to promote bacterial invasion into non-phagocytic cells. Here, we discover that septins, a component of the cytoskeleton that assembles on membranes, are recruited as collar-like structures with actin to S. aureus invasion sites engaging integrin α5β1. To investigate septin recruitment to the plasma membrane in a bacteria-free system, we used FnBPA-coated latex beads and showed that septins are recruited upon activation of integrin α5β1. SEPT2 depletion reduced S. aureus invasion, but increased surface expression of integrin α5 and adhesion of S. aureus to host cells. Consistent with this, SEPT2 depletion increased cellular protein levels of integrin α5 and β1 subunits, as well as FAK. Collectively, these results provide insights into regulation of integrin α5β1 and invasion of S. aureus by the septin cytoskeleton.
Collapse
Affiliation(s)
- Stevens Robertin
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Damián Lobato-Márquez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|
8
|
Mageswaran SK, Grotjahn DA, Zeng X, Barad BA, Medina M, Hoang MH, Dobro MJ, Chang YW, Xu M, Yang WY, Jensen GJ. Nanoscale details of mitochondrial constriction revealed by cryoelectron tomography. Biophys J 2023; 122:3768-3782. [PMID: 37533259 PMCID: PMC10541493 DOI: 10.1016/j.bpj.2023.07.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023] Open
Abstract
Mitochondria adapt to changing cellular environments, stress stimuli, and metabolic demands through dramatic morphological remodeling of their shape, and thus function. Such mitochondrial dynamics is often dependent on cytoskeletal filament interactions. However, the precise organization of these filamentous assemblies remains speculative. Here, we apply cryogenic electron tomography to directly image the nanoscale architecture of the cytoskeletal-membrane interactions involved in mitochondrial dynamics in response to damage. We induced mitochondrial damage via membrane depolarization, a cellular stress associated with mitochondrial fragmentation and mitophagy. We find that, in response to acute membrane depolarization, mammalian mitochondria predominantly organize into tubular morphology that abundantly displays constrictions. We observe long bundles of both unbranched actin and septin filaments enriched at these constrictions. We also observed septin-microtubule interactions at these sites and elsewhere, suggesting that these two filaments guide each other in the cytosolic space. Together, our results provide empirical parameters for the architecture of mitochondrial constriction factors to validate/refine existing models and inform the development of new ones.
Collapse
Affiliation(s)
- Shrawan Kumar Mageswaran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California; Department of Biophysics and Biochemistry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Danielle Ann Grotjahn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California.
| | - Xiangrui Zeng
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Benjamin Asher Barad
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California
| | - Michaela Medina
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California
| | - My Hanh Hoang
- Department of Biophysics and Biochemistry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Yi-Wei Chang
- Department of Biophysics and Biochemistry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Min Xu
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Wei Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Grant J Jensen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California; Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah.
| |
Collapse
|
9
|
Maurice NM, Sadikot RT. Mitochondrial Dysfunction in Bacterial Infections. Pathogens 2023; 12:1005. [PMID: 37623965 PMCID: PMC10458073 DOI: 10.3390/pathogens12081005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Mitochondria are critical in numerous cellular processes, including energy generation. Bacterial pathogens target host cell mitochondria through various mechanisms to disturb the host response and improve bacterial survival. We review recent advances in the understanding of how bacteria cause mitochondrial dysfunction through perturbations in mitochondrial cell-death pathways, energy production, mitochondrial dynamics, mitochondrial quality control, DNA repair, and the mitochondrial unfolded protein response. We also briefly highlight possible therapeutic approaches aimed at restoring the host mitochondrial function as a novel strategy to enhance the host response to bacterial infection.
Collapse
Affiliation(s)
- Nicholas M. Maurice
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Ruxana T. Sadikot
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
10
|
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
|
11
|
Sharma K, Menon MB. Decoding post-translational modifications of mammalian septins. Cytoskeleton (Hoboken) 2023; 80:169-181. [PMID: 36797225 DOI: 10.1002/cm.21747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
Septins are cytoskeletal GTPases that form nonpolar filaments and higher-ordered structures and they take part in a wide range of cellular processes. Septins are conserved from yeast to mammals but absent from higher plants. The number of septin genes vary between organisms and they usually form complex heteropolymeric networks. Most septins are known to be capable of GTP hydrolysis which may regulate septin dynamics. Knowledge on regulation of septin function by post-translational modifications is still in its infancy. In this review article, we highlight the post-translational modifications reported for the 13 human septins and discuss their implications on septin functions. In addition to the functionally investigated modifications, we also try to make sense of the complex septin post-translational modification code revealed from large-scale phospho-proteomic datasets. Future studies may determine how these isoform-specific and homology group specific modifications affect septin structure and function.
Collapse
Affiliation(s)
- Khushboo Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Manoj B Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
12
|
Perry JA, Werner ME, Rivenbark L, Maddox AS. Caenorhabditis elegans septins contribute to the development and structure of the oogenic germline. Cytoskeleton (Hoboken) 2023; 80:215-227. [PMID: 37265173 PMCID: PMC10524836 DOI: 10.1002/cm.21763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023]
Abstract
Oocytes must be exceptionally large cells in order to support embryonic development. Throughout animal phylogeny, a specialized cell called a syncytium, wherein many nuclei share a continuous cytoplasm, achieves oogenesis. The syncytial nature of germline architecture is key to its function and depends on conserved components of the cortical cytoskeleton. Septins form non-polar cytoskeletal polymers that associate with membranes. In the syncytial germline of the nematode Caenorhabditis elegans, septins are highly enriched on the cortex and generally required for fertility, but the role of septins in the germline is poorly understood. We report that the C. elegans septins, UNC-59 and UNC-61, are important for germline extension during development, the maintenance of its syncytial architecture, and production of oocytes. While much of our findings substantiate the idea that the two C. elegans septins act together, we also found evidence that they have distinct functions. Loss of UNC-61 perturbed germline extension during germline development, while the loss of UNC-59 function severely affected germline architecture in adult hermaphrodites. Consultation of clustering results from a large-scale high-throughput screen suggested that septins are involved in germ cell proliferation and/or differentiation. In sum, our findings implicate a conserved cytoskeletal component in the complex architecture of a germline syncytium.
Collapse
Affiliation(s)
- Jenna A Perry
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael E Werner
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Larry Rivenbark
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Amy Shaub Maddox
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Van Ngo H, Robertin S, Brokatzky D, Bielecka MK, Lobato‐Márquez D, Torraca V, Mostowy S. Septins promote caspase activity and coordinate mitochondrial apoptosis. Cytoskeleton (Hoboken) 2023; 80:254-265. [PMID: 35460543 PMCID: PMC10952901 DOI: 10.1002/cm.21696] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/08/2022] [Accepted: 04/05/2022] [Indexed: 11/06/2022]
Abstract
Apoptosis is a form of regulated cell death essential for tissue homeostasis and embryonic development. Apoptosis also plays a key role during bacterial infection, yet some intracellular bacterial pathogens (such as Shigella flexneri, whose lipopolysaccharide can block apoptosis) can manipulate cell death programs as an important survival strategy. Septins are a component of the cytoskeleton essential for mitochondrial dynamics and host defense, however, the role of septins in regulated cell death is mostly unknown. Here, we discover that septins promote mitochondrial (i.e., intrinsic) apoptosis in response to treatment with staurosporine (a pan-kinase inhibitor) or etoposide (a DNA topoisomerase inhibitor). Consistent with a role for septins in mitochondrial dynamics, septins promote the release of mitochondrial protein cytochrome c in apoptotic cells and are required for the proteolytic activation of caspase-3, caspase-7, and caspase-9 (core components of the apoptotic machinery). Apoptosis of HeLa cells induced in response to infection by S. flexneri ΔgalU (a lipopolysaccharide mutant unable to block apoptosis) is also septin-dependent. In vivo, zebrafish larvae are significantly more susceptible to infection with S. flexneri ΔgalU (as compared to infection with wildtype S. flexneri), yet septin deficient larvae are equally susceptible to infection with S. flexneri ΔgalU and wildtype S. flexneri. These data provide a new molecular framework to understand the complexity of mitochondrial apoptosis and its ability to combat bacterial infection.
Collapse
Affiliation(s)
- Hoan Van Ngo
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
| | - Stevens Robertin
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
| | - Dominik Brokatzky
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
| | - Magdalena K. Bielecka
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
| | - Damián Lobato‐Márquez
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
| | - Vincenzo Torraca
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
- School of Life SciencesUniversity of WestminsterLondonUK
| | - Serge Mostowy
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
14
|
Lobato-Márquez D, Conesa JJ, López-Jiménez AT, Divine ME, Pruneda JN, Mostowy S. Septins and K63 ubiquitin chains are present in separate bacterial microdomains during autophagy of entrapped Shigella. J Cell Sci 2023; 136:jcs261139. [PMID: 36939083 PMCID: PMC10264824 DOI: 10.1242/jcs.261139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/21/2023] Open
Abstract
During host cell invasion, Shigella escapes to the cytosol and polymerizes actin for cell-to-cell spread. To restrict cell-to-cell spread, host cells employ cell-autonomous immune responses including antibacterial autophagy and septin cage entrapment. How septins interact with the autophagy process to target Shigella for destruction is poorly understood. Here, we employed a correlative light and cryo-soft X-ray tomography (cryo-SXT) pipeline to study Shigella septin cage entrapment in its near-native state. Quantitative cryo-SXT showed that Shigella fragments mitochondria and enabled visualization of X-ray-dense structures (∼30 nm resolution) surrounding Shigella entrapped in septin cages. Using Airyscan confocal microscopy, we observed lysine 63 (K63)-linked ubiquitin chains decorating septin-cage-entrapped Shigella. Remarkably, septins and K63 chains are present in separate bacterial microdomains, indicating they are recruited separately during antibacterial autophagy. Cryo-SXT and live-cell imaging revealed an interaction between septins and LC3B-positive membranes during autophagy of Shigella. Together, these findings demonstrate how septin-caged Shigella are targeted for autophagy and provide fundamental insights into autophagy-cytoskeleton interactions.
Collapse
Affiliation(s)
- Damián Lobato-Márquez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - José Javier Conesa
- MISTRAL beamline, ALBA Synchrotron Light Source, Cerdanyola del Vallès, 08290 Barcelona, Spain
| | - Ana Teresa López-Jiménez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Michael E. Divine
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jonathan N. Pruneda
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
15
|
García-Rodríguez FJ, Buchrieser C, Escoll P. Legionella and mitochondria, an intriguing relationship. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 374:37-81. [PMID: 36858656 DOI: 10.1016/bs.ircmb.2022.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Legionella pneumophila is the causative agent of Legionnaires' disease, a severe pneumonia. L. pneumophila injects via a type-IV-secretion-system (T4SS) more than 300 bacterial proteins into macrophages, its main host cell in humans. Certain of these bacterial effectors target organelles in the infected cell and hijack multiple processes to facilitate all steps of the intracellular life cycle of this pathogen. In this review, we discuss the interplay between L. pneumophila, an intracellular bacterium fully armed with virulence tools, and mitochondria, the extraordinary eukaryotic organelles playing prominent roles in cellular bioenergetics, cell-autonomous immunity and cell death. We present and discuss key findings concerning the multiple interactions of L. pneumophila with mitochondria during infection and the mechanisms employed by T4SS effectors that target mitochondrial functions to subvert infected cells.
Collapse
Affiliation(s)
| | - Carmen Buchrieser
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires and CNRS UMR 6047, Paris, France.
| | - Pedro Escoll
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires and CNRS UMR 6047, Paris, France.
| |
Collapse
|
16
|
Role of mitochondria in regulating immune response during bacterial infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 374:159-200. [PMID: 36858655 DOI: 10.1016/bs.ircmb.2022.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mitochondria are dynamic organelles of eukaryotes involved in energy production and fatty acid oxidation. Besides maintaining ATP production, calcium signaling, cellular apoptosis, and fatty acid synthesis, mitochondria are also known as the central hub of the immune system as it regulates the innate immune pathway during infection. Mitochondria mediated immune functions mainly involve regulation of reactive oxygen species production, inflammasome activation, cytokine secretion, and apoptosis of infected cells. Recent findings indicate that cellular mitochondria undergo constant biogenesis, fission, fusion and degradation, and these dynamics regulate cellular immuno-metabolism. Several intracellular pathogens target and modulate these normal functions of mitochondria to facilitate their own survival and growth. De-regulation of mitochondrial functions and dynamics favors bacterial infection and pathogens are able to protect themselves from mitochondria mediated immune responses. Here, we will discuss how mitochondria mediated anti-bacterial immune pathways help the host to evade pathogenic insult. In addition, examples of bacterial pathogens modulating mitochondrial metabolism and dynamics will also be elaborated. Study of these interactions between the mitochondria and bacterial pathogens during infection will lead to a better understanding of the mitochondrial metabolism pathways and dynamics important for the establishment of bacterial diseases. In conclusion, detailed studies on how mitochondria regulate the immune response during bacterial infection can open up new avenues to develop mitochondria centric anti-bacterial therapeutics.
Collapse
|
17
|
Weems AD, Welf ES, Driscoll MK, Zhou FY, Mazloom-Farsibaf H, Chang BJ, Murali VS, Gihana GM, Weiss BG, Chi J, Rajendran D, Dean KM, Fiolka R, Danuser G. Blebs promote cell survival by assembling oncogenic signalling hubs. Nature 2023; 615:517-525. [PMID: 36859545 PMCID: PMC10881276 DOI: 10.1038/s41586-023-05758-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/25/2023] [Indexed: 03/03/2023]
Abstract
Most human cells require anchorage for survival. Cell-substrate adhesion activates diverse signalling pathways, without which cells undergo anoikis-a form of programmed cell death1. Acquisition of anoikis resistance is a pivotal step in cancer disease progression, as metastasizing cells often lose firm attachment to surrounding tissue2,3. In these poorly attached states, cells adopt rounded morphologies and form small hemispherical plasma membrane protrusions called blebs4-11. Bleb function has been thoroughly investigated in the context of amoeboid migration, but it has been examined far less in other scenarios12. Here we show by three-dimensional imaging and manipulation of cell morphological states that blebbing triggers the formation of plasma membrane-proximal signalling hubs that confer anoikis resistance. Specifically, in melanoma cells, blebbing generates plasma membrane contours that recruit curvature-sensing septin proteins as scaffolds for constitutively active mutant NRAS and effectors. These signalling hubs activate ERK and PI3K-well-established promoters of pro-survival pathways. Inhibition of blebs or septins has little effect on the survival of well-adhered cells, but in detached cells it causes NRAS mislocalization, reduced MAPK and PI3K activity, and ultimately, death. This unveils a morphological requirement for mutant NRAS to operate as an effective oncoprotein. Furthermore, whereas some BRAF-mutated melanoma cells do not rely on this survival pathway in a basal state, inhibition of BRAF and MEK strongly sensitizes them to both bleb and septin inhibition. Moreover, fibroblasts engineered to sustain blebbing acquire the same anoikis resistance as cancer cells even without harbouring oncogenic mutations. Thus, blebs are potent signalling organelles capable of integrating myriad cellular information flows into concerted cellular responses, in this case granting robust anoikis resistance.
Collapse
Affiliation(s)
- Andrew D Weems
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Erik S Welf
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Meghan K Driscoll
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Felix Y Zhou
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Bo-Jui Chang
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Vasanth S Murali
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Gabriel M Gihana
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Byron G Weiss
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joseph Chi
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Divya Rajendran
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kevin M Dean
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Reto Fiolka
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
18
|
Benoit B, Poüs C, Baillet A. Septins as membrane influencers: direct play or in association with other cytoskeleton partners. Front Cell Dev Biol 2023; 11:1112319. [PMID: 36875762 PMCID: PMC9982393 DOI: 10.3389/fcell.2023.1112319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
The cytoskeleton comprises three polymerizing structures that have been studied for a long time, actin microfilaments, microtubules and intermediate filaments, plus more recently investigated dynamic assemblies like septins or the endocytic-sorting complex required for transport (ESCRT) complex. These filament-forming proteins control several cell functions through crosstalks with each other and with membranes. In this review, we report recent works that address how septins bind to membranes, and influence their shaping, organization, properties and functions, either by binding to them directly or indirectly through other cytoskeleton elements.
Collapse
Affiliation(s)
- Béatrice Benoit
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| | - Christian Poüs
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France.,Laboratoire de Biochimie-Hormonologie, Hôpital Antoine Béclère, AP-HP, Hôpitaux Universitaires Paris-Saclay, Clamart, France
| | - Anita Baillet
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| |
Collapse
|
19
|
Septin barriers protect mammalian host cells against Pseudomonas aeruginosa invasion. Cell Rep 2022; 41:111510. [DOI: 10.1016/j.celrep.2022.111510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/29/2022] [Accepted: 09/23/2022] [Indexed: 11/24/2022] Open
|
20
|
Neuronal-specific septin-3 binds Atg8/LC3B, accumulates and localizes to autophagosomes during induced autophagy. Cell Mol Life Sci 2022; 79:471. [PMID: 35932293 PMCID: PMC9356936 DOI: 10.1007/s00018-022-04488-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/27/2022] [Accepted: 07/13/2022] [Indexed: 11/03/2022]
Abstract
In synapses that show signs of local apoptosis and mitochondrial stress and undergo neuro-immunological synapse pruning, an increase in the levels of the presynaptic protein, neuronal-specific septin-3 can be observed. Septin-3 is a member of the septin GTPase family with the ability to form multimers and contribute to the cytoskeleton. However, the function of septin-3 remains elusive. Here, we provide evidence that septin-3 is capable of binding the most-studied autophagy protein Atg8 homolog microtubule-associated protein 1 light chain 3B (LC3B), besides another homolog, GABA receptor-associated protein-like 2 (GABARAPL2). Moreover, we demonstrate that colocalization of septin-3 and LC3B increases upon chemical autophagy induction in primary neuronal cells. Septin-3 is accumulated in primary neurons upon autophagy enhancement or blockade, similar to autophagy proteins. Using electron microscopy, we also show that septin-3 localizes to LC3B positive membranes and can be found at mitochondria. However, colocalization results of septin-3 and the early mitophagy marker PTEN-induced kinase 1 (PINK1) do not support that binding of septin-3 to mitochondria is mitophagy related. We conclude that septin-3 correlates with synaptic/neuronal autophagy, binds Atg8 and localizes to autophagic membranes that can be enhanced with chemical autophagy induction. Based on our results, elevated septin-3 levels might indicate enhanced or impeded autophagy in neurons.
Collapse
|
21
|
Gönczi M, Ráduly Z, Szabó L, Fodor J, Telek A, Dobrosi N, Balogh N, Szentesi P, Kis G, Antal M, Trencsenyi G, Dienes B, Csernoch L. Septin7 is indispensable for proper skeletal muscle architecture and function. eLife 2022; 11:e75863. [PMID: 35929607 PMCID: PMC9355566 DOI: 10.7554/elife.75863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/03/2022] [Indexed: 11/13/2022] Open
Abstract
Today septins are considered as the fourth component of the cytoskeleton, with the Septin7 isoform playing a critical role in the formation of higher-order structures. While its importance has already been confirmed in several intracellular processes of different organs, very little is known about its role in skeletal muscle. Here, using Septin7 conditional knockdown (KD) mouse model, the C2C12 cell line, and enzymatically isolated adult muscle fibers, the organization and localization of septin filaments are revealed, and an ontogenesis-dependent expression of Septin7 is demonstrated. KD mice displayed a characteristic hunchback phenotype with skeletal deformities, reduction in in vivo and in vitro force generation, and disorganized mitochondrial networks. Furthermore, knockout of Septin7 in C2C12 cells resulted in complete loss of cell division while KD cells provided evidence that Septin7 is essential for proper myotube differentiation. These and the transient increase in Septin7 expression following muscle injury suggest that it may be involved in muscle regeneration and development.
Collapse
Affiliation(s)
- Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Zsolt Ráduly
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - László Szabó
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Andrea Telek
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Nóra Dobrosi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Norbert Balogh
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Gréta Kis
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - György Trencsenyi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| |
Collapse
|
22
|
Abstract
Pyroptosis, a regulated form of pro-inflammatory cell death, is characterised by cell lysis and by the release of cytokines, damage- and pathogen-associated molecular patterns. It plays an important role during bacterial infection, where it can promote an inflammatory response and eliminate the replicative niche of intracellular pathogens. Recent work, using a variety of bacterial pathogens, has illuminated the versatility of pyroptosis, revealing unexpected and important concepts underlying host defence. In this Review, we overview the molecular mechanisms underlying pyroptosis and discuss their role in host defence, from the single cell to the whole organism. We focus on recent studies using three cellular microbiology paradigms - Mycobacterium tuberculosis, Salmonella Typhimurium and Shigella flexneri - that have transformed the field of pyroptosis. We compare insights discovered in tissue culture, zebrafish and mouse models, highlighting the advantages and disadvantages of using these complementary infection models to investigate pyroptosis and for modelling human infection. Moving forward, we propose that in-depth knowledge of pyroptosis obtained from complementary infection models can better inform future studies using higher vertebrates, including humans, and help develop innovative host-directed therapies to combat bacterial infection.
Collapse
Affiliation(s)
- Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
23
|
Hill NS, Welch MD. A glycine-rich PE_PGRS protein governs mycobacterial actin-based motility. Nat Commun 2022; 13:3608. [PMID: 35750685 PMCID: PMC9232537 DOI: 10.1038/s41467-022-31333-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
Many key insights into actin regulation have been derived through examining how microbial pathogens intercept the actin cytoskeleton during infection. Mycobacterium marinum, a close relative of the human pathogen Mycobacterium tuberculosis, polymerizes host actin at the bacterial surface to drive intracellular movement and cell-to-cell spread during infection. However, the mycobacterial factor that commandeers actin polymerization has remained elusive. Here, we report the identification and characterization of the M. marinum actin-based motility factor designated mycobacterial intracellular rockets A (MirA), which is a member of the glycine-rich PE_PGRS protein family. MirA contains an amphipathic helix to anchor into the mycobacterial outer membrane and, surprisingly, also the surface of host lipid droplet organelles. MirA directly binds to and activates the host protein N-WASP to stimulate actin polymerization through the Arp2/3 complex, directing both bacterial and lipid droplet actin-based motility. MirA is dissimilar to known N-WASP activating ligands and may represent a new class of microbial and host actin regulator. Additionally, the MirA-N-WASP interaction represents a model to understand how the enigmatic PE_PGRS proteins contribute to mycobacterial pathogenesis.
Collapse
Affiliation(s)
- Norbert S Hill
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
24
|
Gutierrez MG, Enninga J. Intracellular niche switching as host subversion strategy of bacterial pathogens. Curr Opin Cell Biol 2022; 76:102081. [PMID: 35487154 DOI: 10.1016/j.ceb.2022.102081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 11/03/2022]
Abstract
Numerous bacterial pathogens "confine" themselves within host cells with an intracellular localization as main or exclusive niche. Many of them switch dynamically between a membrane-bound or cytosolic lifestyle. This requires either membrane damage and/or repair of the bacterial-containing compartment. Niche switching has profound consequences on how the host cell recognizes the pathogens in time and space for elimination. Moreover, niche switching impacts how bacteria communicate with host cells to obtain nutrients, and it affects the accessibility to antibiotics. Understanding the local environments and cellular phenotypes that lead to niche switching is critical for developing new host-targeted antimicrobial strategies, and has the potential to shed light into fundamental cellular processes.
Collapse
Affiliation(s)
- Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
| | - Jost Enninga
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France; Université de Paris, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
25
|
Petit TJ, Lebreton A. Adaptations of intracellular bacteria to vacuolar or cytosolic niches. Trends Microbiol 2022; 30:736-748. [DOI: 10.1016/j.tim.2022.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/28/2022]
|
26
|
Baillet A, McMurray MA, Oakes PW. Meeting report - the ever-fascinating world of septins. J Cell Sci 2021; 134:jcs259552. [PMID: 34910818 PMCID: PMC10658896 DOI: 10.1242/jcs.259552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Septins are GTP-binding proteins that assemble into hetero-oligomers. They can interact with each other end-to-end to form filaments, making them the fourth element of the cytoskeleton. To update the current knowledge on the ever-increasing implications of these fascinating proteins in cellular functions, a hundred expert scientists from across the globe gathered from 12 to 15 October 2021 in Berlin for the first hybrid-format (on site and virtual) EMBO workshop Molecular and Cell Biology of Septins.
Collapse
Affiliation(s)
- Anita Baillet
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Michael A. McMurray
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Patrick W. Oakes
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153USA
| |
Collapse
|
27
|
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: 170] [Impact Index Per Article: 42.5] [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
|
28
|
Devlin L, Okletey J, Perkins G, Bowen JR, Nakos K, Montagna C, Spiliotis ET. Proteomic profiling of the oncogenic septin 9 reveals isoform-specific interactions in breast cancer cells. Proteomics 2021; 21:e2100155. [PMID: 34409731 DOI: 10.1002/pmic.202100155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023]
Abstract
Septins are a family of multimeric GTP-binding proteins, which are abnormally expressed in cancer. Septin 9 (SEPT9) is an essential and ubiquitously expressed septin with multiple isoforms, which have differential expression patterns and effects in breast cancer cells. It is unknown, however, if SEPT9 isoforms associate with different molecular networks and functions. Here, we performed a proteomic screen in MCF-7 breast cancer cells to identify the interactome of GFP-SEPT9 isoforms 1, 4 and 5, which vary significantly in their N-terminal extensions. While all three isoforms associated with SEPT2 and SEPT7, the truncated SEPT9_i4 and SEPT9_i5 interacted with septins of the SEPT6 group more promiscuously than SEPT9_i1, which bound predominately SEPT8. Spatial mapping and functional clustering of non-septin partners showed isoform-specific differences in interactions with proteins of distinct subcellular organelles (e.g., nuclei, centrosomes, cilia) and functions such as cell signalling and ubiquitination. The interactome of the full length SEPT9_i1 was more enriched in cytoskeletal regulators, while the truncated SEPT9_i4 and SEPT9_i5 exhibited preferential and isoform-specific interactions with nuclear, signalling, and ubiquitinating proteins. These data provide evidence for isoform-specific interactions, which arise from truncations in the N-terminal extensions of SEPT9, and point to novel roles in the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Louis Devlin
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA.,Sanofi Pasteur, Swiftwater, Pennsylvania, USA
| | - Joshua Okletey
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | | | - Jonathan R Bowen
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Konstantinos Nakos
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Cristina Montagna
- Department of Radiology & Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Liu K, Kong L, Graham DB, Carey KL, Xavier RJ. SAC1 regulates autophagosomal phosphatidylinositol-4-phosphate for xenophagy-directed bacterial clearance. Cell Rep 2021; 36:109434. [PMID: 34320354 PMCID: PMC8327279 DOI: 10.1016/j.celrep.2021.109434] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/21/2020] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Phosphoinositides are important molecules in lipid signaling, membrane identity, and trafficking that are spatiotemporally controlled by factors from both mammalian cells and intracellular pathogens. Here, using small interfering RNA (siRNA) directed against phosphoinositide kinases and phosphatases, we screen for regulators of the host innate defense response to intracellular bacterial replication. We identify SAC1, a transmembrane phosphoinositide phosphatase, as an essential regulator of xenophagy. Depletion or inactivation of SAC1 compromises fusion between Salmonella-containing autophagosomes and lysosomes, leading to increased bacterial replication. Mechanistically, the loss of SAC1 results in aberrant accumulation of phosphatidylinositol-4-phosphate [PI(4)P] on Salmonella-containing autophagosomes, thus facilitating recruitment of SteA, a PI(4)P-binding Salmonella effector protein, which impedes lysosomal fusion. Replication of Salmonella lacking SteA is suppressed by SAC-1-deficient cells, however, demonstrating bacterial adaptation to xenophagy. Our findings uncover a paradigm in which a host protein regulates the level of its substrate and impairs the function of a bacterial effector during xenophagy.
Collapse
Affiliation(s)
- Kai Liu
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingjia Kong
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Daniel B Graham
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Ramnik J Xavier
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
30
|
Lobato-Márquez D, Xu J, Güler GÖ, Ojiakor A, Pilhofer M, Mostowy S. Mechanistic insight into bacterial entrapment by septin cage reconstitution. Nat Commun 2021; 12:4511. [PMID: 34301939 PMCID: PMC8302635 DOI: 10.1038/s41467-021-24721-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/01/2021] [Indexed: 11/22/2022] Open
Abstract
Septins are cytoskeletal proteins that assemble into hetero-oligomeric complexes and sense micron-scale membrane curvature. During infection with Shigella flexneri, an invasive enteropathogen, septins restrict actin tail formation by entrapping bacteria in cage-like structures. Here, we reconstitute septin cages in vitro using purified recombinant septin complexes (SEPT2-SEPT6-SEPT7), and study how these recognize bacterial cells and assemble on their surface. We show that septin complexes recognize the pole of growing Shigella cells. An amphipathic helix domain in human SEPT6 enables septins to sense positively curved membranes and entrap bacterial cells. Shigella strains lacking lipopolysaccharide components are more efficiently entrapped in septin cages. Finally, cryo-electron tomography of in vitro cages reveals how septins assemble as filaments on the bacterial cell surface. Septins are cytoskeletal proteins that assemble into complexes and contribute to immunity by entrapping intracellular bacteria in cage-like structures. Here, Lobato-Márquez et al. reconstitute septin cages in vitro using purified recombinant complexes, and study how these recognize bacterial cells and assemble as filaments on their surface.
Collapse
Affiliation(s)
- Damián Lobato-Márquez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Jingwei Xu
- Department of Biology, Institute of Molecular Biology and Biophysics, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Gizem Özbaykal Güler
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Adaobi Ojiakor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Martin Pilhofer
- Department of Biology, Institute of Molecular Biology and Biophysics, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
31
|
Spiliotis ET, Kesisova IA. Spatial regulation of microtubule-dependent transport by septin GTPases. Trends Cell Biol 2021; 31:979-993. [PMID: 34253430 DOI: 10.1016/j.tcb.2021.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 01/21/2023]
Abstract
The intracellular long-range transport of membrane vesicles and organelles is mediated by microtubule motors (kinesins, dynein) which move cargo with spatiotemporal accuracy and efficiency. How motors navigate the microtubule network and coordinate their activity on membrane cargo are fundamental but poorly understood questions. New studies show that microtubule-dependent membrane traffic is spatially controlled by septins - a unique family of multimerizing GTPases that associate with microtubules and membrane organelles. We review how septins selectively regulate motor interactions with microtubules and membrane cargo. We posit that septins provide a novel traffic code that specifies the movement and directionality of select motor-cargo complexes on distinct microtubule tracks.
Collapse
Affiliation(s)
- Elias T Spiliotis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA.
| | - Ilona A Kesisova
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
32
|
Spiliotis ET, McMurray MA. Masters of asymmetry - lessons and perspectives from 50 years of septins. Mol Biol Cell 2021; 31:2289-2297. [PMID: 32991244 PMCID: PMC7851956 DOI: 10.1091/mbc.e19-11-0648] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Septins are a unique family of GTPases, which were discovered 50 years ago as essential genes for the asymmetric cell shape and division of budding yeast. Septins assemble into filamentous nonpolar polymers, which associate with distinct membrane macrodomains and subpopulations of actin filaments and microtubules. While structurally a cytoskeleton-like element, septins function predominantly as spatial regulators of protein localization and interactions. Septin scaffolds and barriers have provided a long-standing paradigm for the generation and maintenance of asymmetry in cell membranes. Septins also promote asymmetry by regulating the spatial organization of the actin and microtubule cytoskeleton, and biasing the directionality of membrane traffic. In this 50th anniversary perspective, we highlight how septins have conserved and adapted their roles as effectors of membrane and cytoplasmic asymmetry across fungi and animals. We conclude by outlining principles of septin function as a module of symmetry breaking, which alongside the monomeric small GTPases provides a core mechanism for the biogenesis of molecular asymmetry and cell polarity.
Collapse
Affiliation(s)
| | - Michael A McMurray
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
33
|
Kühn S, Bergqvist J, Gil M, Valenzuela C, Barrio L, Lebreton S, Zurzolo C, Enninga J. Actin Assembly around the Shigella-Containing Vacuole Promotes Successful Infection. Cell Rep 2021; 31:107638. [PMID: 32402280 PMCID: PMC7225751 DOI: 10.1016/j.celrep.2020.107638] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/10/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
The enteroinvasive bacterium Shigella flexneri forces its uptake into non-phagocytic host cells through the translocation of T3SS effectors that subvert the actin cytoskeleton. Here, we report de novo actin polymerization after cellular entry around the bacterium-containing vacuole (BCV) leading to the formation of a dynamic actin cocoon. This cocoon is thicker than any described cellular actin structure and functions as a gatekeeper for the cytosolic access of the pathogen. Host CDC42, TOCA-1, N-WASP, WIP, the Arp2/3 complex, cortactin, coronin, and cofilin are recruited to the actin cocoon. They are subverted by T3SS effectors, such as IpgD, IpgB1, and IcsB. IcsB immobilizes components of the actin polymerization machinery at the BCV dependent on its fatty acyltransferase activity. This represents a unique microbial subversion strategy through localized entrapment of host actin regulators causing massive actin assembly. We propose that the cocoon promotes subsequent invasion steps for successful Shigella infection.
A thick actin cocoon forms de novo around the Shigella-containing vacuole upon entry The effector IcsB entraps host actin regulators at the vacuole by lipidation Cdc42, N-WASP, and the Arp2/3 complex are major actin cocoon regulators Cocoon formation promotes subsequent Shigella niche formation and dissemination
Collapse
Affiliation(s)
- Sonja Kühn
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - John Bergqvist
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Magdalena Gil
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Camila Valenzuela
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Laura Barrio
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Stéphanie Lebreton
- Institut Pasteur, Department of Cell Biology and Infection, Membrane Trafficking and Pathogenesis Unit, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Chiara Zurzolo
- Institut Pasteur, Department of Cell Biology and Infection, Membrane Trafficking and Pathogenesis Unit, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Jost Enninga
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
34
|
Kutsch M, González-Prieto C, Lesser CF, Coers J. The GBP1 microcapsule interferes with IcsA-dependent septin cage assembly around Shigella flexneri. Pathog Dis 2021; 79:6246431. [PMID: 33885766 DOI: 10.1093/femspd/ftab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/08/2021] [Indexed: 12/20/2022] Open
Abstract
Many cytosolic bacterial pathogens hijack the host actin polymerization machinery to form actin tails that promote direct cell-to-cell spread, enabling these pathogens to avoid extracellular immune defenses. However, these pathogens are still susceptible to intracellular cell-autonomous immune responses that restrict bacterial actin-based motility. Two classes of cytosolic antimotility factors, septins and guanylate-binding proteins (GBPs), have recently been established to block actin tail formation by the human-adapted bacterial pathogen Shigella flexneri. Both septin cages and GBP1 microcapsules restrict S. flexneri cell-to-cell spread by blocking S. flexneri actin-based motility. While septins assemble into cage-like structures around immobile S. flexneri, GBP1 forms microcapsules around both motile and immobile bacteria. The interplay between these two defense programs remains elusive. Here, we demonstrate that GBP1 microcapsules block septin cage assembly, likely by interfering with the function of S. flexneri IcsA, the outer membrane protein that promotes actin-based motility, as this protein is required for septin cage formation. However, S. flexneri that escape from GBP1 microcapsules via the activity of IpaH9.8, a type III secreted effector that promotes the degradation of GBPs, are often captured within septin cages. Thus, our studies reveal how septin cages and GBP1 microcapsules represent complementary host cell antimotility strategies.
Collapse
Affiliation(s)
- Miriam Kutsch
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Coral González-Prieto
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02115, USA.,Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Cammie F Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02115, USA.,Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
35
|
Robertin S, Mostowy S. The history of septin biology and bacterial infection. Cell Microbiol 2021; 22:e13173. [PMID: 32185906 DOI: 10.1111/cmi.13173] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/29/2022]
Abstract
Investigation of cytoskeleton during bacterial infection has significantly contributed to both cell and infection biology. Bacterial pathogens Listeria monocytogenes and Shigella flexneri are widely recognised as paradigms for investigation of the cytoskeleton during bacterial entry, actin-based motility, and cell-autonomous immunity. At the turn of the century, septins were a poorly understood component of the cytoskeleton mostly studied in the context of yeast cell division and human cancer. In 2002, a screen performed in the laboratory of Pascale Cossart identified septin family member MSF (MLL septin-like fusion, now called SEPT9) associated with L. monocytogenes entry into human epithelial cells. These findings inspired the investigation of septins during L. monocytogenes and S. flexneri infection at the Institut Pasteur, illuminating important roles for septins in host-microbe interactions. In this review, we revisit the history of septin biology and bacterial infection, and discuss how the comparative study of L. monocytogenes and S. flexneri has been instrumental to understand septin roles in cellular homeostasis and host defence.
Collapse
Affiliation(s)
- Stevens Robertin
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Serge Mostowy
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
36
|
Septins in Infections: Focus on Viruses. Pathogens 2021; 10:pathogens10030278. [PMID: 33801245 PMCID: PMC8001386 DOI: 10.3390/pathogens10030278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/17/2021] [Accepted: 02/25/2021] [Indexed: 11/17/2022] Open
Abstract
Human septins comprise a family of 13 genes that encode conserved GTP-binding proteins. They form nonpolar complexes, which assemble into higher-order structures, such as bundles, scaffolding structures, or rings. Septins are counted among the cytoskeletal elements. They interact with the actin and microtubule networks and can bind to membranes. Many cellular functions with septin participation have been described in the literature, including cytokinesis, motility, forming of scaffolding platforms or lateral diffusion barriers, vesicle transport, exocytosis, and recognition of micron-scale curvature. Septin dysfunction has been implicated in diverse human pathologies, including neurodegeneration and tumorigenesis. Moreover, septins are thought to affect the outcome of host–microbe interactions. Implication of septins has been demonstrated in fungal, bacterial, and viral infections. Knowledge on the precise function of a particular septin in the different steps of the virus infection and replication cycle is still limited. Published data for vaccinia virus (VACV), hepatitis C virus (HCV), influenza A virus (H1N1 and H5N1), human herpesvirus 8 (HHV-8), and Zika virus (ZIKV), all of major concern for public health, will be discussed here.
Collapse
|
37
|
Kesisova IA, Robinson BP, Spiliotis ET. A septin GTPase scaffold of dynein-dynactin motors triggers retrograde lysosome transport. J Cell Biol 2021; 220:211663. [PMID: 33416861 PMCID: PMC7802366 DOI: 10.1083/jcb.202005219] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/22/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
The metabolic and signaling functions of lysosomes depend on their intracellular positioning and trafficking, but the underlying mechanisms are little understood. Here, we have discovered a novel septin GTPase-based mechanism for retrograde lysosome transport. We found that septin 9 (SEPT9) associates with lysosomes, promoting the perinuclear localization of lysosomes in a Rab7-independent manner. SEPT9 targeting to mitochondria and peroxisomes is sufficient to recruit dynein and cause perinuclear clustering. We show that SEPT9 interacts with both dynein and dynactin through its GTPase domain and N-terminal extension, respectively. Strikingly, SEPT9 associates preferentially with the dynein intermediate chain (DIC) in its GDP-bound state, which favors dimerization and assembly into septin multimers. In response to oxidative cell stress induced by arsenite, SEPT9 localization to lysosomes is enhanced, promoting the perinuclear clustering of lysosomes. We posit that septins function as GDP-activated scaffolds for the cooperative assembly of dynein-dynactin, providing an alternative mechanism of retrograde lysosome transport at steady state and during cellular adaptation to stress.
Collapse
|
38
|
Györffy BA, Tóth V, Török G, Gulyássy P, Kovács RÁ, Vadászi H, Micsonai A, Tóth ME, Sántha M, Homolya L, Drahos L, Juhász G, Kékesi KA, Kardos J. Synaptic mitochondrial dysfunction and septin accumulation are linked to complement-mediated synapse loss in an Alzheimer's disease animal model. Cell Mol Life Sci 2020; 77:5243-5258. [PMID: 32034429 PMCID: PMC7671981 DOI: 10.1007/s00018-020-03468-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/25/2019] [Accepted: 01/21/2020] [Indexed: 12/21/2022]
Abstract
Synaptic functional disturbances with concomitant synapse loss represent central pathological hallmarks of Alzheimer's disease. Excessive accumulation of cytotoxic amyloid oligomers is widely recognized as a key event that underlies neurodegeneration. Certain complement components are crucial instruments of widespread synapse loss because they can tag synapses with functional impairments leading to their engulfment by microglia. However, an exact understanding of the affected synaptic functions that predispose to complement-mediated synapse elimination is lacking. Therefore, we conducted systematic proteomic examinations on synaptosomes prepared from an amyloidogenic mouse model of Alzheimer's disease (APP/PS1). Synaptic fractions were separated according to the presence of the C1q-tag using fluorescence-activated synaptosome sorting and subjected to proteomic comparisons. The results raised the decline of mitochondrial functions in the C1q-tagged synapses of APP/PS1 mice based on enrichment analyses, which was verified using flow cytometry. Additionally, proteomics results revealed extensive alterations in the level of septin protein family members, which are known to dynamically form highly organized pre- and postsynaptic supramolecular structures, thereby affecting synaptic transmission. High-resolution microscopy investigations demonstrated that synapses with considerable amounts of septin-3 and septin-5 show increased accumulation of C1q in APP/PS1 mice compared to the wild-type ones. Moreover, a strong positive correlation was apparent between synaptic septin-3 levels and C1q deposition as revealed via flow cytometry and confocal microscopy examinations. In sum, our results imply that deterioration of synaptic mitochondrial functions and alterations in the organization of synaptic septins are associated with complement-dependent synapse loss in Alzheimer's disease.
Collapse
Affiliation(s)
- Balázs A Györffy
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Vilmos Tóth
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - György Török
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Péter Gulyássy
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Réka Á Kovács
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Henrietta Vadászi
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - András Micsonai
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - László Homolya
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| | - László Drahos
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gábor Juhász
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- CRU Hungary Ltd., Göd, Hungary
| | - Katalin A Kékesi
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - József Kardos
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary.
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
39
|
Huang T, Pu Q, Zhou C, Lin P, Gao P, Zhang X, Chu Y, Yue B, Wu M. MicroRNA-302/367 Cluster Impacts Host Antimicrobial Defense via Regulation of Mitophagic Response Against Pseudomonas aeruginosa Infection. Front Immunol 2020; 11:569173. [PMID: 33117356 PMCID: PMC7576609 DOI: 10.3389/fimmu.2020.569173] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023] Open
Abstract
Mitophagy has recently been implicated in bacterial infection but the underlying mechanism remains largely unknown. Here, we uncover a role of microRNA-302/367 cluster in regulating mitophagy and its associated host response against bacterial infection. We demonstrate that miR-302/367 cluster expression was significantly increased after Pseudomonas aeruginosa infection. Enhanced expression of miR-302/367 cluster accelerated the mitophagic response in macrophages, thus increasing clearance of invading P. aeruginosa by regulating production of reactive oxygen species (ROS), while application of miR-302/367 cluster inhibitors decreased bacterial clearance. Blocking mitophagy with siRNA against mitophagy receptor prohibitin 2 (PHB2) reduced the effect of miR-302/367 cluster on induction of mitophagy and its-associated P. aeruginosa elimination. In addition, we found that miR-302/367 cluster also increased bacterial clearance in mouse model. Mechanistically, we illustrate that miR-302/367 cluster binds to the 3′-untranslated region of nuclear factor kappa B (NF-κB), a negative regulator of mitophagy, accelerated the process of mitophagy by inhibiting NF-κB. Furthermore, inhibition of NF-κB in macrophages attenuated the ROS or cytokines production and may reduce cell injury by P. aeruginosa infection to maintain cellular homeostasis. Collectively, our findings elucidate that miR-302/367 cluster functions as potent regulators in mitophagy-mediated P. aeruginosa elimination and pinpoint an unexpected functional link between miRNAs and mitophagy.
Collapse
Affiliation(s)
- Ting Huang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China.,Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China.,Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chuanmin Zhou
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Ping Lin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Pan Gao
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuyue Zhang
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Yiwen Chu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Bisong Yue
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| |
Collapse
|
40
|
Novel Functions of the Septin Cytoskeleton: Shaping Up Tissue Inflammation and Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:40-51. [PMID: 33039354 DOI: 10.1016/j.ajpath.2020.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/24/2020] [Accepted: 09/16/2020] [Indexed: 12/19/2022]
Abstract
Chronic inflammatory diseases cause profound alterations in tissue homeostasis, including unchecked activation of immune and nonimmune cells leading to disease complications such as aberrant tissue repair and fibrosis. Current anti-inflammatory therapies are often insufficient in preventing or reversing these complications. Remodeling of the intracellular cytoskeleton is critical for cell activation in inflamed and fibrotic tissues; however, the cytoskeleton has not been adequately explored as a therapeutic target in inflammation. Septins are GTP-binding proteins that self-assemble into higher order cytoskeletal structures. The septin cytoskeleton exhibits a number of critical cellular functions, including regulation of cell shape and polarity, cytokinesis, cell migration, vesicle trafficking, and receptor signaling. Surprisingly, little is known about the role of the septin cytoskeleton in inflammation. This article reviews emerging evidence implicating different septins in the regulation of host-pathogen interactions, immune cell functions, and tissue fibrosis. Targeting of the septin cytoskeleton as a potential future therapeutic intervention in human inflammatory and fibrotic diseases is also discussed.
Collapse
|
41
|
DUSP5 (dual-specificity protein phosphatase 5) suppresses BCG-induced autophagy via ERK 1/2 signaling pathway. Mol Immunol 2020; 126:101-109. [PMID: 32795663 DOI: 10.1016/j.molimm.2020.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/17/2022]
Abstract
Autophagy is considered as an effective strategy for host cells to eliminate intracellular Mycobacterium tuberculosis (Mtb). Dual-specificity phosphatase 5 (DUSP5) is an endogenous phosphatase of ERK1/2, and plays an important role in host innate immune responses, its function in autophagy regulation however remains unexplored. In the present study, the function of DUSP5 in autophagy in Mycobacterium bovis Bacillus Calmette-Guerin (BCG)-infected RAW264.7 cells, a murine macrophage-like cell line, was examined by assessing the alteration of the cell morphology, expression of autophagy markers, and ERK1/2 signaling activation. The results demonstrated that the BCG infection could induce DUSP5 expression and activate ERK1/2 signaling in RAW264.7 cells; an activation of ERK1/2 signaling contributed to autophagic process in RAW264.7 cells. Moreover, DUSP5 knockdown increased the expression of autophagy-related proteins (Atgs), including LC3-II, Beclin1, Atg5 and Atg7. However, an overexpression of DUSP5 exhibited an opposite effect. Mechanistically, DUSP5 could inhibit the formation of autophagosome by suppressing the phosphorylation of signaling molecules in ERK1/2 signaling cascade. This study thus demonstrated a novel role of DUSP5 in modulating autophagy inRAW264.7 cells in response to BCG infection in particular, and autophagy macrophage to Mtb in general.
Collapse
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
Soultawi C, Fortier Y, Soundaramourty C, Estaquier J, Laforge M. Mitochondrial Bioenergetics and Dynamics During Infection. EXPERIENTIA. SUPPLEMENTUM 2019; 109:221-233. [PMID: 30535601 DOI: 10.1007/978-3-319-74932-7_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Microbes have developed a series of strategies to overcome the defense mechanisms of the infected host. During pathogen-host coevolution, they develop strategy to manipulate cellular machinery particularly in subverting mitochondrion function. Mitochondria are highly dynamic organelles that constantly remodel their structure. In particular, shaping and cellular distribution of the mitochondrial network is maintained in large part by the conserved activities of mitochondrial division, fusion, motility, and tethering. Mitochondria have been long recognized for their role in providing energy production, calcium metabolism, and apoptosis. More recently, mitochondria have been also shown to serve as a platform for innate immune response. In this context, mitochondrial dynamics and shaping is not only essential to maintain cristae structure and bioenergetic to fuel cellular demands but contribute to regulate cellular function such as innate immune response and mitochondrial permeabilization. Due to their key role in cell survival, mitochondria represent attractive targets for pathogens. Therefore, microbes by manipulating mitochondrial dynamics may escape to host cellular control. Herein, we describe how mitochondrial bioenergetics, dynamics, and shaping are impacted during microbe infections and how this interplay benefits to pathogens contributing to the diseases.
Collapse
Affiliation(s)
- Cynthia Soultawi
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France
| | - Yasmina Fortier
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France
| | | | - Jérôme Estaquier
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France. .,Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medicine, Laval University, Québec, QC, Canada.
| | - Mireille Laforge
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France.
| |
Collapse
|
46
|
Krokowski S, Atwal S, Lobato-Márquez D, Chastanet A, Carballido-López R, Salje J, Mostowy S. Shigella MreB promotes polar IcsA positioning for actin tail formation. J Cell Sci 2019; 132:jcs.226217. [PMID: 30992346 PMCID: PMC6526709 DOI: 10.1242/jcs.226217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/01/2019] [Indexed: 01/17/2023] Open
Abstract
Pathogenic Shigella bacteria are a paradigm to address key issues of cell and infection biology. Polar localisation of the Shigella autotransporter protein IcsA is essential for actin tail formation, which is necessary for the bacterium to travel from cell-to-cell; yet how proteins are targeted to the bacterial cell pole is poorly understood. The bacterial actin homologue MreB has been extensively studied in broth culture using model organisms including Escherichia coli, Bacillus subtilis and Caulobacter crescentus, but has never been visualised in rod-shaped pathogenic bacteria during infection of host cells. Here, using single-cell analysis of intracellular Shigella, we discover that MreB accumulates at the cell pole of bacteria forming actin tails, where it colocalises with IcsA. Pharmacological inhibition of host cell actin polymerisation and genetic deletion of IcsA is used to show, respectively, that localisation of MreB to the cell poles precedes actin tail formation and polar localisation of IcsA. Finally, by exploiting the MreB inhibitors A22 and MP265, we demonstrate that MreB polymerisation can support actin tail formation. We conclude that Shigella MreB promotes polar IcsA positioning for actin tail formation, and suggest that understanding the bacterial cytoskeleton during host–pathogen interactions can inspire development of new therapeutic regimes for infection control. This article has an associated First Person interview with the first author of the paper. Summary: The pathogen Shigella forms actin tails to move through the cytosol of infected cells. We show that the bacterial actin homologue MreB can help to position the autotransporter protein IcsA for such actin tail formation.
Collapse
Affiliation(s)
- Sina Krokowski
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK.,Department of Immunology & Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Sharanjeet Atwal
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7JT, UK.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400 PHRI 07103, Thailand.,Public Health Research Institute, Rutgers Biomedical and Health Science, Newark, New Jersey NJ 07103, USA
| | - Damián Lobato-Márquez
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK.,Department of Immunology & Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Arnaud Chastanet
- MICALIS Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Rut Carballido-López
- MICALIS Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Jeanne Salje
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7JT, UK.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400 PHRI 07103, Thailand.,Public Health Research Institute, Rutgers Biomedical and Health Science, Newark, New Jersey NJ 07103, USA
| | - Serge Mostowy
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK .,Department of Immunology & Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
47
|
Abstract
Septins are widely recognized as a component of the cytoskeleton that is essential for cell division, and new work has shown that septins can recognise cell shape by assembling into filaments on membrane regions that display micrometer-scale curvature (e.g. at the cytokinetic furrow). Moreover, infection biology studies have illuminated important roles for septins in mediating the outcome of host-microbe interactions. In this Review, we discuss a selection of mechanistic insights recently gained from studying three infection paradigms: the rice blast fungus Magnaporthe oryzae, the poxvirus family member vaccinia virus and the Gram-negative bacterium Shigella flexneri These studies have respectively discovered that higher-order septin assemblies enable fungal invasion into plant cells, entrap viral particles at the plasma membrane and recognize dividing bacterial cells for delivery to lysosomes. Collectively, these insights illustrate how studying septin biology during microbial infection can provide fundamental advances in both cell and infection biology, and suggest new concepts underlying infection control.
Collapse
Affiliation(s)
- Hoan Van Ngo
- Department of Immunology & Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Serge Mostowy
- Department of Immunology & Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
48
|
Schnupf P, Sansonetti PJ. Shigella Pathogenesis: New Insights through Advanced Methodologies. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0023-2019. [PMID: 30953429 PMCID: PMC11588159 DOI: 10.1128/microbiolspec.bai-0023-2019] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Indexed: 02/07/2023] Open
Abstract
Shigella is a genus of Gram-negative enteropathogens that have long been, and continue to be, an important public health concern worldwide. Over the past several decades, Shigella spp. have also served as model pathogens in the study of bacterial pathogenesis, and Shigella flexneri has become one of the best-studied pathogens on a molecular, cellular, and tissue level. In the arms race between Shigella and the host immune system, Shigella has developed highly sophisticated mechanisms to subvert host cell processes in order to promote infection, escape immune detection, and prevent bacterial clearance. Here, we give an overview of Shigella pathogenesis while highlighting innovative techniques and methods whose application has significantly advanced our understanding of Shigella pathogenesis in recent years.
Collapse
Affiliation(s)
- Pamela Schnupf
- Institut Imagine, Laboratory of Intestinal Immunity, INSERM UMR1163; Institut Necker Enfants Malades, Laboratory of Host-Microbiota Interaction, INSERM U1151; and Université Paris Descartes-Sorbonne, 75006 Paris, France
| | - Philippe J Sansonetti
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, and College de France, Paris, France
| |
Collapse
|
49
|
Spier A, Stavru F, Cossart P. Interaction between Intracellular Bacterial Pathogens and Host Cell Mitochondria. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0016-2019. [PMID: 30848238 PMCID: PMC11590420 DOI: 10.1128/microbiolspec.bai-0016-2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Indexed: 12/31/2022] Open
Abstract
Mitochondria are essential and highly dynamic organelles whose morphology is determined by a steady-state balance between fusion and fission. Mitochondrial morphology and function are tightly connected. Because they are involved in many important cellular processes, including energy production, cell-autonomous immunity, and apoptosis, mitochondria present an attractive target for pathogens. Here, we explore the relationship between host cell mitochondria and intracellular bacteria, with a focus on mitochondrial morphology and function, as well as apoptosis. Modulation of apoptosis can allow bacteria to establish their replicative niche or support bacterial dissemination. Furthermore, bacteria can manipulate mitochondrial morphology and function through secreted effector proteins and can also contribute to the establishment of a successful infection, e.g., by favoring access to nutrients and/or evasion of the immune system.
Collapse
Affiliation(s)
- Anna Spier
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
- Bio Sorbonne Paris Cité, Université Paris Diderot, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
| | - Fabrizia Stavru
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
- Centre National de la Recherche Scientifique, SNC 5101, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
| |
Collapse
|
50
|
Biskou O, Casanova V, Hooper KM, Kemp S, Wright GP, Satsangi J, Barlow PG, Stevens C. The type III intermediate filament vimentin regulates organelle distribution and modulates autophagy. PLoS One 2019; 14:e0209665. [PMID: 30699149 PMCID: PMC6353089 DOI: 10.1371/journal.pone.0209665] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
The cytoskeletal protein vimentin plays a key role in positioning of organelles within the cytosol and has been linked to the regulation of numerous cellular processes including autophagy, however, how vimentin regulates autophagy remains relatively unexplored. Here we report that inhibition of vimentin using the steroidal lactone Withaferin A (WFA) causes vimentin to aggregate, and this is associated with the relocalisation of organelles including autophagosomes and lysosomes from the cytosol to a juxtanuclear location. Vimentin inhibition causes autophagosomes to accumulate, and we demonstrate this results from modulation of mechanistic target of rapamycin (mTORC1) activity, and disruption of autophagosome-lysosome fusion. We suggest that vimentin plays a physiological role in autophagosome and lysosome positioning, thus identifying vimentin as a key factor in the regulation of mTORC1 and autophagy.
Collapse
Affiliation(s)
- Olga Biskou
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Victor Casanova
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Kirsty M. Hooper
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Sadie Kemp
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Graham P. Wright
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Jack Satsangi
- Centre for Genomic & Experimental Medicine, University of Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh, United Kingdom
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Peter G. Barlow
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|